ArticlePDF AvailableLiterature Review

New approaches regarding the in vitro maturation of oocytes: Manipulating cyclic nucleotides and their partners in crime

Authors:

Abstract and Figures

Several discoveries have been described recently (5-10 years) about the biology of ovarian follicles (oocyte, cumulus cells and granulosa cells), including new aspects of cellular communication, the control of oocyte maturation and the acquisition of oocyte competence for fertilization and further embryo development. These advances are nourishing assisted reproduction techniques (ART) with new possibilities, in which novel culture systems are being developed and tested to improve embryo yield and quality. This mini-review aims to describe how the recent knowledge on the physiological aspects of mammalian oocyte is reflecting as original or revisited approaches into the context of embryo production. These new insights include recent findings on the mechanisms that control oocyte maturation, especially modulating intraoocyte levels of cyclic nucleotides during in vitro maturation using endogenous or exogenous agents. In this mini-review we also discuss the positive and negative effects of these manipulations on the outcoming embryo
Content may be subject to copyright.
35
Received July 13, 2016
Accepted November 01, 2016
Review Article
New approaches regarding the in vitro maturation of oocytes:
manipulating cyclic nucleotides and their partners in crime
Ramon Cesar Botigelli 1, Eduardo Montanari Razza1, Elisa Mariano Pioltine1, Marcelo Fábio Gouveia Nogueira1,2
1Department of Pharmacology, Institute of Bioscience, University of São Paulo State, Botucatu, São Paulo,
Brazil
2Department of Biological Sciences, Faculty of Sciences and Letters, University of São Paulo State, Assis, São
Paulo, Brazil
ABSTRACT
Several discoveries have been described recently (5-
10 years) about the biology of ovarian follicles (oocyte,
cumulus cells and granulosa cells), including new aspects
of cellular communication, the control of oocyte maturation
and the acquisition of oocyte competence for fertilization
and further embryo development. These advances are
nourishing assisted reproduction techniques (ART) with
new possibilities, in which novel culture systems are
being developed and tested to improve embryo yield and
quality. This mini-review aims to describe how the recent
knowledge on the physiological aspects of mammalian
oocyte is reecting as original or revisited approaches
into the context of embryo production. These new insights
include recent ndings on the mechanisms that control
oocyte maturation, especially modulating intraoocyte
levels of cyclic nucleotides during in vitro maturation using
endogenous or exogenous agents. In this mini-review we
also discuss the positive and negative eects of these
manipulations on the outcoming embryo
Keywords: Oocytes, cumulus cells, oocyte in vitro
maturation, cyclic nucleotides, in vitro oocyte maturation
techniques
JBRA Assisted Reproduction 2017;21(1):35-44
doi: 10.5935/1518-0557.20170010
BACKGROUND
The literature concerning oocyte competence and
embryo quality has become abundant in the last few
years. Several factors are involved in oocyte metabolism,
cyto-skeletal remodeling, accumulation of molecules
(RNAs), meiosis arrest/resumption and fertilization, all
of which are key events for initiating and sustaining early
embryogenesis. This large amount of published data has
allowed researchers to pursue new strategies in assisted
reproduction techniques (ART). This mini-review focuses
on the recent progress in understanding the events
controlling the acquisition of competence in oocytes and
the new mechanisms involved in the maintenance of oocyte
meiotic arrest; and thus, it may yield future approaches
regarding the development of novel systems of in vitro
culture, and hopefully bring the status of in vitro production
of embryos (IVP) to a whole new level. For the purpose
of this mini-review the literature search was performed in
the PubMed database, to nd all relevant papers focusing
in “mammalian oocyte maturation”, “in vitro oocyte
maturation techniques” and crossing data with “cyclic
nucleotides” (“cyclic adenosine monophosphate”, “cyclic
guanosine monophosphate”), “meiosis arrest/resumption”
and “embryo yield”, from which we have selected 112
papers, among original and review papers, to discuss the
most interesting ndings and also included some of our
own.
INTRODUCTION
Mammalian oocytes pass through a long and complex
process to acquire the competence necessary for
fertilization and embryogenesis. Oocytes are formed in
fetal life, when the primordial germ cells (PGC), rst seen
in the epiblast, move outside the embryo to the yolk sac,
and then migrate from the yolk sac to the early gonad
(genital ridges). After genital ridges are colonized by PGC
they are denominated primordial gonads (review by van
den Hurk & Zhao, 2005).
The migration and colonization of the gonads by PGC
in females give rise to the oogonia, which, associated with
somatic cells, undergo a phase of mitotic proliferation
with an incomplete cytokinesis. In the developing ovary,
the oogonia and pregranulosa somatic cells progressively
organize into epithelial structures eventually recognized as
ovarian follicles. However, before follicle formation, germ
cells change from mitotic to meiotic and become primary
oocytes, committed to follicle development (reviewed by
Guigon & Magre, 2006).
Still in fetal phase, all female germ cells reach the
prophase of the rst meiotic division, but instead of
progressing to metaphase, they are kept arrested in the
diplotene stage or germinal vesicle (GV). After birth,
oocytes undergo some important processes for their
growth and maturation, such as storage of mRNA, proteins,
metabolic substrates and organelle reorganization. The
primary oocytes, arrested in the rst meiotic division, now
become enveloped by a layer of attened pregranulosa
cells and a basal membrane to become primordial follicles
(Picton, 2001).
Followed by the activation of growth, the primordial
follicle is surrounded by a complete layer of cuboidal
granulosa cells making it a primary follicle (Picton,
2001). During follicular growth, granulosa cells continue
to proliferate and the theca layer is developed, which is
required to produce androgens and to form the network
of cells that support the vascular system of the growing
follicle (Young & McNeilly, 2010).
Primordial follicles remain ‘dormant’ in the ovaries until
recruitment into the population of growing cells. Every
day, a group of primordial follicles are recruited and start
to grow based on the order in which they are initially
formed. Consequently, certain primordial follicles are rst
transformed into primary follicles after a few days and
others only after more than a year as in rodents, or after
one to ve decades later, as in women (van den Hurk &
Zhao, 2005).
Follicles are called primary follicles when the single
layer of granulosa cells surrounding the oocyte becomes
cuboidal. The transition of primordial follicles into primary
follicles is slow and the diameter of its oocyte hardly
changes. This process is associated with commitment
36
Review Article
JBRA Assist. Reprod. | v.21 | no1| Jan-Feb-Mar/ 2017
and subsequent stages of follicular development, and it
is independent of direct FSH action (Méduri et al., 2002).
Furthermore, when the follicle reaches several layers
of granulosa cells, it starts forming the antrum, and the
granulosa cells dierentiate into two compartments: the
mural cells, which internally surround the basal membrane,
and the cumulus oophorus cells (CCs), that are closely
associated with the oocyte. This structure forms the so-
called cumulus-oocyte complex (COC) and its intricate
interaction confers the oocyte with the competence to
resume meiosis and be fertilized (Hyttel et al., 2010;
Picton, 2001).
Bidirectional communication between oocytes
and somatic cells
The granulosa cells have clearly established roles to
support oocyte growth and the acquisition of developmental
competence (Brower & Schultz, 1982), but also participate
in the control of meiosis progression (Eppig, 1991), and
in the modulation of global transcriptional activity and
chromatin remodeling in the oocyte (De La Fuente & Eppig,
2001).
In recent years, researchers have focused on the
understanding how the oocyte can inuence granulosa
cells through the so-called oocyte derived paracrine factors
(ODPFs). Among them, the main players are the proteins
of the transforming growth factor β (TGF-β) superfamily,
such as the growth dierentiation factor 9 (GDF9) and
the bone morphogenetic protein 15 (BMP15). Fibroblast
growth factors (FGFs) are also secreted by oocytes and
are reported to regulate granulosa cell development and
function cooperatively with TGF-β proteins (reviewed by
Emori & Sugiura, 2014).
Especially within antral follicles, ODPFs guide the
dierentiation and maintenance of granulosa and CCs
(Eppig et al., 1997). In addition, ODPFs can stimulate
growth and apoptosis (Gilchrist et al., 2001), energy
metabolism (Sugiura et al., 2005; Sutton et al., 2003;
Sutton-McDowall et al., 2010), sterol biosynthesis (Su
et al., 2008) and the CCs expansion (Elvin et al., 1999;
Varani et al., 2002). Thus, the oocyte can aect the
functions of the CCs for their own benet, since the oocyte
is not able to produce all the substrates required for its
maturation. To ensure an eective development, oocyte
and cumulus/granulosa cells must communicate through a
perfectly orchestrated signaling system.
One mechanisms of bidirectional communication
between the CCs and oocyte is through the gap junctional
communication (GJC). Gap junctions (GJ) are specialized
membrane proteins occurring in points of very close
contact between both cells. They consist of arrays of
intercellular channels that allow direct sharing of small
(less than 1 kD) molecules between the cells (Harris,
2001). Indeed, many of the molecules are known to be
transferred from granulosa cells to the growing oocyte via
GJC, e.g., amino acids, glucose, and ribonucleotides (Eppig
et al., 2005; Sugiura et al., 2005). GJ are comprised of
connexins, a homologous family of more than 20 proteins.
The connexin 43 (Cx43) is predominantly expressed by
cumulus/granulosa cells whereas Cx37 seems to be the
only connexin connecting oocyte to the granulosa cells
(Juneja et al., 1999), and the loss of Cx37 expression
is detrimental to the oocyte-granulosa communication
(Simon et al., 1997).
Macaulay et al. (2014, 2016) demonstrated by confocal
and transmission electron microscopy, in combination with
transcript detection, that somatic cells contribute to the
maternal reserves of oocytes, including mRNA and long
noncoding RNA. This communication is performed by
transzonal projections (TZPs). These recent discoveries
rened our understanding of the small molecule transport
mechanism (GJC/TZP) synthesized by cumulus cells, which
are transferred into the ooplasm.
Recently, a new mechanism of cell communication within
the ovarian follicle was demonstrated; this mechanism is
performed by extracellular vesicles (EVs). Initially, EVs
were described in ovarian follicular uid of mares using
ow cytometer and transmission electron microscopy
techniques (da Silveira et al., 2012). These EVs are lipid
bilayer structures secreted by many cell types into the
extracellular uid, serving as a vehicle for membrane and
cytosolic proteins, lipids, and RNA (Raposo & Stoorvogel,
2013). Several articles identied miRNAs in bovine (Miles
et al., 2012), equine (da Silveira et al., 2012) and human
(Santonocito et al., 2014) follicular uid, suggesting EVs
as a potential mediator of cell-to-cell communication,
impacting oocyte and follicle growth (reviewed by da
Silveira et al., 2015).
Cyclic nucleotides and maturation control
Other important molecules that also use the GJC/TZP
system to move around between CCs and oocyte are the
cyclic nucleotides. Among those, we should highlight the
adenosine 3’,5’-cyclic monophosphate (cAMP). This second
messenger acts mostly in the phosphorylation of the cAMP-
dependent protein kinase A (PKA), leading to the activation
of various cellular pathways. The cAMP is synthesized from
adenosine triphosphate (ATP) by adenylate cyclase (AC),
following the dissociation of the stimulatory-G (Gs) protein
from specic classes of G-protein-coupled receptors (Wright
et al., 2015). Variation in the intraoocyte concentration of
cAMP can modulate the resumption of meiosis. Optimum
concentration of cAMP maintains PKA active, which inhibits
the maturation-promoting factor (MPF) and keeps the
oocyte arrested at the GV stage (Sirard et al., 1998).
Another cyclic nucleotide, the cyclic guanosine
monophosphate (cGMP), also plays its role in controlling
meiotic arrest/resumption. cGMP is synthesized via
dierent pathways, such as through nitric oxide (NO),
bicarbonate, natriuretic peptides (NPPA, NPPB and NPPC),
guanylins, uroguanylins and guanylyl cyclase activating
proteins (GCAPs); those guanylin molecules can activate
various enzymes, e.g., guanyl, adenylyl cyclases and
guanylate, which act in the catalytic conversion of guanosine
triphosphate (GTP) into cGMP and pyrophosphate (Potter,
2011).
Like cAMP, the cGMP molecules participate in protein
kinase phosphorylation (cGMP-dependent protein kinase,
PKG) and inuence the activity of several phosphodiesterases
(PDEs). The PDEs are intracellular enzymes that catalyze
the hydrolysis of the cyclic phosphate bond into cAMP and
cGMP to generate the inactive products 5’-AMP and 5’-
GMP (Francis et al., 2011). The PDEs are classied into 11
families according to their anity, although each family can
have multiple isoforms (Francis et al., 2011). PDE activities
can be of short or long term, and are modulated by signals
including hormones, neurotransmitters, cytokines, light,
and oxidative inuences. The concentration of nucleotides
(cAMP and cGMP) is controlled by the balance between
their synthesis and degradation, which is carried out by
the PDEs themselves (reviewed by Francis et al., 2011).
PDEs decrease cAMP concentration in immature
oocytes to allow for meiosis resumption and, consequently,
the onset of oocyte maturation (Sadler & Maller, 1989).
Tsafriri et al. (1996) reported that the location of PDE3A
is restricted to the oocyte and they showed an eectively
inhibition of spontaneous meiosis resumption in vitro
using specic inhibitors. Additionally, Norris et al. (2009)
demonstrated that cGMP synthesized by CCs moves across
GJ/TZP to the oocyte and inhibit cAMP degradation by
PDE3A. This process assures that the cAMP concentration,
37
Cyclic nucleotides oocytes maturation - Botigelli, R C
JBRA Assist. Reprod. | v.21 | no1| Jan-Feb-Mar/ 2017
demanded by the GV-arrest, be maintained at optimum
levels.
During the normal reproductive cycle, a surge of LH
induces oocyte maturation and ovulation (Richards et al.,
2002). Triggered by LH, a receptor coupled to G protein
is activated in the theca and granulosa cells (Breen et al.,
2013; Gudermann et al., 1992; Rajagopalan-Gupta et al.,
1998), inducing a rapid reduction in follicle cGMP, which
is diused out of the oocyte through GJ/TZP (Shuhaibar
et al., 2015). Simultaneously, LH-induced phosphorylation
and activation of PDE5 leads to decreasing levels of cGMP
and relieves the inhibition of PDE3A in the oocyte, lowering
cAMP content and allowing meiosis to resume (Egbert
et al., 2016).
Knowledge of the physiology involved in oocyte
meiotic arrest/resumption and maturation has enabled
the development and improvement of techniques for
the in vitro maturation (IVM) of oocytes. The IVM of
mammalian oocytes is an essential tool for the basic or
applied aspects of assisted reproductive technology (ART)
such as developmental biology, in vitro production (IVP)
of embryos, cloning, stem cells and embryology (Smitz
et al., 2011). However, the eciency of IVM is still low
when compared to in vivo maturation, which limits its
application in ART (Gilchrist, 2011). Drawbacks of IVM
include decreased preimplantation embryo development,
low pregnancy rates and poor live birth index (Child et al.,
2002; Eppig et al., 2009). This is probably caused by the
ineciency of the oocyte to avoid the drastic decrease
in cAMP concentration when removed from the follicular
environment during ART procedures (Luciano et al., 2004;
Mattioli et al., 1994). This spontaneous resumption of
oocyte meiosis causes incomplete cytoplasmic maturation,
and the asynchrony between cytoplasmic and nuclear
maturation, aects oocyte development and embryo
quality (Blondin et al., 1997; Gilchrist & Thompson, 2007;
Lonergan et al., 2003).
Several authors have reported reversible inhibition
of spontaneous meiotic resumption by pharmacological
methods and most of these strategies are described in the
following section of this review.
Pharmacological approaches to modulate cyclic
nucleotides during in vitro maturation
Reversible inhibition of meiotic resumption by
pharmacological methods have been long tried by many
researchers, but results on the subsequent developmental
competence is variable and often lower than in COCs
cultured without inhibition (Fulka et al., 1991; Lonergan
et al., 1997; Avery et al., 1998; Kubelka et al., 2000;
Mermillod et al., 2000). In addition, pharmacological
manipulations may, occasionally, aect oocytes and
embryos at the ultrastructural level as well (Faerge et al.,
2001; Lonergan et al., 2003; Nogueira et al., 2003, 2005;
Vanhoutte et al., 2007).
In mammalian oocytes, among the pharmacological
approaches used for in vitro maturation to maintain
meiotic arrest - or at least to retard meiotic spontaneous
resumption - we have the cAMP modulators: dbcAMP
(Sirard & First, 1988) and 8-bromo-cAMP (Chen et al.,
2009), phosphodiesterase inhibitors: specic inhibitors
of the PDE3, such as, cilostamide (Gharibi et al., 2013;
Mayes & Sirard, 2002; Shu et al., 2008; Vanhoutte et al.,
2008) and milrinone (Mayes & Sirard, 2002; Thomas et al.,
2002, 2004b), PDE4: rolipram (Mayes & Sirard, 2002;
Thomas et al., 2002, 2004b) and PDE8: dipyridamole
(Sasseville et al., 2009), nonspecic inhibitor: IBMX (Albuz
et al., 2010; Rose et al., 2013; Thomas et al., 2002) and
stimulators of adenylate cyclase: forskolin (Albuz et al.,
2010; Richani et al., 2014; Shu et al., 2008; Zeng et al.,
2014) and iAC (Aktas et al., 1995; Guixue et al., 2001;
Luciano et al., 2004).
The overall objective of these pharmacological
manipulations is to avoid premature nuclear maturation
in vitro by means of maintaining higher concentration of
cAMP within the ooplasm. This should provide enough
time for the COC to synchronize nuclear and cytoplasmic
maturation, as similar as possible to the in vivo event
that would allegedly result in more competent oocytes
and embryo (Thomas et al., 2004a). Unsurprisingly, the
mechanism of synthesis and hydrolysis of cGMP is one of
the main targets of pharmacological strategies to control
oocyte maturation.
Nakamura et al. (2002) described an important role of
inducible nitric oxide (NO) synthase (iNOS)/NO/cGMP in the
control of oocyte maturation in rats. This technique uses
a NO donor (S-nitroso-L-acetyl penicillamine - SNAP) for 5
hours to reversibly prevent GV breakdown. This discovery
paved the way for many other studies that also reported
the activation of this pathway in several mammalian
species, including rats (Sela-Abramovich et al., 2008),
mice (Norris et al., 2009), pigs (Chmelíková et al., 2010;
Tichovská et al., 2011) and bovines (Pires et al., 2009;
Sasseville et al., 2008; Schwarz et al., 2008, 2010).
New approaches for modifying IVM and improve
developmental competence take into consideration the
knowledge from cGMP/cAMP and the use of dynamic
systems. Furthermore, non-pharmacological strategies
are trending since the 2010 paper from Dr. John Eppig,
describing the role of the granulosa cell ligand natriuretic
peptide precursor type C (NPPC) and its receptor NPR2 in
maintaining meiotic arrest in mice oocytes.
Novel systems of in vitro maturation and their
impacts in the resulting embryo
Based on the signicant advances of the mechanisms
that control oocyte maturation and their interaction with
the CCs, new paths were opened to improve the IVM
technique. One of which is the use of dynamic in vitro
systems to improve embryo quality and quantity, the so-
called prematuration or pre-IVM systems.
Interesting approaches for modifying IVM to improve
developmental competence is the use of a two-step culture
or pre-maturation systems, where during the initial step a
medium that does not promote nuclear maturation is used
(Albuz et al., 2010; Franciosi et al., 2014; Luciano et al.,
2004; Oliveira e Silva et al., 2011; Ponderato et al., 2002).
Among the systems that have been developed, the most
promising are those that pharmacologically inhibit or retard
meiotic resumption by elevating cAMP concentration in the
oocyte while sustaining GJ communication functionality
(Albuz et al., 2010; Luciano et al., 2004). It was previously
reported that modulation of cAMP levels within mammalian
COCs during IVM could substantially improve oocyte
developmental competence in several species (Albuz et al.,
2010; Funahashi et al., 1997; Luciano et al., 1999, 2004;
Nogueira et al., 2003, 2006; Shu et al., 2008; Thomas
et al., 2004b; Vanhoutte et al., 2009; Zeng et al., 2013;
Figure 1).
In 2003, Shimada et al. (2003) investigated the
formation of LH receptor in cumulus cells of swine COCs
and, after its detection, a new two-step culture system
was developed. In the rst step, the COCs were cultured
in medium supplemented with FSH and IBMX for 20h,
followed by culture in medium supplemented with LH
(second-step). This two-step system with FSH and 0.5
mM IBMX induced the expression of LH receptors in CCs,
improved the rate of blastocyst formation and increased
the number of cells in IVF blastocysts (Figure 1).
38
Review Article
JBRA Assist. Reprod. | v.21 | no1| Jan-Feb-Mar/ 2017
Figure 1. Summarized representation of dierent strategies used during in vitro maturation to modulate the intraoocyte
concentration of cyclic nucleotides and improve embryo yield in several mammalian species (pigs, cattle, mice, sheep or
goat). After each approach, there is a brief schematic description of the methods the authors used. The gure shows the
rates of blastocyst in each treatment in comparison to their respective control group. Statistical signicance is indicated by
the p value. Note that some authors calculate the in vitro performance by dividing the number of blastocyst by the number
of oocytes and others by the number of cleaved embryos. IVF: in vitro fertilization; IVC: in vitro culture; PA: parthenogenetic
activation.
To better understand the inuence of hormones and
growth factor production on the mechanisms controlling
the in vivo maturation in pigs, Kawashima et al. (2008)
updated the two-step system into a new one called “novel
culture system - NCS”. In the NCS design, COCs are
recovered from small antral follicles (3-5mm in diameter);
rst pre-IVM uses FSH, E2 and IBMX for 10h to induce
cell proliferation; second pre-IVM takes place with FSH,
E2, IBMX and P4 for 10h to suppress cell proliferation and
induce LH receptor mRNA expression and nally, an IVM
with LH, EGF and P4 for additional 24h (Figure 1). Using
NCS system, Kawashima et al. (2008) reported the full
expansion of porcine COCs, decreased number of cumulus
cells in apoptotic process and, when oocytes obtained from
NCS were used for IVF, the developmental competence
to blastocyst was signicantly improved when compared
with the conventional culture system (FSH+LH for 48h), or
with the two-step culture system (Funahashi et al., 1997;
Shimada et al., 2003; Figure 1).
Early in the decade, Albuz et al. (2010) proposed a new
IVM system. Their methodology was seeking to mimic the
processes that occurred in the in vivo maturation; hence
their system was called simulated physiological oocyte
maturation (SPOM; Figure 1). This system consisted of
a small pre-IVM (1-2h) where the adenylate cyclase was
stimulated with forskolin, increasing cAMP levels and IBMX,
a PDE inhibitor, to prevent hydrolysis of cyclic nucleotides
(cAMP and cGMP), and after the pre-IVM, the COCs were
subjected to an extended IVM for 24 hours, where the
culture medium was supplemented with cilostamide (PDE3
inhibitor) and recombinant human (rh)-FSH. Results of the
SPOM system were quite exciting, with rates of 69% of
blastocysts per cleaved embryo. Later, the SPOM protocol
was adapted to sheep oocytes and, even though no
signicant eect on blastocyst rates were achieved, there
was an improvement in blastocyst quality observed by an
increase in total cell number (Rose et al., 2013; Figure 1).
The promising results of the rst version of the SPOM
system (SPOMv1) greatly impacted ART research; still, the
SPOM system was updated by their creators in subsequent
studies. Zeng et al. (2013) used heparin during pre-MIV
and removed the cilostamide from the extended IVM. This
approach positively aected oocyte energy metabolism,
oocyte meiotic maturation and embryo development
(SPOMv2; Figure 1).
One year later, Zeng et al. (2014) tested the presence
of rh-FSH during extended IVM (Figure 1). By now it
seems that cilostamide in extended IVM phase is gone
39
Cyclic nucleotides oocytes maturation - Botigelli, R C
JBRA Assist. Reprod. | v.21 | no1| Jan-Feb-Mar/ 2017
for good, and now the system is no longer called SPOM,
but Prematuration System (or Pre-IVM system) instead.
They reported successful results in IVP of mice embryos
(± 70% of blastocysts per cleaved embryos), better
quality in the expansion of CCs, reduction of abnormal
spindles and a positive inuence of Pre-IVM system in the
glycolic metabolism of COCs (suggesting an eect of cAMP
production predominantly on glycolytic activity).
Several laboratories and research groups around the
world sought to repeat the success obtained by the SPOM or
Pre-IVM system; however, most failed in doing so (Bernal-
Ulloa et al., 2016; Buell et al., 2015; Guimarães et al.,
2015; Razza et al., 2015; Figure 1). Ulloa et al. (2015),
using bovine oocytes cultured in SPOM system, produced a
smaller number of blastocyst compared with the standard
IVM. However, the pattern of DNA methylation of embryos
produced in SPOM system was more similar to embryos
produced in vivo. Also, Santiquet et al. (2014a) tested a
pre-IVM treatment and could improve the developmental
competence of oocytes, as demonstrated by increased
embryo development. Additionally, pre-IVM performed
with IBMX and forskolin in the Pre-IVM system can change
ultrastructural characteristics of oocytes and blastocysts
(Razza et al., 2015; unpublished data from our group).
With the strategy used in the SPOM system of performing
a two-step culture with dierent drugs to induce dierent
eects in CCs and oocytes during IVM, new drugs and
signaling pathways have emerged as potential targets for
research seeking to improve IVM and embryo production.
Earlier in this decade, a new model discovered that the
binding of NPPC to its receptor (NPR2) in granulosa and CCs
are the main cause for the modulation of cGMP levels (Zhang
et al., 2010). Until now, studies relating the new mechanism
of NPPC/NPR2/cGMP in maturation control have been reported
in mice (Tsuji et al., 2012; Zhang et al., 2011), bovines
(Franciosi et al., 2014), swine (Santiquet et al., 2014b; Zhang
et al., 2014) and sheep (Zhang et al., 2015; Figure 1).
Using the NPPC during pre-IVM (8h) in bovine COCs,
Franciosi et al. (2014) could successfully arrest meiosis re-
sumption and extend the functional communication among
oocytes and CCs through GJ. After IVF and embryo culture,
the NPPC treatment in pre-IVM has also increased blasto-
cyst cell number and hatching rates.
In a more recent approach of two-step maturation with
caprine COCs, Zhang et al. (2015) used the NPPC and
estradiol during pre-IVM (8h), followed by conventional IVM
(18h). With this system, meiosis was eectively arrested in
pre-IVM and the maturation rate was also increased after
conventional IVM. They also increased embryo production
and quality, evaluated by total cell number per blastocyst
(Figure 1).
Conventionally, oocyte competence has been assessed
by embryo morphology and blastocyst rates; however,
these aspects alone do not provide sucient information to
fully endorse the IVM system eciency. Several strategies
are being used to study the quality of oocytes (before and
after IVM) and embryos. Some of the most promising and
approaches that focus on the identication of biomarkers.
New perspectives and nal considerations
A few non-invasive strategies are already being used
to predict oocyte competence to become a viable embryo,
even before the oocyte is fertilized. These approaches aim
to identify oocyte competence biomarkers mostly in cumulus
cells. In this context, the morphology of CCs can be used to
rst categorize oocyte potential (de Loos et al., 1991) and then
to compare the morphological data with CCs transcriptome
(dierentially expressed genes in CCs surrounding the good
oocytes versus poor-quality oocytes). At present, many genes
are identied as potential biomarkers (reviewed by Labrecque
& Sirard, 2014). Still, many research groups are working on
the identication of miRNAs as biomarkers as well. Proles
of miRNAs isolated from EVs present in follicular uid were
described and associated with proper cytoplasmic oocyte
maturation; hence, these miRNA proles can be used to
predict oocyte competence (Sohel et al., 2013).
The use of non-invasive strategies, such as analysis
of follicular uid and culture media (after culture) also
appears to be quite useful on the search for molecular
biomarkers for oocyte competence. The presence of
cytokines and growth factors in follicular uid is crucial for
determining oocyte quality (reviewed by Dumesic et al.,
2015). In this context, the metabolic characterization of
the culture media, in which IVP embryos are kept for many
hours, may represent an important non-invasive tool to
either indicate possible predictive biomarkers of viability
or to explain IVP outcome afterwards (Muñoz et al., 2014).
Lipid metabolism is induced in COCs during oocyte
maturation and contributes to oocyte and embryo
development (Gardner & Harvey, 2015). Specic fatty acids
have distinct eects on oocyte maturation. In general,
saturated fatty acids (palmitic acid and stearic acid) are
elevated in follicular uid and, in some metabolic contexts,
are detrimental, while the presence of unsaturated
non-esteried fatty acids (oleic acid and linoleic acid)
can counteract these detrimental eects and promote
developmental competence (reviwed by Dunning et al.,
2014). Thus, in vitro oocyte and embryo development may
be optimized through the provision of appropriate energy
substrates and essential co-factors during ART in domestic
animals and subfertility women.
Despite the large number of publications in the eld
we still have a long way to go to deeply understand and
manipulate the mechanisms controlling oocyte maturation.
Overcoming these gaps may allow us to improve ART
results. Therefore, it is necessary to design studies aiming
at nding eective biomarkers for oocyte competence. The
eld of the OMICs seems to be quite promising, especially
regarding the new ndings in transcriptomics, proteomics
and lipidomics in oocytes, CCs, embryos and in the EVs
within the follicular uid. Future studies on this subject
might enable the design of more complex, dened and
ecient culture conditions for oocytes to be fully matured
and able to generate optimum IVP embryos.
ACKNOWLEDGEMENTS
We acknowledge the São Paulo Research Foundation
(FAPESP) for funding (2012/50533-2 and 2013/05083-1) and
fellowships for RCB (2014/25072-7), EMR (12/23409-9) and
EMP (13/07730-4).
CONFLICT OF INTERESTS
No conict of interests has been declared.
Corresponding author:
Ramon Cesar Botigelli
Multi-user laboratory Phytomedicine
Pharmacology and Biotechnology (FitoFarmaTec)
Institute of Bioscience, University of São Paulo State
Botucatu - SP, Brazil
E-mail: ramonbotigelli@gmail.com
REFERENCES
Aktas H, Wheeler MB, Rosenkrans CF Jr, First NL, Leibfried-Rut-
ledge ML. Maintenance of bovine oocytes in prophase of meio-
sis I by high [cAMP]i. J Reprod Fertil. 1995;105:227-35. PMID:
8568765 DOI: http://dx.doi.org/10.1530/jrf.0.1050227
40
Review Article
JBRA Assist. Reprod. | v.21 | no1| Jan-Feb-Mar/ 2017
Albuz FK, Sasseville M, Lane M, Armstrong DT, Thompson
JG, Gilchrist RB. Simulated physiological oocyte matura-
tion (SPOM): a novel in vitro maturation system that sub-
stantially improves embryo yield and pregnancy outcomes.
Hum Reprod. 2010;25:2999-3011. PMID: 20870682
DOI: http://dx.doi.org/10.1093/humrep/deq246
Avery B, Hay-Schmidt A, Hyttel P, Greve T. Embryo develop-
ment, oocyte morphology, and kinetics of meiotic maturation
in bovine oocytes exposed to 6-dimethylaminopurine prior
to in vitro maturation. Mol Reprod Dev. 1998;50:334-44.
PMID: 9621310 DOI: http://dx.doi.org/10.1002/(SICI)1098-
2795(199807)50:3<334::AID-MRD10>3.0.CO;2-4
Bernal-Ulloa SM, Heinzmann J, Herrmann D, Hadeler KG,
Aldag P, Winkler S, Pache D, Baulain U, Lucas-Hahn A,
Niemann H. Cyclic AMP Aects Oocyte Maturation and
Embryo Development in Prepubertal and Adult Cattle.
PLoS One. 2016;11:e0150264. PMID: 26926596 DOI:
http://dx.doi.org/10.1371/journal.pone.0150264
Blondin P, Coenen K, Guilbault LA, Sirard MA. In vi-
tro production of bovine embryos: developmen-
tal competence is acquired before maturation. Ther-
iogenology. 1997;47:1061-75. PMID: 16728056 DOI:
http://dx.doi.org/10.1016/S0093-691X(97)00063-0
Breen SM, Andric N, Ping T, Xie F, Oermans S, Gos-
sen JA, Ascoli M. Ovulation involves the luteinizing hor-
mone-dependent activation of G(q/11) in granulosa cells.
Mol Endocrinol. 2013;27:1483-91. PMID: 23836924 DOI:
http://dx.doi.org/10.1210/me.2013-1130
Brower PT, Schultz RM. Intercellular communication be-
tween granulosa cells and mouse oocytes: existence
and possible nutritional role during oocyte growth.
Dev Biol. 1982;90:144-53. PMID: 7199496 DOI:
http://dx.doi.org/10.1016/0012-1606(82)90219-6
Buell M, Chitwood JL, Ross PJ. cAMP modulation during sheep
in vitro oocyte maturation delays progression of meiosis with-
out aecting oocyte parthenogenetic developmental compe-
tence. Anim Reprod Sci. 2015;154:16-24. PMID: 25595334
DOI: http://dx.doi.org/10.1016/j.anireprosci.2014.12.012
Chen J, Chi MM, Moley KH, Downs SM. cAMP pulsing
of denuded mouse oocytes increases meiotic resump-
tion via activation of AMP-activated protein kinase. Re-
production. 2009;138:759-70. PMID: 19700529 DOI:
http://dx.doi.org/10.1530/REP-08-0535
Child TJ, Phillips SJ, Abdul-Jalil AK, Gulekli B, Tan SL.
A comparison of in vitro maturation and in vitro fertil-
ization for women with polycystic ovaries. Obstet Gy-
necol. 2002;100:665-70. PMID: 12383531 DOI:
http://dx.doi.org/10.1097/00006250-200210000-00009
Chmelíková E, Jeseta M, Sedmíková M, Petr J, Tumová L,
Kott T, Lipovová P, Jílek F. Nitric oxide synthase isoforms and
the eect of their inhibition on meiotic maturation of porcine
oocytes. Zygote. 2010;18:235-44. PMID: 20109267 DOI:
http://dx.doi.org/10.1017/S0967199409990268
da Silveira JC, Veeramachaneni DN, Winger QA, Carnev-
ale EM, Bouma GJ. Cell-secreted vesicles in equine ovar-
ian follicular uid contain miRNAs and proteins: a pos-
sible new form of cell communication within the ovarian
follicle. Biol Reprod. 2012;86:71. PMID: 22116803 DOI:
http://dx.doi.org/10.1095/biolreprod.111.093252
da Silveira JC, de Andrade GM, Nogueira MF, Meirelles FV,
Perecin F. Involvement of miRNAs and Cell-Secreted Ves-
icles in Mammalian Ovarian Antral Follicle Development.
Reprod Sci. 2015;22:1474-83. PMID: 25736328 DOI:
http://dx.doi.org/10.1177/1933719115574344
De La Fuente R, Eppig JJ. Transcriptional activi-
ty of the mouse oocyte genome: companion granulo-
sa cells modulate transcription and chromatin remodel-
ing. Dev Biol. 2001;229:224-36. PMID: 11133166 DOI:
http://dx.doi.org/10.1006/dbio.2000.9947
de Loos F, Kastrop P, Van Maurik P, Van Beneden TH,
Kruip TA. Heterologous cell contacts and metabol-
ic coupling in bovine cumulus oocyte complexes. Mol
Reprod Dev. 1991;28:255-9. PMID: 2015084 DOI:
http://dx.doi.org/10.1002/mrd.1080280307
Dunning KR, Russell DL, Robker RL. Lipids and oocyte devel-
opmental competence: the role of fatty acids and ß-oxida-
tion. Reproduction. 2014;148:R15-27. PMID: 24760880 DOI:
http://dx.doi.org/10.1530/REP-13-0251
Dumesic DA, Meldrum DR, Katz-Jae MG, Krisher RL,
Schoolcraft WB. Oocyte environment: follicular u-
id and cumulus cells are critical for oocyte health. Fer-
til Steril. 2015;103:303-16. PMID: 25497448 DOI:
http://dx.doi.org/10.1016/j.fertnstert.2014.11.015
Egbert JR, Uliasz TF, Shuhaibar LC, Geerts A, Wunder F, Klei-
man RJ, Humphrey JM, Lampe PD, Artemyev NO, Rybalkin
SD, Beavo JA, Movsesian MA, Jae LA. Luteinizing Hor-
mone Causes Phosphorylation and Activation of the cGMP
Phosphodiesterase PDE5 in Rat Ovarian Follicles, Contrib-
uting, Together with PDE1 Activity, to the Resumption of
Meiosis. Biol Reprod. 2016;94:110. PMID: 27009040 DOI:
http://dx.doi.org/10.1095/biolreprod.115.135897
Elvin JA, Yan C, Wang P, Nishimori K, Matzuk MM. Mo-
lecular characterization of the follicle defects in the
growth dierentiation factor 9-decient ovary. Mol En-
docrinol. 1999;13:1018-34. PMID: 10379899 DOI:
http://dx.doi.org/10.1210/mend.13.6.0309
Emori C, Sugiura K. Role of oocyte-derived paracrine factors
in follicular development. Anim Sci J. 2014;85:627-33. PMID:
24717179 DOI: http://dx.doi.org/10.1111/asj.12200
Eppig JJ. Intercommunication between mammalian oocytes and
companion somatic cells. Bioessays. 1991;13:569-74. PMID:
1772412 DOI: http://dx.doi.org/10.1002/bies.950131105
Eppig JJ, Chesnel F, Hirao Y, O’Brien MJ, Pendola FL, Watanabe
S, Wigglesworth K. Oocyte control of granulosa cell develop-
ment: how and why. Hum Reprod. 1997;12:127-32. PMID:
9433969
Eppig JJ, Pendola FL, Wigglesworth K, Pendola JK.
Mouse oocytes regulate metabolic cooperativity be-
tween granulosa cells and oocytes: amino acid trans-
port. Biol Reprod. 2005;73:351-7. PMID: 15843493 DOI:
http://dx.doi.org/10.1095/biolreprod.105.041798
Eppig JJ, O’Brien MJ, Wigglesworth K, Nicholson A, Zhang
W, King BA. Eect of in vitro maturation of mouse oo-
cytes on the health and lifespan of adult ospring.
Hum Reprod. 2009;24:922-8. PMID: 19151027 DOI:
http://dx.doi.org/10.1093/humrep/den466
41
Cyclic nucleotides oocytes maturation - Botigelli, R C
JBRA Assist. Reprod. | v.21 | no1| Jan-Feb-Mar/ 2017
Faerge I, Mayes M, Hyttel P, Sirard MA. Nuclear ultrastructure
in bovine oocytes after inhibition of meiosis by chemical and
biological inhibitors. Mol Reprod Dev. 2001;59:459-67. PMID:
11468783 DOI: http://dx.doi.org/10.1002/mrd.1053
Franciosi F, Coticchio G, Lodde V, Tessaro I, Modina SC, Fadini R,
Dal Canto M, Renzini MM, Albertini DF, Luciano AM. Natriuretic
peptide precursor C delays meiotic resumption and sustains
gap junction-mediated communication in bovine cumulus-en-
closed oocytes. Biol Reprod. 2014;91:61. PMID: 25078681
DOI: http://dx.doi.org/10.1095/biolreprod.114.118869
Francis SH, Blount MA, Corbin JD. Mammalian cyclic nucleotide
phosphodiesterases: molecular mechanisms and physiologi-
cal functions. Physiol Rev. 2011;91:651-90. PMID: 21527734
DOI: http://dx.doi.org/10.1152/physrev.00030.2010
Fulka J Jr, Leibfried-Rutledge ML, First NL. Eect of 6-dime-
thylaminopurine on germinal vesicle breakdown of bovine
oocytes. Mol Reprod Dev. 1991;29:379-84. PMID: 1888517
DOI: http://dx.doi.org/10.1002/mrd.1080290410
Funahashi H, Cantley TC, Day BN. Synchronization of meiosis
in porcine oocytes by exposure to dibutyryl cyclic adenosine
monophosphate improves developmental competence follow-
ing in vitro fertilization. Biol Reprod. 1997;57:49-53. PMID:
9209079 DOI: http://dx.doi.org/10.1095/biolreprod57.1.49
Gardner DK, Harvey AJ. Blastocyst metabolism. Re-
prod Fertil Dev. 2015;27:638-54. PMID: 25751298 DOI:
http://dx.doi.org/10.1071/RD14421
Gharibi Sh, Hajian M, Ostadhosseini S, Hosseini SM, Forouzan-
far M, Nasr-Esfahani MH. Eect of phosphodiesterase type 3 in-
hibitor on nuclear maturation and in vitro development of ovine
oocytes. Theriogenology. 2013;80:302-12. PMID: 23683693
DOI: http://dx.doi.org/10.1016/j.theriogenology.2013.04.012
Gilchrist RB. Recent insights into oocyte-follicle cell interactions
provide opportunities for the development of new approach-
es to in vitro maturation. Reprod Fertil Dev. 2011;23:23-31.
PMID: 21366977 DOI: http://dx.doi.org/10.1071/RD10225
Gilchrist RB, Ritter LJ, Armstrong DT. Mouse oocyte mitogenic ac-
tivity is developmentally coordinated throughout folliculogene-
sis and meiotic maturation. Dev Biol. 2001;240:289-98. PMID:
11784064 DOI: http://dx.doi.org/10.1006/dbio.2001.0451
Gilchrist RB, Thompson JG. Oocyte maturation: emerging con-
cepts and technologies to improve developmental potential in
vitro. Theriogenology. 2007;67:6-15. PMID: 17092551 DOI:
http://dx.doi.org/10.1016/j.theriogenology.2006.09.027
Gudermann T, Birnbaumer M, Birnbaumer L. Evidence for dual
coupling of the murine luteinizing hormone receptor to adeny-
lyl cyclase and phosphoinositide breakdown and Ca2+ mobi-
lization. Studies with the cloned murine luteinizing hormone
receptor expressed in L cells. J Biol Chem. 1992;267:4479-
88. PMID: 1311310
Guigon CJ, Magre S. Contribution of germ cells to the dierenti-
ation and maturation of the ovary: insights from models of germ
cell depletion. Biol Reprod. 2006;74:450-8. PMID: 16339043
DOI: http://dx.doi.org/10.1095/biolreprod.105.047134
Guimarães AL, Pereira SA, Leme LO, Dode MA. Evalua-
tion of the simulated physiological oocyte maturation sys-
tem for improving bovine in vitro embryo production.
Theriogenology. 2015;83:52-7. PMID: 25447152 DOI:
http://dx.doi.org/10.1016/j.theriogenology.2014.07.042
Guixue Z, Luciano AM, Coenen K, Gandol F, Sirard MA.
The inuence of cAMP before or during bovine oocyte mat-
uration on embryonic developmental competence. Ther-
iogenology. 2001;55:1733-43. PMID: 11393223 DOI:
http://dx.doi.org/10.1016/S0093-691X(01)00516-7
Harris AL. Emerging issues of connexin channels: biophysics lls
the gap. Q Rev Biophys. 2001;34:325-472. PMID: 11838236
DOI: https://doi.org/10.1017/S0033583501003705
Hyttel P, Sinowatz F, Vejlsted M, Betteridge K. Essentials of
Domestic Animal Embryology. Philadelphia: Saunders; 2010.
Juneja SC, Barr KJ, Enders GC, Kidder GM. Defects in
the germ line and gonads of mice lacking connexin43.
Biol Reprod. 1999;60:1263-70. PMID: 10208994 DOI:
http://dx.doi.org/10.1095/biolreprod60.5.1263
Kawashima I, Okazaki T, Noma N, Nishibori M, Yamashi-
ta Y, Shimada M. Sequential exposure of porcine cu-
mulus cells to FSH and/or LH is critical for appropriate
expression of steroidogenic and ovulation-related genes
that impact oocyte maturation in vivo and in vitro. Re-
production. 2008;136:9-21. PMID: 18456902 DOI:
http://dx.doi.org/10.1530/REP-08-0074
Kubelka M, Motlík J, Schultz RM, Pavlok A. Butyrolactone I
reversibly inhibits meiotic maturation of bovine oocytes,
Without inuencing chromosome condensation activi-
ty. Biol Reprod. 2000;62:292-302. PMID: 10642565 DOI:
http://dx.doi.org/10.1095/biolreprod62.2.292
Labrecque R, Sirard MA. The study of mammalian oocyte
competence by transcriptome analysis: progress and chal-
lenges. Mol Hum Reprod. 2014;20:103-16. PMID: 24233546
DOI: http://dx.doi.org/10.1093/molehr/gat082
Lonergan P, Khatir H, Carolan C, Mermillod P. Bovine blasto-
cyst production in vitro after inhibition of oocyte meiotic re-
sumption for 24 h. J Reprod Fertil. 1997;109:355-65. PMID:
9155746 DOI: http://dx.doi.org/10.1530/jrf.0.1090355
Lonergan P, Rizos D, Gutierrez-Adan A, Fair T, Boland
MP. Oocyte and embryo quality: eect of origin, culture
conditions and gene expression patterns. Reprod Do-
mest Anim. 2003;38:259-67. PMID: 12887565 DOI:
http://dx.doi.org/10.1046/j.1439-0531.2003.00437.x
Luciano AM, Modina S, Vassena R, Milanesi E, Lauria A,
Gandol F. Role of intracellular cyclic adenosine 3’,5’-mo-
nophosphate concentration and oocyte-cumulus cells
communications on the acquisition of the developmental
competence during in vitro maturation of bovine oocyte.
Biol Reprod. 2004;70:465-72. PMID: 14568913 DOI:
http://dx.doi.org/10.1095/biolreprod.103.020644
Luciano AM, Pocar P, Milanesi E, Modina S, Rieger D, Lau-
ria A, Gandol F. Eect of dierent levels of intracellular
cAMP on the in vitro maturation of cattle oocytes and
their subsequent development following in vitro fertiliza-
tion. Mol Reprod Dev. 1999;54:86-91. DOI: https://doi.
org/10.1002/(SICI)1098-2795(199909)54:1<86::AID-
MRD13>3.0.CO;2-C
Macaulay AD, Gilbert I, Caballero J, Barreto R,
Fournier E, Tossou P, Sirard MA, Clarke HJ, Khand-
jian ÉW, Richard FJ, Hyttel P, Robert C. The ga-
metic synapse: RNA transfer to the bovine oocyte.
Biol Reprod. 2014;91:90. PMID: 25143353 DOI:
http://dx.doi.org/10.1095/biolreprod.114.119867
42
Review Article
JBRA Assist. Reprod. | v.21 | no1| Jan-Feb-Mar/ 2017
Macaulay AD, Gilbert I, Scantland S, Fournier E, Ash-
kar F, Bastien A, Saadi HA, Gagné D, Sirard MA, Khand-
jian ÉW, Richard FJ, Hyttel P, Robert C. Cumulus Cell Tran-
scripts Transit to the Bovine Oocyte in Preparation for
Maturation. Biol Reprod. 2016;94:16. PMID: 26586844 DOI:
http://dx.doi.org/10.1095/biolreprod.114.127571
Mattioli M, Galeati G, Barboni B, Seren E. Concentration of
cyclic AMP during the maturation of pig oocytes in vivo and in
vitro. J Reprod Fertil. 1994;100:403-9. PMID: 8021856 DOI:
http://dx.doi.org/10.1530/jrf.0.1000403
Mayes MA, Sirard MA. Eect of type 3 and type 4 phosphodi-
esterase inhibitors on the maintenance of bovine oocytes in
meiotic arrest. Biol Reprod. 2002;66:180-4. PMID: 11751280
DOI: http://dx.doi.org/10.1095/biolreprod66.1.180
Méduri G, Charnaux N, Driancourt MA, Combettes L,
Granet P, Vannier B, Loosfelt H, Milgrom E. Follicle-stim-
ulating hormone receptors in oocytes? J Clin Endocri-
nol Metab. 2002;97:2266-76. PMID: 11994374 DOI:
http://dx.doi.org/10.1210/jc.87.5.2266
Mermillod P, Tomanek M, Marchal R, Meijer L. High developmen-
tal competence of cattle oocytes maintained at the germinal
vesicle stage for 24 hours in culture by specic inhibition of MPF
kinase activity. Mol Reprod Dev. 2000;55:89-95. DOI: https://
doi.org/10.1002/(SICI)1098-2795(200001)55:1<89::AID-
MRD12>3.0.CO;2-M
Miles JR, McDaneld TG, Wiedmann RT, Cushman RA, Ech-
ternkamp SE, Vallet JL, Smith TP. MicroRNA expression pro-
le in bovine cumulus-oocyte complexes: possible role of
let-7 and miR-106a in the development of bovine oocytes.
Anim Reprod Sci. 2012;130:16-26. PMID: 22269106 DOI:
http://dx.doi.org/10.1016/j.anireprosci.2011.12.021
Muñoz M, Uyar A, Correia E, Díez C, Fernandez-Gonzalez
A, Caamaño JN, Martínez-Bello D, Trigal B, Humblot P, Pon-
sart C, Guyader-Joly C, Carrocera S, Martin D, Marquant
Le Guienne B, Seli E, Gomez E. Prediction of pregnancy
viability in bovine in vitro-produced embryos and recipi-
ent plasma with Fourier transform infrared spectroscopy.
J Dairy Sci. 2014;97:5497-507. PMID: 24997663 DOI:
http://dx.doi.org/10.3168/jds.2014-8067
Nakamura Y, Yamagata Y, Sugino N, Takayama H, Kato
H. Nitric oxide inhibits oocyte meiotic maturation. Biol
Reprod. 2002;67:1588-92. PMID: 12390892 DOI:
http://dx.doi.org/10.1095/biolreprod.102.005264
Nogueira D, Cortvrindt R, De Matos DG, Vanhoutte L, Smitz
J. Eect of phosphodiesterase type 3 inhibitor on devel-
opmental competence of immature mouse oocytes in vi-
tro. Biol Reprod. 2003;69:2045-52. PMID: 12930710 DOI:
http://dx.doi.org/10.1095/biolreprod.103.021105
Nogueira D, Cortvrindt R, Everaerdt B, Smitz J. Eects
of long-term in vitro exposure to phosphodiesterase
type-3 inhibitors on follicle and oocyte development. Re-
production. 2005;130:177-86. PMID: 16049155 DOI:
http://dx.doi.org/10.1095/biolreprod.105.040485
Nogueira D, Ron-EI R, Friedler S, Schachter M, Raziel A,
Cortvrindt R, Smitz J. Meiotic arrest in vitro by phosphodi-
esterase 3-inhibitor enhances maturation capacity of hu-
man oocytes and allows subsequent embryonic develop-
ment. Biol Reprod. 2006;74:177-84. PMID: 16207840 DOI:
http://dx.doi.org/10.1095/biolreprod.105.040485
Norris RP, Ratzan WJ, Freudzon M, Mehlmann LM, Krall
J, Movsesian MA, Wang H, Ke H, Nikolaev VO, Jae LA.
Cyclic GMP from the surrounding somatic cells regu-
lates cyclic AMP and meiosis in the mouse oocyte. De-
velopment. 2009;136:1869-78. PMID: 19429786 DOI:
http://dx.doi.org/10.1242/dev.035238
Oliveira e Silva I, Vasconcelos RB, Caetano JV, Gulart LV,
Camargo LS, Báo SN, Rosa e Silva AA. Induction of re-
versible meiosis arrest of bovine oocytes using a two-step
procedure under dened and nondened conditions. Ther-
iogenology. 2011;75:1115-24. PMID: 21220166 DOI:
http://dx.doi.org/10.1016/j.theriogenology.2010.11.021
Picton HM. Activation of follicle development: the primordial
follicle. Theriogenology. 2001;55:1193-210. PMID: 11327680
DOI: http://dx.doi.org/10.1016/S0093-691X(01)00478-2
Pires PR, Santos NP, Adona PR, Natori MM, Schwarz
KR, de Bem TH, Leal CL. Endothelial and inducible ni-
tric oxide synthases in oocytes of cattle. Anim Re-
prod Sci. 2009;116:233-43. PMID: 19327921 DOI:
http://dx.doi.org/10.1016/j.anireprosci.2009.02.019
Ponderato N, Crotti G, Turini P, Duchi R, Galli C, Lazzari G. Em-
bryonic and foetal development of bovine oocytes treated with
a combination of butyrolactone I and roscovitine in an enriched
medium prior to IVM and IVF. Mol Reprod Dev. 2002;62:513-8.
PMID: 12112585 DOI: http://dx.doi.org/10.1002/mrd.10134
Potter LR. Guanylyl cyclase structure, function and regula-
tion. Cell Signal. 2011;23:1921-6. PMID: 21914472 DOI:
http://dx.doi.org/10.1016/j.cellsig.2011.09.001
Rajagopalan-Gupta RM, Lamm ML, Mukherjee S, Rasenick
MM, Hunzicker-Dunn M. Luteinizing hormone/choriogonado-
tropin receptor-mediated activation of heterotrimeric guanine
nucleotide binding proteins in ovarian follicular membranes.
Endocrinology. 1998;139:4547-55. PMID: 28200179 DOI:
http://dx.doi.org/10.1210/en.139.11.4547
Raposo G, Stoorvogel W. Extracellular vesi-
cles: exosomes, microvesicles, and friends. J Cell
Biol. 2013;200:373-83. PMID: 23420871 DOI:
http://dx.doi.org/10.1083/jcb.201211138
Razza E, Pedersen H, Stroebech L, Machado M, Nogueira
M, Kadarmideen H, Callesen H, Hyttel P. 192 Prematuration
of bovine cumulus-oocyte complexes with cylic adenosine
monophosphate modulators aects both oocyte and blasto-
cyt ultrastructure. Reprod Fertil Dev. 2015;28:227. DOI:
http://dx.doi.org/10.1071/RDv28n2Ab192
Richani D, Wang X, Zeng HT, Smitz J, Thompson JG,
Gilchrist RB. Pre-maturation with cAMP modulators in con-
junction with EGF-like peptides during in vitro matura-
tion enhances mouse oocyte developmental competence.
Mol Reprod Dev. 2014;81:422-35. PMID: 24488930 DOI:
http://dx.doi.org/10.1002/mrd.22307
Richards JS, Russell DL, Ochsner S, Espey LL. Ovulation: new di-
mensions and new regulators of the inammatory-like response.
Annu Rev Physiol. 2002;64:69-92. PMID: 11826264 DOI:
http://dx.doi.org/10.1146/annurev.physiol.64.081501.131029
Rose RD, Gilchrist RB, Kelly JM, Thompson JG, Sutton-McDowall
ML. Regulation of sheep oocyte maturation using cAMP modu-
lators. Theriogenology. 2013;79:142-8. PMID: 23102843 DOI:
http://dx.doi.org/10.1016/j.theriogenology.2012.09.020
43
Cyclic nucleotides oocytes maturation - Botigelli, R C
JBRA Assist. Reprod. | v.21 | no1| Jan-Feb-Mar/ 2017
Sadler SE, Maller JL. A similar pool of cyclic AMP phosphodi-
esterase in Xenopus oocytes is stimulated by insulin, insu-
lin-like growth factor 1, and [Val12,Thr59]Ha-ras protein. J
Biol Chem. 1989;264:856-61. PMID: 2463250
Santiquet N, Papillon-Dion E, Djender N, Guillemette C,
Richard FJ. New elements in the C-type natriuretic peptide
signaling pathway inhibiting swine in vitro oocyte meiotic re-
sumption. Biol Reprod. 2014a;91:16. PMID: 24899572 DOI:
http://dx.doi.org/10.1095/biolreprod.113.114132
Santiquet N, Greene AF, Schoolcraft WB, Krisher RL.
313 Pre-in vitro maturation with cylic amp and cyclic
gmp modulators improves developmental competece of
mouse oocytes. Reprod Fertil Dev. 2014b;27:245. DOI:
http://dx.doi.org/10.1071/RDv27n1Ab313
Santonocito M, Vento M, Guglielmino MR, Battaglia R, Wahl-
gren J, Ragusa M, Barbagallo D, Borzì P, Rizzari S, Maugeri M,
Scollo P, Tatone C, Valadi H, Purrello M, Di Pietro C. Molecu-
lar characterization of exosomes and their microRNA cargo in
human follicular uid: bioinformatic analysis reveals that exo-
somal microRNAs control pathways involved in follicular mat-
uration. Fertil Steril. 2014;102:1751-61.e1. PMID: 25241362
DOI: http://dx.doi.org/10.1016/j.fertnstert.2014.08.005
Sasseville M, Albuz FK, Côté N, Guillemette C,
Gilchrist RB, Richard FJ. Characterization of nov-
el phosphodiesterases in the bovine ovarian follicle.
Biol Reprod. 2009;81:415-25. PMID: 19357367 DOI:
http://dx.doi.org/10.1095/biolreprod.108.074450
Sasseville M, Côté N, Gagnon MC, Richard FJ. Up-regu-
lation of 3’5’-cyclic guanosine monophosphate-specic
phosphodiesterase in the porcine cumulus-oocyte com-
plex aects steroidogenesis during in vitro maturation. En-
docrinology. 2008;149:5568-76. PMID: 18669600 DOI:
http://dx.doi.org/10.1210/en.2008-0547
Schwarz KR, Pires PR, Adona PR, Câmara de Bem TH, Leal
CL. Inuence of nitric oxide during maturation on bovine
oocyte meiosis and embryo development in vitro. Re-
prod Fertil Dev. 2008;20:529-36. PMID: 18462616 DOI:
http://dx.doi.org/10.1071/RD07209
Schwarz KR, Pires PR, de Bem TH, Adona PR, Leal CL. Con-
sequences of nitric oxide synthase inhibition during bovine
oocyte maturation on meiosis and embryo development. Re-
prod Domest Anim. 2010;45:75-80. PMID: 20137060 DOI:
http://dx.doi.org/10.1111/j.1439-0531.2008.01242.x
Sela-Abramovich S, Galiani D, Nevo N, Dekel N. Inhibition of rat
oocyte maturation and ovulation by nitric oxide: mechanism
of action. Biol Reprod. 2008;78:1111-8. PMID: 18337515
DOI: http://dx.doi.org/10.1095/biolreprod.107.065490
Shimada M, Nishibori M, Isobe N, Kawano N, Terada T. Lutein-
izing hormone receptor formation in cumulus cells surrounding
porcine oocytes and its role during meiotic maturation of por-
cine oocytes. Biol Reprod. 2003;68:1142-9. PMID: 12606491
DOI: http://dx.doi.org/10.1095/biolreprod.102.010082
Shu YM, Zeng HT, Ren Z, Zhuang GL, Liang XY, Shen HW, Yao
SZ, Ke PQ, Wang NN. Eects of cilostamide and forskolin on
the meiotic resumption and embryonic development of imma-
ture human oocytes. Hum Reprod. 2008;23:504-13. PMID:
1821603 DOI: http://dx.doi.org/10.1093/humrep/dem344
Shuhaibar LC, Egbert JR, Norris RP, Lampe PD, Nikolaev
VO, Thunemann M, Wen L, Feil R, Jae LA. Intercellular
signaling via cyclic GMP diusion through gap junctions
restarts meiosis in mouse ovarian follicles. Proc Natl
Acad Sci U S A. 2015;112:5527-32. PMID: 25775542
DOI: http://dx.doi.org/10.1073/pnas.1423598112
Simon AM, Goodenough DA, Li E, Paul DL. Female infertility
in mice lacking connexin 37. Nature. 1997;385:525-9. PMID:
9020357 DOI: http://dx.doi.org/10.1038/385525a0
Sirard MA, First NL. In vitro inhibition of oocyte nuclear mat-
uration in the bovine. Biol Reprod. 1988;39:229-34. PMID:
2460146 DOI: http://dx.doi.org/10.1095/biolreprod39.2.229
Sirard MA, Richard F, Mayes M. Controlling meiot-
ic resumption in bovine oocytes: a review. Theriog-
enology. 1998;49:483-97. PMID: 10732029 DOI:
http://dx.doi.org/10.1016/S0093-691X(97)00420-2
Smitz JE, Thompson JG, Gilchrist RB. The promise of in vitro
maturation in assisted reproduction and fertility preservation.
Semin Reprod Med. 2011;29:24-37. PMID: 21207332 DOI:
http://dx.doi.org/10.1055/s-0030-1268701
Sohel MM, Hoelker M, Noferesti SS, Salilew-Wondim D,
Tholen E, Looft C, Rings F, Uddin MJ, Spencer TE, Schel-
lander K, Tesfaye D. Exosomal and Non-Exosomal Trans-
port of Extra-Cellular microRNAs in Follicular Fluid:
Implications for Bovine Oocyte Developmental Compe-
tence. PLoS One. 2013;8:e78505. PMID: 24223816 DOI:
https://doi.org/10.1371/journal.pone.0078505
Su YQ, Sugiura K, Wigglesworth K, O’Brien MJ, Aourtit JP,
Pangas SA, Matzuk MM, Eppig JJ. Oocyte regulation of meta-
bolic cooperativity between mouse cumulus cells and oocytes:
BMP15 and GDF9 control cholesterol biosynthesis in cumu-
lus cells. Development. 2008;135:111-21. PMID: 18045843
DOI: http://dx.doi.org/10.1242/dev.009068
Sugiura K, Pendola FL, Eppig JJ. Oocyte control of metabolic co-
operativity between oocytes and companion granulosa cells: en-
ergy metabolism. Dev Biol. 2005;279:20-30. PMID: 15708555
DOI: http://dx.doi.org/10.1016/j.ydbio.2004.11.027
Sutton ML, Cetica PD, Beconi MT, Kind KL, Gilchrist RB,
Thompson JG. Inuence of oocyte-secreted factors and cul-
ture duration on the metabolic activity of bovine cumulus cell
complexes. Reproduction. 2003;126:27-34. PMID: 12814344
DOI: http://dx.doi.org/10.1530/rep.0.1260027
Sutton-McDowall ML, Gilchrist RB, Thompson JG. The pivotal
role of glucose metabolism in determining oocyte develop-
mental competence. Reproduction. 2010;139:685-95. PMID:
20089664 DOI: http://dx.doi.org/10.1530/REP-09-0345
Thomas RE, Armstrong DT, Gilchrist RB. Dierential eects
of specic phosphodiesterase isoenzyme inhibitors on bovine
oocyte meiotic maturation. Dev Biol. 2002;244:215-5. PMID:
11944932 DOI: http://dx.doi.org/10.1006/dbio.2002.0609
Thomas RE, Armstrong DT, Gilchrist RB. Bovine cumu-
lus cell-oocyte gap junctional communication during in
vitro maturation in response to manipulation of cell-spe-
cic cyclic adenosine 3’,5’-monophosophate levels.
Biol Reprod. 2004a;70:548-56. PMID: 14568915 DOI:
http://dx.doi.org/10.1095/biolreprod.103.021204
44
Review Article
JBRA Assist. Reprod. | v.21 | no1| Jan-Feb-Mar/ 2017
Thomas RE, Thompson JG, Armstrong DT, Gilchrist RB. Eect
of specic phosphodiesterase isoenzyme inhibitors during in
vitro maturation of bovine oocytes on meiotic and developmen-
tal capacity. Biol Reprod. 2004b;71:1142-9. PMID: 15189837
DOI: http://dx.doi.org/10.1095/biolreprod.103.024828
Tichovská H, Petr J, Chmelíková E, Sedmíková M,
Tůmová L, Krejcová M, Dörerová A, Rajmon R. Ni-
tric oxide and meiotic competence of porcine oocytes.
Animal. 2011;5:1398-405. PMID: 22440285 DOI:
http://dx.doi.org/10.1017/S1751731111000565
Tsafriri A, Chun SY, Zhang R, Hsueh AJ, Conti M. Oocyte
maturation involves compartmentalization and oppos-
ing changes of cAMP levels in follicular somatic and germ
cells: studies using selective phosphodiesterase inhibi-
tors. Dev Biol. 1996;178:393-402. PMID: 8812137 DOI:
http://dx.doi.org/10.1006/dbio.1996.0226
Tsuji T, Kiyosu C, Akiyama K, Kunieda T. CNP/NPR2 signaling
maintains oocyte meiotic arrest in early antral follicles and is
suppressed by EGFR-mediated signaling in preovulatory fol-
licles. Mol Reprod Dev. 2012;79:795-802. PMID: 22987720
DOI: http://dx.doi.org/10.1002/mrd.22114
Ulloa SM, Heinzmann J, Herrmann D, Timmermann B,
Baulain U, Großfeld R, Diederich M, Lucas-Hahn A, Nie-
mann H. Eects of dierent oocyte retrieval and in vitro
maturation systems on bovine embryo development and
quality. Zygote. 2015;23:367-77. PMID: 24423448 DOI:
http://dx.doi.org/10.1017/S0967199413000658
van den Hurk R, Zhao J. Formation of mammalian oocytes and
their growth, dierentiation and maturation within ovarian folli-
cles. Theriogenology. 2005;63:1717-51. PMID: 15763114 DOI:
http://dx.doi.org/10.1016/j.theriogenology.2004.08.005
Vanhoutte L, De Sutter P, Nogueira D, Gerris J, Dhont M, Van der
Elst J. Nuclear and cytoplasmic maturation of in vitro matured
human oocytes after temporary nuclear arrest by phosphodi-
esterase 3-inhibitor. Hum Reprod. 2007;22:1239-46. PMID:
17303631 DOI: http://dx.doi.org/10.1093/humrep/dem007
Vanhoutte L, Nogueira D, Dumortier F, De Sutter P. As-
sessment of a new in vitro maturation system for mouse
and human cumulus-enclosed oocytes: three-dimensional
prematuration culture in the presence of a phosphodies-
terase 3-inhibitor. Hum Reprod. 2009;24:1946-59. PMID:
19395363 DOI: http://dx.doi.org/10.1093/humrep/dep104
Vanhoutte L, Nogueira D, Gerris J, Dhont M, De Sut-
ter P. Eect of temporary nuclear arrest by phospho-
diesterase 3-inhibitor on morphological and function-
al aspects of in vitro matured mouse oocytes. Mol
Reprod Dev. 2008;75:1021-30. PMID: 18163445 DOI:
http://dx.doi.org/10.1002/mrd.20851
Varani S, Elvin JA, Yan C, DeMayo J, DeMayo FJ, Hor-
ton HF, Byrne MC, Matzuk MM. Knockout of pen-
traxin 3, a downstream target of growth dier-
entiation factor-9, causes female subfertility. Mol
Endocrinol. 2002;16:1154-67. PMID: 12040004 DOI:
http://dx.doi.org/10.1210/mend.16.6.0859
Wright PT, Schobesberger S, Gorelik J. Studying GPCR/
cAMP pharmacology from the perspective of cellular struc-
ture. Front Pharmacol. 2015;6:148. PMID: 26236239 DOI:
https://doi.org/10.3389/fphar.2015.00148
Young JM, McNeilly AS. Theca: the forgotten cell of the ovarian
follicle. Reproduction. 2010;140:489-504. PMID: 20628033
DOI: http://dx.doi.org/10.1530/REP-10-0094
Zeng HT, Ren Z, Guzman L, Wang X, Sutton-McDow-
all ML, Ritter LJ, De Vos M, Smitz J, Thompson JG,
Gilchrist RB. Heparin and cAMP modulators interact
during pre-in vitro maturation to aect mouse and hu-
man oocyte meiosis and developmental competence.
Hum Reprod. 2013;28:1536-45. PMID: 23559189 DOI:
http://dx.doi.org/10.1093/humrep/det086
Zeng HT, Richani D, Sutton-McDowall ML, Ren Z, Smitz
JE, Stokes Y, Gilchrist RB, Thompson JG. Prematuration
with cyclic adenosine monophosphate modulators alters
cumulus cell and oocyte metabolism and enhances de-
velopmental competence of in vitro-matured mouse oo-
cytes. Biol Reprod. 2014;91:47. PMID: 24966394 DOI:
http://dx.doi.org/10.1095/biolreprod.114.118471
Zhang J, Wei Q, Cai J, Zhao X, Ma B. Eect of C-Type
Natriuretic Peptide on Maturation and Developmen-
tal Competence of Goat Oocytes Matured In Vitro.
PLoS One. 2015;10:e0132318. PMID: 26151446 DOI:
https://doi.org/10.1371/journal.pone.0132318
Zhang M, Su YQ, Sugiura K, Wigglesworth K, Xia G, Eppig JJ.
Estradiol promotes and maintains cumulus cell expression of na-
triuretic peptide receptor 2 (NPR2) and meiotic arrest in mouse
oocytes in vitro. Endocrinology. 2011;152:4377-85. PMID:
21914782 DOI: http://dx.doi.org/10.1210/en.2011-1118
Zhang M, Su YQ, Sugiura K, Xia G, Eppig JJ. Granulosa cell ligand
NPPC and its receptor NPR2 maintain meiotic arrest in mouse
oocytes. Science. 2010;330:366-9. PMID: 20947764 DOI:
http://dx.doi.org/10.1126/science.1193573
Zhang W, Yang Y, Liu W, Chen Q, Wang H, Wang X, Zhang
Y, Zhang M, Xia G. Brain natriuretic peptide and C-type na-
triuretic peptide maintain porcine oocyte meiotic arrest.
J Cell Physiol. 2014;230:71-81. PMID: 24912131 DOI:
http://dx.doi.org/10.1002/jcp.24682
... These events start with the binding of gonadotropins to their receptors and consequent activation of the associated G protein, which triggers a phosphorylation cascade in cyclic adenosine monophosphate (cAMP)-dependent protein kinases (Russell et al., 2016). Physiologically, in the follicular environment, cAMP is one of the main agents responsible for controlling the meiotic maturation of oocytes, constituting the second messenger of the gonadotropin signaling pathway in ovarian tissue (Botigelli et al., 2017). ...
... In both situations, when gonadotropins bind to their receptors, the activation of the associated G protein is initiated, inducing the conversion of guanosine triphosphate into GDP (guanosine diphosphate). Guanosine diphosphate then connects to the α subunit of the G protein, stimulating adenylate cyclase to generate cAMP, one of the main responsible factors for controlling oocyte meiotic maturation (Botigelli et al., 2017). ...
Article
Full-text available
O objetivo deste estudo foi avaliar o efeito da gonadotrofina coriônica equina (eCG) e da gonadotrofina coriônica humana (hCG), em substituição ao uso de hormônio folículo estimulante (FSH) na maturação in vitro (MIV) de oócitos ovinos. Após a coleta de ovários (n=300) ovinos, os complexos cúmulus-oócitos (CCOs) foram aspirados, selecionados e divididos em quatro grupos de acordo com o meio de MIV: grupo CON, em que foi utilizado o meio MIV base; e grupos ECG, HCG e FSH, em que os oócitos foram imersos em meio MIV base adicionado de 10 UI/mL de eCG, 10 UI/mL de hCG e 10 µg/mL de FSH-p, respectivamente. A MIV dos oócitos foi realizada a 38,5°C, em atmosfera umidificada de 5% de CO2 em ar, durante 24 horas. Posteriormente, os oócitos foram avaliados, quanto grau de expansão das células do cumulus, configuração da cromatina, níveis de GSH e mitocôndrias ativas. Não foram observadas diferenças significativas com relação à taxa de expansão de células do cumulus. A percentagem de oócitos em MII foi maior no grupo ECG do que no grupo CON e HCG (P<0,05) e semelhante ao grupo FSH. Em conclusão, a eCG pode ser utilizada em substituição ao FSH na MIV de oócitos ovinos.
... Sheep breeding is an important enterprise in animal production yielding foods such as meat and milk which are very important for people's nourishment all over the world (Rather et al., 2020) there has been meat consumption increased throughout the course of recent years and this growth attributed to increases in per person consumption of sheep meat (Godfray et al., 2018). In vitro embryo production (IVEP) systems as new reproductive technology may play an important role for the production of sheep in the future to accelerate sheep breeding and improve the efficiency of production (Botigelli et al., 2017;Gharban, 2021). Also to increase the productivity in the small ruminant industry the genetic material of these species should be improved so, the in vitro embryo production could be important technology to reach this goal by combining selected male and female gametes (Paramio et al., 2016). ...
... The IVEP allows the production of a high and cheap number of embryos essential for studies which need a high number of embryos using ovaries recovered from slaughterhouse as a source of oocyte such as sexing, stem cells, cloning and in general studies needing a high number of embryos (Paramio and Izquierdo, 2014). There are currently facing many technical challenges in improving the efficiency of the ovine IVEP systems such as low efficiency and poor quality of embryos, so need to find solutions to overcome the problems for that the system (Botigelli et al., 2017). ...
Article
Full-text available
This study was designed to investigation the effect of oocytes collected methods (aspiration and slicing) to oocytes recovery per ovary and the quality oocytes (normal and abnormal) in local Iraqi ewes. The female genitalia was brought from Al-shulla abattoir from the animals after slaughtered, the samples was collected and transport to reproductive techniques laboratory with 1-3 hrs in thermal box containing normal saline (0.9%) supplemented with 100 IU/ml penicillin, and 1mg / ml streptomycin. In laboratory the ovaries washed in phosphate-buffered solution with antibiotics three times. Each time the oocytes collection in different method aspiration or slicing to compared in recovery rate and quality of oocyte collected after classification four group according number of cumulus layers and uniform the cytoplasm to (A + B) when contained multiple layer of cumulus cells with homogenous cytoplasm considered normal oocytes and (C+D) when absent cumulus cells or degenerated with heterogeneous cytoplasm considered abnormal oocytes in two different methods. The result of this current study showed the high significant effect (P≤0.01) in recovery rate to slicing than aspiration (4.91, 3.23) oocyte /ovary respectively.
... The aim to use cAMP modulators is to avoid premature nuclear maturation in vitro by means of maintaining higher concentration of cAMP within the ooplasm. This provides time for the COC to synchronize nuclear and cytoplasmic maturation, like that in vivo event that may result in more competent oocytes and embryo (Botigelli et al., 2017). Unsurprisingly, the mechanism of synthesis and hydrolysis of cGMP is one of the main targets of pharmacological strategies to control oocyte maturation. ...
... New approaches in in vitro maturation and their impacts in the resulting embryo: Based on the significant advances of the mechanisms that control oocyte maturation and their interaction with the CCs, new paths were opened to improve the IVM technique. One of which is the use of dynamic in vitro systems to improve embryo quality and quantity, the so-called pre-maturation or pre-IVM systems (Botigelli et al., 2017). (Sugimura et al, 2018) reported that the administration of follicle-stimulating hormone (FSH) prior to oocyte retrieval improves oocyte developmental competence. ...
Article
Full-text available
Cattle play a vital role in national economy; therefore, the cattle population must be raised by all possible ways of production. Maturation of oocytes outside the body of animal is principal method in dairy production field which is used to produce the mature oocytes for pre-implantation embryo development. This technology has great potential to increase efficiency for both clinically and commercially purposes. In the last few decades, the progress in in-vitro Maturation (IVM) of oocytes has been very slow. Many factors affects the IVM of oocytes. However pre-IVM with cAMP modulators could contribute to the acquisition of developmental competence by bovine oocytes from small antral follicles through the modulation of EGF receptor signaling and oocyte-cumulus/cumulus-cumulus gap junctional communication, however, the main aim of the review is to understand the ability of oocyte development and its control mechanism during in-vivo and in-vitro processes. In addition, the various perspectives of in-vitro maturation of oocytes in bovines have also been discussed.
... Ένα τέτοιο παράδειγμα αποτελεί και ο GDF9, όπου στην πολύ πρόσφατη μελέτη των Li et al., 2022 ερευνήθηκε και υποστηρίχθηκε η δυνατότητα του GDF9 (mRNA και πρωτεΐνη), από καλλιέργειες ανθρώπινων εμβρύων 3 ης ημέρας, να αποτελέσει μη επεμβατικό βιοδείκτη για την επιλογή των εμβρύων προς μεταφορά. Όπως άλλωστε καταγράφεται και σε άλλες μελέτες, η παρουσία κυτοκινών και αυξητικών παραγόντων είναι ζωτικής σημασίας για τον προσδιορισμό της ποιότητας των ωαρίων (Dumesic et al., 2015;Botigelli et al., 2017). ...
Thesis
Full-text available
During folliculogenesis, the interaction between the oocyte and the cells of the ovary is essential to achieve oocyte growth and maturation. Growth Differentiation Factor 9 (GDF9) is thought to play an important role in such process. GDF9 belongs to the TGF-β superfamily, produced mainly by the oocyte participates in the ovarian function by exerting paracrine and/or autocrine activity. The regulation of its expression is complex and species-specific. The aim of the present study was to investigate the dependence of the oocyte on granulosa cells of the corona radiata (Cumulus Cells, CC) regarding GDF9 protein production. Porcine ovaries were collected, and Cumulus Oocyte Complexes (COC), Denuded Oocytes (DO) and Cumulus cells (CC) were isolated from follicles 1-3mm and >3mm in diameter for each group respectively and cultured separately. After 48 and 96 hours of culture, the culture medium was collected and analyzed by the ELISA method to determine the amount of GDF9 produced by each cell culture. The results showed that COC and DO collected from follicles >3mm in diameter (COC B and DO B), produced a higher amount of GDF9 compared to oocytes collected from follicles 1-3mm in diameter cultured with or without their corona radiata (COC S and DO S) respectively. DO B produced more GDF9 than in the co-culture with their cumulus cells (COC B), indicating a reaction, of the mature oocyte, in the absence of the surrounding cumulus cells (CC B), which is expressed by excess secretion of GDF9 protein. Cumulus cells from small 1-3mm diameter follicles (CC S) produced more GDF9 than their oocytes (DO S) or their co-culture (COC S). This observation is a novel in this study, and as previously assumed, is a reaction of the small cumulus cells due to the absence of the surrounded oocyte. In contrast, cumulus cells from large follicles >3mm in diameter (CC B) produced less GDF9 than their oocytes (DO B). Differences in GDF9 production were also observed between 48 and 96 hours of culture. COC B produced less GDF9 at 48 than at 96 hours of culture, while CC B produced more at 48 than at 96 hours of culture. The results of the present study indicate a complex interdependence between the oocyte and the cumulus cells, even by measuring one growth differential factor (GDF9). This emphasizes the importance of the contribution of cumulus cells to maturation and the various processes that lead to a high-quality egg. There is also an incentive to reappraisal cumulus cells as co-culture cells or markers of the developmental competence of the oocyte in assisted reproductive technologies (ARTs).
... The cryopreservation and in vitro culture of ovaries and isolated preantral follicles have been introduced as an alternative approach for fertility preservation. Several factors such as oxidative stress, hypoxia, and temperature alterations may influence the survival and development of ovarian follicles during in vitro growth and many attempts have focused on improving these procedures (1)(2)(3). Moreover, in the literature, it has been shown that cell death may be involved in follicular damage and degeneration following ovarian cryopreservation and in vitro culture (2,(3)(4)(5)(6)(7). ...
Article
Full-text available
Background: Lysophosphatidic acid (LPA) contributes to follicular activation, oocyte maturation, in vitro fertilization, and embryo implantation. Objective: This study was designed to evaluate the effects of LPA to improve the development of isolated follicles derived from whole mouse cultured vitrified ovaries. Materials and methods: In this experimental study, first, the 1-wk-old mouse ovaries in the non-vitrified and vitrified groups were cultured in the presence of 20 µM of LPA for 1 wk. Then, their isolated preantral follicles were cultured individually for 12 days in the presence or absence of 40 µM of LPA. The following evaluations were done for the cultured follicles: a viability test using Calcein AM staining, flow cytometry using annexin V/Pi, and analysis of the expression of genes by real-time reverse transcription polymerase chain reaction. The maturation rates of the oocytes were compared among groups and some of the released metaphase II oocytes were subjected to in vitro fertilization. Results: In all LPA treated groups, the rates of survival and follicular development were higher, and the incidence of cell death and expression of pro-apoptotic genes were lower, than in the non-LPA supplemented groups (p = 0.035). There was no significant difference between the vitrified and non-vitrified groups regarding follicular or oocyte development, but the expression of Bad and LPA receptors genes was significantly altered in the vitrified LPA supplemented group in comparison with the non-vitrified LPA supplemented group (p = 0.028). Conclusion: LPA improved the survival and developmental potential of the isolated follicles. Despite some alterations in the expression of apoptosis-related genes in the vitrified ovaries, LPA had positive effects on the survival and development of these follicles.
... Хоча виробництво ембріонів in vitro супроводжується багатьма технічними проблемами (низька ефективність технології та низька якість ембріонів), воно залишається важливим, особливо для підвищення генетичного потенціалу овець порівняно з природним розмноженням і може бути використане для забезпечення сталого розвитку вів-чарства. Водночас останнім часом появилося багато позитивних відгуків щодо виробництва ембріонів in vitro в овець (Cognié et al., 2003;Cox & Alfaro, 2007;Amiridisa & Cseh, 2012;De Souza-Fabjan et al., 2014;Paramio & Izquierdo, 2016;Botigelli et al., 2017), у яких не лише описано досягнення в галузі, а й вказано напрям технології на майбутнє. ...
Article
Full-text available
New literature data on research aimed at improving the in vitro production of sheep embryos presents in the article. An analysis of the achievements of scientists from different countries to increase the efficiency of the main stages of embryo production in vitro: maturation of oocytes in vitro, their in vitro fertilization and in vitro embryo culture. In the literature experience has shown that the efficiency of oocyte maturation in vitro is significantly influenced by the experience and qualifications of scientists, the age of the egg donor, the improvement of the environment by adding roscovitin to inhibit meiosis, α-linolenic acid, cerium dioxide nanoparticles (CeO2 NPs) and sericin to accelerate nuclear maturation and increase the number of oocytes of the second meiotic metaphase (MII). The main factors influencing the effectiveness of in vitro fertilization have been identified, and the parameters of the limited time of fertilization ability of sperm and the ability of oocytes to fertilize, which is called the “fertile span”, have been determined. The main effective medium that increases the effectiveness of in vitro fertilization – synthetic oviduct fluid (SOF) with the addition of heparin and serum of cattle or sheep. The main parameters of sheep embryo culture in vitro are presented with the definition of the most commonly used media and their influence on embryonic development. Potential ways to improve the production of sheep embryos in vitro with the determination of morphological evaluation of categories of oocytes, methods of synchronization of their maturation in vitro are also highlighted. At the same time, literature data on the synchronization of oocyte-cumulus complexes with the use of a large number of inhibitors of meiotic division are presented, which according to many scientists may be a key factor in improving the efficiency of sheep embryo production in vitro. In addition, the results of studies of many scientists on the expansion of the fertile gap of oocytes of sheep cultured in vitro using certain biologically active substances were analyzed. In conclusion, the prospect of using the technology of in vitro production of sheep embryos in biomedical research is highlighted.
Article
This study investigated the effects of cyclic adenosine monophosphate modulating during cumulus-oocyte complexes (COCs) pre-maturation and the role of melatonin on in vitro maturation (IVM) of bovine COCs. In experiment one, COCs were pre-matured for 8 h in control medium or with 3-isobutyl-1-methylxanthine (IBMX) and forskolin, IBMX and C-type natriuretic peptide, c-type natriuretic peptide and forskolin or IBMX, forskolin and c-type natriuretic peptide. Then, meiotic progression was evaluated. In experiment two, COCs were pre-matured, followed by IVM in control medium alone or with 10-6, 10-7 or 10-8 M melatonin. After IVM, chromatin configuration, transzonal projections (TZPs), reactive oxygen species, mitochondrial distribution, ultrastructure and mRNA expression for antioxidant enzymes were evaluated. In experiment 1, COCs pre-matured with both C-type natriuretic peptide and forskolin or C-type natriuretic peptide, forskolin and IBMX had lower meiotic resumption rate when compared to control. Considering that IBMX had not an additional effect to potentiate inhibition of meiotic resumption, a combination of C-type natriuretic peptide and forskolin was chosen. In experiment 2, COCs matured with 10-8 M melatonin had greater rates of meiotic resumption when compared to the other treatments (P < 0.05). The COCs matured with 10-7 or 10-8 M melatonin had greater mitochondrial activity (P < 0.05), while those matured with 10-6 or 10-8 M of melatonin had greater levels of TZPs. Ultrastructure of oocyte and cumulus cells after IVM with melatonin was relatively well preserved. COCs matured with 10-8 M melatonin increased mRNA expression for superoxide dismutase (SOD) and catalase (CAT) (P < 0.05), when compared to non-cultured and pre-matured COCs, respectively. In conclusion, bovine COC pre-maturation with C-type natriuretic peptide and forskolin, followed by IVM with 10-8 M melatonin improves meiotic resumption rates, TZPs, mitochondrial distribution and mRNA expression for SOD and CAT.
Article
During the past decades, pharmaceutical companies have been making efforts to find alternative methods to animal-based tests. Several tests have been proposed as models for in vitro testing, including oocyte and bovine embryo produced in vitro in developmental toxicity screening. Under optimal experimental conditions, the in vitro maturation rate of bovine oocytes approaches 90%, the fertilisation rate is close to 80%; however, the success in progressing to blastocyst is almost half of those, attaining only 30–40%. It is also known that after in vivo insemination of normally cycling cows, approximately 85% of the ovulated oocytes will develop into an embryo. In contrast to this, in most in vitro production practises, only 15–20% of the oocytes punctured yield transferable embryos. In fact, from oocyte to embryo development, the competency of the female gamete is mandatory to assure the repeatability and feasibility of the results. The developmental competency of an oocyte can be influenced by several factors, including external components: such as the age of the cow, its nutritional status, body condition score, genetic merit for milk yield, proper function of intracellular molecular mechanisms, as well as the season. In this review, the factors influencing the quality of bovine oocytes that could possibly influence the success of producing embryos in vitro are highlighted.
Article
Full-text available
As the use of assisted reproductive technologies (ART) and in vitro embryo production (IVP) expand in the equine industry, it has become necessary to further our understanding of available semen selection techniques. This segment of our two-section review will focus on the selection of spermatozoa based on quality and sex for equine intracytoplasmic sperm injection (ICSI), as well as current and future developments in sperm sorting technologies. Ultimately, novel methods of semen selection will be assessed based on their efficacy in other species and their relevance and future application towards ARTs in the horse.
Chapter
How to Prepare the Egg and Embryo to Maximize IVF Success - edited by Gabor Kovacs January 2019
Article
Full-text available
The meiotic cell cycle of mammalian oocytes in preovulatory follicles is held in prophase arrest by diffusion of cGMP from the surrounding granulosa cells into the oocyte. Luteinizing hormone (LH) then releases meiotic arrest by lowering cGMP in the granulosa cells. The LH-induced reduction of cGMP is caused in part by a decrease in guanylyl cyclase activity, but the observation that the cGMP phosphodiesterase PDE5 is phosphorylated during LH signaling suggests that an increase in PDE5 activity could also contribute. To investigate this idea, we measured cGMP-hydrolytic activity in rat ovarian follicles. Basal activity was due primarily to PDE1A and PDE5, and LH increased PDE5 activity. The increase in PDE5 activity was accompanied by phosphorylation of PDE5 at serine 92, a protein kinase A/G consensus site. Both the phosphorylation and the increase in activity were promoted by elevating cAMP and opposed by inhibiting protein kinase A, supporting the hypothesis that LH activates PDE5 by stimulating its phosphorylation by protein kinase A. Inhibition of PDE5 activity partially suppressed LH-induced meiotic resumption as indicated by nuclear envelope breakdown, but inhibition of both PDE5 and PDE1 activities was needed to completely inhibit this response. These results show that activities of both PDE5 and PDE1 contribute to the LH-induced resumption of meiosis in rat oocytes, and that phosphorylation and activation of PDE5 is a regulatory mechanism.
Article
Full-text available
Oocytes resume meiosis spontaneously when subjected to in vitro maturation (IVM). Cyclic adenosine monophosphate (cAMP) elevating agents have been used for artificial blocking of meiotic resumption (pre-IVM) to allow the oocyte to prepare for maturation, potentially increasing its developmental competence. However, the ultrastructural effects of this pharmacological approach on oocytes and embryos remain to be addressed. We assessed the effects of pre-IVM with cAMP modulators in oocytes (10 for each group) at the end of IVM and in blastocyst (10 for each group) after 7 days of culture. Cumulus-oocyte complexes (COC) were subjected to pre-IVM for 2 h with forskolin (Sigma, St. Louis, MO, USA; 100 μM) and 3-isobutyl-1-methylxanthine) (IBMX, Sigma, 500 μM) followed by 24 h of IVM with FSH-enriched media (IVF Vet Solutions, Adelaide, Australia). Simultaneously, another group of COC was subjected to conventional IVM (con-IVM) for 24 h (EmbryoTransBiotech, Copenhagen, Denmark) with BSA (4 mg mL–1, Sigma), gentamycin (50 mg mL–1), and FSH (0.1 IU mL–1). Matured oocytes were collected for qualitative ultrastructural analysis or followed to IVF. The morphology was carefully evaluated on serially sectioned oocytes and embryos, where each serial section (~60.2-μm section per oocyte/embryo) was analysed under light microscopy. Subsequently, the equatorial section from each oocyte and the section giving the optimal representation of the inner cell mass in each blastocyst was re-embedded and sectioned for electron microcopy as previously described (Hyttel and Madsen 1987 Acta Anat. 129, 12–14). Blastocyst rates did not differ between groups. Ultrastructural analyses revealed subtle ultrastructural differences between pre-IVM and con-IVM conditions. In both groups, oocytes had matured to metaphase II. The perivitelline space of pre-IVM oocytes was significantly narrower than con-IVM. The cytoplasmic vesicles were more abundant and globally distributed in pre-IVM oocytes, whereas at con-IVM a vesicle-free periphery of the ooplasm was frequent, except for cortical granules and clusters of mitochondria associated with smooth endoplasmic reticulum (SER). We observed typical hooded mitochondria and cortical granules either clustered in the periphery or solitarily distributed in the cortical ooplasmic region for both groups. In the blastocysts, differences were noted with respect to especially distribution of ribosomes. In pre-IVM blastocysts, ribosomes were mostly organised in free clusters (polysomes) and peripherally located in cells of the inner cell mass. Con-IVM blastocysts showed ribosomes preferentially associated with the rough ER and often associated with mitochondria. Lipid droplets and rounded mitochondria were observed in both groups as well as apically located tight junctions and desmosomes between adjacent trophectoderm (TE) cells. Pleomorphic and elongated mitochondria were abundant in the TE of pre-IVM blastocysts, whereas the mitochondrial population was more homogenous at con-IVM. These findings suggest that pre-IVM for 2 h affects oocyte and blastocyst ultrastructure. Research was supported by grants 12/50533-2 and 12/23409-9 from FAPESP.
Article
Full-text available
So far, the characteristics of a good quality egg have been elusive, similar to the nature of the physiological, cellular, and molecular cues leading to its production both in vivo and in vitro. Current understanding highlights a strong and complex interdependence between the follicular cells and the gamete. Secreted factors induce cellular responses in the follicular cells, and direct exchange of small molecules from the cumulus cells to the oocyte through gap-junctions control meiotic arrest. Studying the interconnection between the cumulus cells and the oocyte, we previously demonstrated that the somatic cells also contribute transcripts to the gamete. Here we show that these transcripts can be visualized moving down the transzonal projections (TZPs) to the oocyte, and that a time course analysis revealed progressive RNA accumulation in the TZPs, indicating that RNA transfer occurs before the initiation of meiosis resumption under a time table fitting with the acquisition of developmental competence. A comparison of the identity of the nascent transcripts trafficking in the TZPs, to those in the oocyte increasing in abundance during maturation, and that are present on the oocyte's polyribosomes, revealed transcripts common to all three fractions suggesting the utilization of transferred transcripts for translation. Furthermore, the removal of potential RNA trafficking by stripping the cumulus cells caused a significant reduction in maturation rates indicating the need for the cumulus cell RNA transfer to the oocyte. These results offer a new perspective to t he determinants of oocyte quality and female fertility, as well as provide insight that may eventually be used to improve IVM conditions.
Article
Full-text available
Signal transduction via G-protein coupled receptors (GPCRs) relies upon the production of cAMP and other signaling cascades. A given receptor and agonist pair, produce multiple effects upon cellular physiology which can be opposite in different cell types. The production of variable cellular effects via the signaling of the same GPCR in different cell types is a result of signal organization in space and time (compartmentation). This organization is usually based upon the physical and chemical properties of the membranes in which the GPCRs reside and the repertoire of downstream effectors and co-factors that are available at that location. In this review we explore mechanisms of GPCR signal compartmentation and broadly review the state-of-the-art methodologies which can be utilized to study them. We provide a clear rationale for a “localized” approach to the study of the pharmacology and physiology of GPCRs and particularly the secondary messenger cAMP.
Article
Full-text available
The developmental competence of oocytes during in vitro maturation (IVM) is compromised due to asynchronous nuclear and cytoplasmic maturation. To improve IVM efficiency, a pre-maturation culture or two-step maturation strategy has been established, involving meiosis arrest induced by pharmacological agents to provide oocytes with sufficient time to synchronize the maturation of the nucleus and cytoplasm. C-type natriuretic peptide (CNP), which has been demonstrated to function as an oocyte maturation inhibitor (OMI) in many species, provides a new alternative to improve the developmental capacity of oocytes matured in vitro. However, the effect of CNP on meiosis arrest and the maturation of goat oocytes remains unclear. In the present study, CNP was shown to function as an OMI in goat oocytes. CNP could temporarily maintain the meiotic arrest of goat oocytes cultured in vitro for 4 hours. This transient effect was partly due to the reduction of natriuretic peptide receptor 2 (Npr2). Estradiol could delay the decrease in Npr2 expression and prolong the duration of meiosis arrest up to 6 hours. Based on the above results, a two-step method was established for goat oocyte maturation, in which the oocyte maturation rate was significantly increased. After parthenogenetic activation, the cleavage rate, blastocyst rate and total cell number of blastocysts were significantly improved. Our results suggested that CNP can be used to delay meiotic resumption and enhance the developmental competence of goat oocytes matured in vitro.
Article
Full-text available
The connexins are a family of at least 15 proteins that form the intercellular membrane channels of gap junctions. Numerous connexins, including connexin43 (Cx43), have been implicated in reproductive processes by virtue of their expression in adult gonads. In the present study, we examined the gonads of fetal and neonatal mice homozygous for a null mutation in the Gja1 gene encoding Cx43 to determine whether the absence of this connexin has any consequences for gonadal development. We found that in both sexes at the time of birth, the gonads of homozygous mutants were unusually small. This appears to be caused, at least in part, by a deficiency of germ cells. The germ cell deficiency was traced back as far as Day 11.5 of gestation, implying that it arises during early stages of germ line development. We also used an organ culture technique to examine postnatal folliculogenesis in the mutant ovaries, an approach necessitated by the fact that Gja1 null mutant offspring die soon after birth because of a heart abnormality. The results demonstrated that folliculogenesis can proceed to the primary (unilam-inar) follicle stage in the absence of Cx43 but that subsequent development is impaired. In neonatal ovaries of normal mice, Cx43 could be detected in the somatic cells as early as Day 1, when primordial follicles begin to appear, supporting the conclusion that this connexin is required for the earliest stages of folliculogenesis. These results imply that gap junctional coupling mediated by Cx43 channels plays indispensable roles in both germ line development and postnatal folliculogenesis.
Article
Several in vitro systems using cumulus oocyte complexes (COCs) recovered from early antral follicles have been designed to permit meiotic maturation of oocytes in the pig and other species. To date, these have been partially but not completely successful, however the in vitro matured oocytes exhibit limited developmental competence as compared with that in oocytes matured in vivo. Therefore, this study was undertaken to determine the precise temporal patterns of steroid hormone production and the expression of specific genes in follicular cell types in vivo and then apply these patterns to stimulate oocyte maturation in COCs in culture. For this, follicular fluid, granulosa cells and cumulus cells were collected from early stage, antral and ovulatory follicles at specific time intervals following eCG and hCG treatment of gilts. The results showed several similarities but also distinct differences from the well-characterized patterns in mouse and rat models. As in the rodent, FSH/eCG stimulated expression of Cyp19 mRNA and increased production of estrogen coordinately with up-regulated proliferation of granulosa cells and cumulus cells. However, unlike the rodent, eCG induced the expression of Lhcgr and Pgr in porcine cumulus cells as well as granulosa cells and progesterone accumulation was detected in follicular fluid prior to the LH/hCG surge in the pig model. Moreover, progesterone and PGR were critical for FSH-induced expression of Lhcgr mRNA in cumulus cells of cultured COCs. The expression of Lhcgr mRNA in cumulus cells was associated with the appearance of functional LH receptors and the ability of LH to induce, in cumulus cells as well as granulosa cells, prostaglandin production, release of EGF like factors and ADAMTS-1 expression, promoting COCs expansion and oocyte maturation. Based on the unique expression and regulation of Pgr and Lhcgr in cumulus cells, we designed a novel porcine COC culture system in which hormones were added sequentially to mimic changes observed in vivo. Specifically, COCs from early stage follicles were pre-cultured with FSH and estradiol for 10h at which time progesterone was added for another 10h. After 20 hr, COCs were moved to fresh medium containing LH, EGF and progesterone. The COCs exhibited improved cumulus cell proliferation (and no apoptosis), greater expansion and more highly regulated changes in oocyte maturation. Moreover, the oocytes matured in this revised COC culture system exhibited greater developmental competence to the blastocyst stage. From these results, we conclude that to achieve optimal COC expansion and oocyte maturation in culture the unique gene expression patterns in cumulus cells of each species, such as mouse, pig and human need to be characterized and used to increase the effectiveness of hormone stimulation regimens in each species. Supported in part by JSPS-18688016 (MS)
Article
High cAMP levels during in vitro maturation (IVM) have been related to improved blastocyst yields. Here, we employed the cAMP/cGMP modulators, forskolin, IBMX, and cilostamide, during IVM to unravel the role of high cAMP in early embryonic development produced from prepubertal and adult bovine oocytes. Oocytes were collected via transvaginal aspiration and randomly assigned to three experimental groups: TCM24 (24h IVM/control), cAMP30 (2h pre-IVM (forskolin-IBMX), 30h IVM-cilostamide), and DMSO30 (Dimethyl Sulfoxide/vehicle control). After IVM, oocytes were fertilized in vitro and zygotes were cultured in vitro to blastocysts. Meiotic progression, cAMP levels, mRNA abundance of selected genes and DNA methylation were evaluated in oocytes. Blastocysts were used for gene expression or DNA methylation analyses. Blastocysts from the cAMP30 groups were transferred to recipients. The cAMP elevation delayed meiotic progression, but developmental rates were not increased. In immature oocytes, mRNA abundance of PRKACA was higher for cAMP30 protocol and no differences were found for PDE3A, SMAD2, ZAR1, PRDX1 and SLC2A8. EGR1 gene was up-regulated in prepubertal cAMP30 immature oocytes and down-regulated in blastocysts from all in vitro treatments. A similar gene expression profile was observed for DNMT3b, BCL2L1, PRDX1 and SLC2A8 in blastocysts. Satellite DNA methylation profiles were different between prepubertal and adult oocytes and blastocysts derived from the TCM24 and DMSO30 groups. Blastocysts obtained from prepubertal and adult oocytes in the cAMP30 treatment displayed normal methylation profiles and produced offspring. These data indicate that cAMP regulates IVM in prepubertal and adult oocytes in a similar manner, with impact on the establishment of epigenetic marks and acquisition of full developmental competency.
Article
The LH/CG receptor signals to adenylyl cyclase via the stimulatory heterotrimeric GTP binding regulatory protein, Gs, and to phospholipase C and potentially to other effectors, such as ion channels, via a G protein or proteins that have not been identified in gonadal cells. To identify G proteins activated in a physiological membrane environment upon LH/CG receptor activation, we used the ability of activated G proteins to bind GTP and incubated ovarian follicular membranes with the photoaffinity GTP analog, P³-(4-azidoanilido)-P¹-5′-GTP ([³²P]AAGTP). Results showed that human CG (hCG) stimulated the binding of [³²P]AAGTP to a 45-kDa protein(s) in follicular membranes that comigrated with immunoreactive Gαs, Gαq/11, and Gα13. When Gα proteins were immunoprecipitated from Triton X-100 solubilized membrane extracts after photoaffinity labeling with [³²P]AAGTP, a time-dependent increase in hCG-dependent [³²P]AAGTP binding to Gαs, Gαq/11, and Gαi was detected. hCG-dependent [³²P]AAGTP binding to Gα13 was also detected. These results demonstrate that agonist-dependent LH/CG receptor activation promotes the activation of Gs, Gi, Gq/11, and G13 in porcine ovarian follicular membranes. These results further suggest that Gαs remains coupled to the agonist-bound LH/CG receptor during at least the initial 10 min after agonist-dependent receptor activation.