You are on page 1of 11

REPRODUCTION

RESEARCH

C-type natriuretic peptide enhances mouse preantral


follicle growth
Guangyin Xi1,*, Wenjing Wang1,*, Sarfaraz A Fazlani1,2,*, Fusheng Yao1, Mingyao Yang1,
Jing Hao1, Lei An1 and Jianhui Tian1
1
Ministry of Agriculture Key Laboratory of Animal Genetics, Breeding and Reproduction, National Engineering
Laboratory for Animal Breeding, College of Animal Sciences and Technology, China Agricultural University, Beijing,
China and 2Lasbela University of Agriculture, Water and Marine Science, Lasbela, Balochistan, Pakistan
Correspondence should be addressed to J Tian; Email: tianjh@cau.edu.cn
*(G Xi, W Wang and S A Fazlani contributed equally to this work)

Abstract
Compared to ovarian antral follicle development, the mechanism underlying preantral follicle growth has not been well documented.
Although C-type natriuretic peptide (CNP) involvement in preantral folliculogenesis has been explored, its detailed role has not been
fully defined. Here, we used mouse preantral follicles and granulosa cells (GCs) as a model for investigating the dynamic expression
of CNP and natriuretic peptide receptor 2 (NPR2) during preantral folliculogenesis, the regulatory role of oocyte-derived growth
factors (ODGFs) in natriuretic peptide type C (Nppc) and Npr2 expression, and the effect of CNP on preantral GC viability. Both
mRNA and protein levels of Nppc and Npr2 were gradually activated during preantral folliculogenesis. CNP supplementation in
culture medium significantly promoted the growth of in vitro-cultured preantral follicles and enhanced the viability of cultured GCs
in a follicle-stimulating hormone (FSH)-independent manner. Using adult and prepubertal mice as an in vivo model, CNP pre-
treatment via intraperitoneal injection before conventional superovulation also had a beneficial effect on promoting the ovulation
rate. Furthermore, ODGFs enhanced Nppc and Npr2 expression in the in vitro-cultured preantral follicles and GCs. Mechanistic
study demonstrated that the regulation of WNT signaling and estrogen synthesis may be implicated in the promoting role of CNP in
preantral folliculogenesis. This study not only proves that CNP is a critical regulator of preantral follicle growth, but also provides
new insight in understanding the crosstalk between oocytes and somatic cells during early folliculogenesis.
Reproduction (2019) 157 445–455

Introduction of intraovarian factors, which include R-Spondin 2,


WNT, GDF9 (growth differentiation factor 9), BMP15
Folliculogenesis is divided into three stages (Orisaka
(bone morphogenetic protein 15) and KITL (KIT ligand),
et  al. 2013). The initial stage covers primordial to
are supportive factors of preantral follicle development
primary follicle transition, which is believed to be
(Eppig et  al. 2001, 2002, Boyer et  al. 2010, Cheng
independent of gonadotropin influence (Mcnatty et  al.
et  al. 2013), the molecular and cellular mechanisms
2007). The second stage covers preantral to antral follicle
transition. Although it is a gonadotropin-responsive responsible for this process have not been fully defined.
phase, folliculogenesis during this stage is primarily A recent study (Sato et al. 2012) showed evidence that
controlled by intraovarian regulators, not gonadotropins C-type natriuretic peptide (CNP) may play an important
(Cattanach et  al. 1977, Halpin et  al. 1986). In the last role as an upstream regulator in stimulating preantral
stage, growing follicles may be recruited to continue follicle growth. Using a cultured ovarian explant model,
their development until they are ovulated (Craig et  al. they reported that CNP supplementation in culture
2007) in a gonadotropin-dependent manner (Hunter medium-facilitated primary and early secondary follicle
et al. 2004). Compared with the well-documented role transition to the late secondary stage, which covers
of pituitary gonadotropins (e.g., follicle-stimulating preantral follicle development, in a dose-dependent
hormone (FSH) and luteinizing hormone (LH)) in manner (0.25–3  μM). In addition, in vivo studies,
regulating antral follicle development, intraovarian where juvenile and prepubertal mice were treated via
factors, which support autocrine and paracrine signals CNP intraperitoneal (i.p.) injection for 4  days, further
from oocytes and granulosa cells (GCs), may be more indicated the role of CNP in promoting follicle growth
important for preantral follicle growth. Although a series (Sato et al. 2012).

© 2019 Society for Reproduction and Fertility https://doi.org/10.1530/REP-18-0470


ISSN 1470–1626 (paper) 1741–7899 (online) Online version via https://rep.bioscientifica.com
Downloaded from Bioscientifica.com at 11/05/2020 04:09:28PM
via free access
446 G Xi, W Wang, S A Fazlani and others

CNP belongs to the natriuretic peptide family, which development. CNP facilitated the in vitro preantral
consists of three major types: atrial natriuretic peptide, follicle development and the viability of cultured GCs
brain natriuretic peptide and CNP. These peptides in a dose-dependent and FSH-independent manner.
show high structural homology, characterized by a Furthermore, based on the role of CNP in facilitating
highly conserved 17-member ring structure formed by preantral follicle development, in vivo study also
intramolecular disulfide linkage (Nishikimi et al. 2011). confirmed the beneficial effect of CNP on early follicle
Natriuretic peptides execute their biological function by growth. This observation also provides a promising
stimulating cyclic guanosine monophosphate (cGMP) reference for developing a novel strategy for inducing
production via guanylyl cyclase–coupled receptors ovulation by CNP-based pre-treatment in females. In
(Chinkers et  al. 1989). CNP acts exclusively through addition, the ODGFs regulated both Nppc and Npr2
natriuretic peptide receptor 2 (NPR2 or NRRB) to expression in preantral follicles, providing new insight
stimulate downstream cGMP signaling (Potter et  al. into understanding the crosstalk between oocytes and
2006). CNP–NPR2–cGMP signaling acts locally as a somatic cells during early folliculogenesis. Finally, we
positive regulator of endochondral ossification and is demonstrate that CNP may regulate WNT signaling and
essential for promoting physiological longitudinal bone enhance estrogen synthesis by modulating the expression
growth (Chusho et al. 2001). CNP is widely distributed of key ovarian genes (e.g., Wnt2b (wingless-type MMTV
in the body, including in the brain, chondrocytes and integration site family, member 2B), Wnt5a, Cyp11a1
endothelial cells and is considered a paracrine/autocrine (chrome P450, family 11, subfamily a, polypeptide 1)
regulator (Lumsden et al. 2010). More recently, genetic and Cdkn1a (cyclin-dependent kinase inhibitor 1A))
evidence has elucidated that loss-of-function mutation closely related to follicle development.
in Nppc and Npr2 results in female infertility due to
premature oocyte meiotic resumption (Zhang et  al.
Materials and methods
2010, Geister et  al. 2013). In Graafian follicles, CNP
produced by mural GCs stimulates cGMP generation by Animal studies and ethical approval
activating NPR2, which is expressed by cumulus cells All female ICR mice used in this study were from Beijing Vital
(CCs) surrounding and associating with oocytes and River Laboratory Animal Technology Co. Ltd. (Beijing, China).
diffuses into oocytes via gap junctions. In oocytes, cGMP The adult mice were maintained in a climate-controlled room
inhibits phosphodiesterase 3A, thereby preventing on a 12-h light/darkness cycle and allowed food and water ad
cAMP hydrolysis and maintaining meiotic arrest with libitum. The China Agricultural University Institutional Animal
high intra-oocyte cAMP levels (Zhang et al. 2010). The Care and Use Committee (XK662) approved this study and it
results of Sato et al. (Sato et al. 2012) are in accordance was performed in accordance with the committee guidelines.
with an earlier study (Mcgee et  al. 1997), in which All efforts were made to minimize animal suffering.
cGMP analogs promoted the development of cultured
rat preantral follicles; the authors hypothesized that
GCs collection and culture
cGMP pathway activators may be essential for preantral
follicle survival and development (Mcgee et  al. 1997). The preantral GCs were collected as previously reported
These findings not only support the possible role of CNP (Latham et al. 2004). Briefly, ICR mice (10–13 days old) were
in initiating antrum formation, but also implicate CNP killed by cervical dislocation. The ovaries were dissected in
regulation of preantral follicle response to gonadotropin, Dulbecco’s modified Eagle’s medium (DMEM)/F12 medium
which is essential for follicle development. (Gibco, Thermo Fisher Scientific) supplemented with 1%
CNP–NPR2 signaling is the critical determinant for fetal bovine serum (FBS; Gibco, Thermo Fisher Scientific),
oocyte meiotic arrest in antral follicles in different species 100 IU/mL penicillin (Invitrogen, Thermo Fisher Scientific) and
(Zhang et  al. 2010, Hiradate et  al. 2014, Zhong et  al. 100 µg/mL streptomycin sulfate (Invitrogen, Thermo Fisher
2015, Xi et al. 2018), and its regulatory mechanisms via Scientific) after three washes in physiological saline solution.
gonadotropins or oocyte-derived growth factors (ODGFs) After mechanical dissection, the follicles (100–130 µm) were
digested in medium containing 1  mg/mL collagenase IV
are well documented (Kawamura et al. 2011, Lee et al.
(Sigma), 0.025% trypsin (Gibco, Thermo Fisher Scientific)
2013). However, the upstream regulatory mechanism
and 0.02 mg/mL DNase I (Sigma) for 10 min at 37°C. The
of CNP–NPR2 signaling in preantral follicles and the GC–oocyte complexes were aspirated repeatedly in and
mechanism underlying the promoting effect on preantral out of a borosilicate glass pipette with an internal diameter
follicle growth remain to be elucidated. Accordingly, we slightly smaller than that of an oocyte. After two washes, the
investigated (1) CNP and NPR2 expression dynamics cells were seeded with DMEM/F12 culture medium (Gibco,
during preantral follicle development; (2) the effect of Thermo Fisher Scientific) supplemented with 10% FBS (Gibco,
ODGFs, as upstream regulators, on Nppc and Npr2 Thermo Fisher Scientific), 100 IU/mL penicillin (Invitrogen,
expression in mouse preantral follicles and (3) the Thermo Fisher Scientific) and 100  μg/mL streptomycin
effect of CNP on the viability of GCs isolated from sulfate (Invitrogen, Thermo Scientific) and cultured for 4 h
mouse preantral follicles. We show that CNP and NPR2 for adhesion. Subsequently, the cells were cultured in fresh
expression is gradually activated during preantral follicle medium with or without CNP, 8-Br-cGMP (a cGMP analog),

Reproduction (2019) 157 445–455 https://rep.bioscientifica.com

Downloaded from Bioscientifica.com at 11/05/2020 04:09:28PM


via free access
CNP stimulates ovarian GC viability 447

BMP15, GDF9 or FGF8 (fibroblast growth factor 8) according mare serum gonadotropin; Ningbo Second Hormone Factory).
to the different experimental treatments. The ovulated oocytes in the oviducts were counted after
16 h to evaluate ovulation efficiency. Meanwhile, oocyte
morphology was evaluated under an inverted microscope
Preantral follicle isolation and culture (Olympus). Nonviable oocytes had shape abnormalities, dark
The ovaries of prepubertal mice (13 days old) were aseptically cytoplasm, fragmented first polar body, ooplasm vacuolization
removed after the animals had been killed by cervical and fragments in the cytoplasm (Balaban & Urman 2006).
dislocation and placed in 3 mL prewarmed isolation medium Prepubertal mice (13  days old) were processed as described
consisting of L15 Leibovitz medium (Gibco, Thermo Fisher earlier, but were treated with 50 μg/kg body weight CNP. The
Scientific) supplemented with 10% FBS (Gibco, Thermo control group was injected with the same volume of normal
Fisher Scientific) and 1% antibiotics (100 units/mL penicillin saline instead of CNP.
and 100  μg/mL streptomycin; Invitrogen, Thermo Fisher
Scientific) (Orisaka et  al. 2006). Then, the preantral follicles
Cell viability assay
were dissected microscopically using 26-gauge needles. To
minimize experimental variation during the isolation, only Cell viability was assessed by measuring the conversion
follicles with two layers of GCs; a visible, centrally located of tetrazolium salt (WST-8) to formazan according to the
oocyte; 100–130-µm diameter, and which were enclosed by manufacturer’s instructions of an Enhanced CCK-8 assay kit
an intact basal membrane and had at least some attached (Beyotime Biotechnology, Jiangsu, China). The cells were
thecal cells were collected (Cortvrindt et  al. 1996). Instead plated onto 96-well plates at a density of 3 × 103 cells per well
of enzymatic digestion, the ovary and early preantral follicles and cultured for 24 h, followed by 3-day treatment without
(EPFs) were mechanically dissected to conserve all cell types (control) or with CNP (Sigma) and 8-Br-cGMP (Sigma). CCK-8
and receptor systems of the ovarian follicle for culture. assays were performed 24, 48 and 72 h after treatment. The
After three washes in isolation medium and two washes cells were washed with PBS (Gibco, Thermo Fisher Scientific),
in culture medium, the preantral follicles were cultured and then 200 μL CCK-8 solution was added to each well
individually in 100 µL culture medium in 96-well tissue culture and incubated for 3 h at 37°C. The optical density of each
plates for up to 4 days at 37°C in a humidified atmosphere of well at 450 nm was recorded on a Tecan microplate reader
5% CO2 in air (Cortvrindt et  al. 1998). The culture medium (Infinite M200, Tecan Nordic AB, Stockholm, Sweden). The
consisted of α-minimal essential medium (with 10 mM HEPES; results were calculated as the mean values of eight wells per
Gibco, Thermo Fisher Scientific) enriched with 1% ITS (5 µg/ treatment group.
mL insulin, 5 µg/mL transferrin, 5 ng/mL selenium; Sigma),
0.1% bovine serum albumin (BSA; Sigma), 100 µg/mL sodium
pyruvate and 1% antibiotics in the absence (control) or RNA extraction and quantitative real-time reverse
presence of CNP (N8768; Sigma-Aldrich) (Huang et al. 2011, transcription (RT)-PCR (RT-qPCR)
Zhong et  al. 2015, Zhang et  al. 2017). CNP was dissolved Preantral follicle or cultured cell RNA isolation and qRT-PCR
in phosphate-buffered saline (PBS; Gibco, Thermo Fisher were performed as previously described (Ren et al. 2015). In
Scientific) and stored at −20°C until used. For control cultures, summary, cultured cells in a six-well culture dish or about
an equal volume of PBS was added to the culture medium in 100–150 follicles were treated as indicated, rinsed twice
place of CNP. All selected follicles were pooled and randomly with cold PBS and collected in 1.0 mL TRIzol (Invitrogen);
divided over the culture conditions under study. Half the RNA was isolated as per the manufacturer’s instructions. The
medium was replaced with fresh medium every other day. extracted total RNA concentration and quality were assessed
The follicle morphological characteristics and diameters were by the absorbance at 260 nm (A260)/280 and A260/230
recorded every day as the average distance between the outer ratios as determined using a DS-11 spectrophotometer
edges of the basal membrane in two perpendicular planes (DeNovix, Wilmington, NC, USA). The RNA to be used
(Kobayashi et  al. 2009). To study ODGF regulation of Nppc for the next experiment should have an A260/280 ratio of
and Npr2 expression, the follicles were cultured in culture 1.8–2.0 and an A260/230 ratio of 2.0–2.2. Before RT, 1 μg
medium with 100 ng/mL BMP15 (R&D Systems), 100 ng/mL total RNA samples were digested with DNase I (Fermentas,
GDF9 (R&D Systems), 100 ng/mL FGF8 (R&D Systems) or Hanover, MD, USA) to remove contaminating genomic DNA.
combinations thereof for 24 h (Su et  al. 2003, Zhang et  al. The primer of this cDNA synthesis kit was an oligo-dT and
2010, Miyoshi et al. 2012, Machado et al. 2015). random primer mix. Aliquot RNA RT was performed using
a commercially available first-strand cDNA synthesis kit
(iScript cDNA Synthesis Kit; Bio-Rad Laboratories). The real-
In vivo CNP treatment
time PCR was performed in triplicate in a CFX96 Real-Time
Adult female mice (7  weeks old) were treated with CNP PCR System (Bio-Rad Laboratories) using SsoFast EvaGreen
(120 μg/kg body weight; N8768; Sigma-Aldrich) by i.p. Supermix (Bio-Rad Laboratories). The thermal cycling
injection daily for 4  days. CNP was dissolved in PBS and conditions were denaturation (95°C for 3 min), 40 cycles of
diluted by physiological saline solution before injection. amplification (95°C for 10 s) and quantification (61°C for 30 s)
The mice were injected with 5 IU hCG (human chorionic with a single fluorescence measurement and melting curve
gonadotropin; Ningbo Second Hormone Factory, Zhejiang, analysis (65–95°C with 0.5°C/s increments and continuous
China) after 48-h administration of 5 IU PMSG (pregnant fluorescence measurements). Table 1 summarizes the related

https://rep.bioscientifica.com Reproduction (2019) 157 445–455

Downloaded from Bioscientifica.com at 11/05/2020 04:09:28PM


via free access
448 G Xi, W Wang, S A Fazlani and others

Table 1  Primers for RT-PCR and qRT-PCR.

Gene Primer sequence (5′-3′) Tm (°C) Amplification efficiency (%) Accession number
Nppc Forward: GGTCTGGGATGTTAGTGCAGCTA 58 96.7 NM_010933.5
Reverse: TAAAAGCCACATTGCGTTGGA
Npr2 Forward: GCTGACCCGGCAAGTTCTGT 58 95.2 NM_173788.4
Reverse: ACAATACTCGGTGACAATGCAGAT
Wnt2b Forward: GACACGTCCTGGTGGTACATAGG 58 102.4 NM_009520.3
Reverse: TGGGTAGCGTTGACACAACTG
Wnt5a Forward: GCAGGCCGTAGGACAGTATACAA 60 100.9 NM_009524.4
Reverse: CGCCGCGCTATCATACTTCT
Cyp1a1 Forward: ATCCCCCACAGCACCACAA 58 97.8 NM_001136059.2
Reverse: AGTTCCCGGTCATGGTTAACC
Cyp11a1 Forward: GACGCATCAAGCAGCAAAATTC 58 99.1 NM_019779.4
Reverse: TCCACGATCTCCTCCAGCAT
Cdkn1a Forward: CTGTCTTGCACTCTGGTGTCTG 60 95.0 NM_007669.5
Reverse: AGAAATCTGTCAGGCTGGTCTG
Gapdh Forward: CCTGGAGAAACCTGCCAAGTAT 60 104.3 NM_008084.3
Reverse: GGAAGAGTGGGAGTTGCTGTTG

primer information. The specificity of the qRT-PCR products antibodies (0.2 μg/mL; Golden Bridge, Beijing, China). After
was confirmed with melting curve analysis. The amplification three washes in TBST, the membranes were incubated with
efficiency of each primer pair was calculated based on the enhanced chemiluminescence reagents (Millipore), exposed
slope of the standard curve. The amplification efficiency of digitally with Image Reader LAS-4000 (FujiFilm Life Science,
the primers used in our study was 95–105%. The relative Tokyo, Japan).
quantity of each gene was calculated by the comparative
threshold cycle (2−ΔΔCt) method as described previously (Livak
et  al. 2001). All experiments were repeated three or four Immunofluorescence staining
times using independent samples; the relative abundance of Mouse ovaries were fixed in 4% paraformaldehyde overnight
specific genes was normalized to the relative abundance of and stored at 4°C. The tissue was dehydrated and paraffin
glyceraldehyde-3-phosphate dehydrogenase (Gapdh) levels embedded and sections (5 μm) were cut with a microtome. The
(Xu et al. 2013, Sadr et al. 2015, Yan et al. 2019). slides were deparaffinized in xylene, and then rehydrated in
a series of ethanol dilutions. After dewaxing and rehydration,
antigen retrieval was performed in 0.01% sodium citrate
Protein extraction and western blotting buffer (pH 6.0), followed by cooling at room temperature
The CNP and NPR2 levels in the mouse preantral follicles at for at least 1 h. The sections were blocked in 0.5% BSA in
different stages were assayed by western blotting. About 250 PBS for 1 h at room temperature. Then, the sections were
preantral follicles were prepared by 20-min homogenization immunostained overnight at 4°C in a wet chamber with anti-
in ice-cold radioimmunoprecipitation assay lysis buffer CNP primary antibody (10 μg/mL; Abnova, Taiwan, China)
(CWBio Co., Ltd., Beijing, China) containing 1% Halt Protease and anti-NPR2 antibody (10 μg/mL; Abcam), followed by 1-h
and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific). incubation at room temperature with Alexa Fluor 594 Goat
The supernatant was collected after 10-min centrifugation Anti-Rabbit IgG (H+L) Secondary Antibody (1 μg/mL; Thermo
at 15,000 g at 4°C, and proteins were quantified using an Fisher Scientific) and DAPI (4′,6-diamidino-2-phenylindole;
Enhanced Bicinchoninic Acid Protein Assay Kit (Beyotime Thermo Fisher Scientific) counterstaining. The slides were
Biotechnology). The samples were denatured in the same imaged with a fluorescence microscope (Olympus). For the
volume of 2× Laemmli sample loading buffer (Bio-Rad negative control, rabbit IgG (immunoglobulin G, 10 μg/mL;
Laboratories) with 5% β-mercaptoethanol (Sigma) for 5 min Beyotime) was used in place of the primary antibody in
at 100°C and stored at −80°C for future use. Equal amounts parallel reactions.
of protein (about 20 µg) were loaded in each well for 12%
SDS-PAGE, and then transferred onto microporous PVDF
17β-Estradiol measurement
membranes (Millipore). Membranes containing the transferred
proteins were blocked with Tris-buffered saline containing Media cultured with individual mouse follicles were pooled
0.1% Tween 20 (TBST; 20 mM Tris–HCl, 150 mM NaCl and on the fourth day and stored at −80°C until the measurement
0.1% Tween 20, pH 7.6) and 5% skim milk for 1 h at room of estradiol levels. Estradiol levels in the collected culture
temperature. After three washes with TBST, the membranes media were determined by radioimmunoassay with the Iodine
were incubated overnight at 4°C with rabbit anti-CNP [125I] Estradiol Radioimmunoassy Kit (Beijing North Institute
polyclonal antibody (2  μg/mL; Santa Cruz Biotechnology of Biological Technology, Beijing, China). The measurements
Inc.), rabbit anti-NPR2 antibody (2 μg/mL; Abcam) and rabbit were taken in the General Hospital of the Nanjing Military
anti-GAPDH antibody (0.2 μg/mL; Sigma). After three washes Command, China. The sensitivity of the assay was 1.4 pg/mL.
with TBST, the membranes were incubated for 1 h at room The intra-assay coefficient of variation (CV) was 6.2% and the
temperature with horseradish peroxidase–linked secondary interassay CV was 8.9%.

Reproduction (2019) 157 445–455 https://rep.bioscientifica.com

Downloaded from Bioscientifica.com at 11/05/2020 04:09:28PM


via free access
CNP stimulates ovarian GC viability 449

Statistical analysis GCs. Unexpectedly, it appeared that CNP did not alter GC
viability at any concentration in culture conditions with
Statistical differences in the data were evaluated using
SPSS (SPSS Inc.). Dunnett’s t-test was used for statistical
typical 10% FBS supplementation (Fig. 3A). Considering
comparisons between two groups. Multiple groups were CNP is ubiquitously present in serum (Yagci et al. 2008,
compared statistically using one-way ANOVA, followed by Kake et al. 2009), we reduced FBS supplementation to
a least significant difference test. Significance was indicated 5% and 1% to exclude its possible influence (Fig.  3B
when P < 0.05. and C). With 1% FBS supplementation, the control
group also showed sustained, but lower viability than
that with 10% FBS supplementation. Under the 1%
Results FBS supplementation culture condition, 10, 100 and
Nppc and Npr2 were gradually activated during 500 nM CNP significantly increased preantral GC
preantral follicle development viability (Fig. 3C).
Next, as the biological function of CNP is dependent
Follicle growth and development is largely dependent on intracellular cGMP production via its exclusive
on the activation of a series of paracrine, autocrine receptor NPR2, we detected the effect of the cGMP
and endocrine signals. Nppc and Npr2 transcription analog (8-Br-cGMP) on preantral GC viability. At
were activated in the EPFs and late preantral follicles >50 µM, 8-Br-cGMP exhibited a significant viability-
(LPFs) of the prepubertal mice (Fig.  1A and B). promoting effect (Fig. 3D). Taken together, these findings
The immunofluorescence results confirmed these suggest that both CNP and cGMP benefit preantral GC
observations (Fig.  1C). Interestingly, qRT-PCR showed viability in vitro.
significantly increased Nppc and Npr2 transcripts
during early to late preantral follicle development
(Fig.  1D). Nppc is predominantly expressed in GCs, CNP pre-treatment enhanced the ovulation rate of
whereas Npr2 is primarily expressed in CCs (Zhang superovulated mice
et  al. 2010). Here, we also detected antral GCs and To detect the function of CNP for stimulating preantral
CCs as positive controls, which had higher Nppc and follicle development in vivo, adult (8-week-old)
Npr2 expression, respectively (Fig.  1D). These results and prepubertal mice (13-day-old) were treated
imply that, compared with antral follicles, in which high i.p. with CNP daily for 4  days, followed by a single
CNP levels are needed for maintaining oocyte meiotic i.p. injection of 5 IU equine chorionic gonadotropin
arrest, relatively lower CNP levels may be sufficient for for 48 h, and further treated with an ovulatory dose
promoting preantral follicle growth. Similarly, western of 5 IU hCG to examine superovulation efficiency.
blotting showed an upward trend of CNP and NPR2 in We divided the total ovulated oocytes based on the
LPFs (Fig. 1E). Together, these results indicated a gradual quality of the recovered oocytes: nonviable oocytes
upregulation of CNP and NPR2 during mouse preantral always presented a non-uniform or even fragmented
follicle growth. cytoplasm (Fig.  4A). CNP pre-treatment (120 μg/kg
body weight) increased ovulation in the adult mice,
CNP facilitated the growth of in vitro-cultured as revealed by the increased total and viable oocytes
preantral follicles (Fig.  4B). Similar results were obtained when the
prepubertal mice were pre-treated with CNP (50 μg/kg
We isolated preantral follicles (100–130 µm diameter) body weight) (Fig. 4C). These findings suggest the ability
from prepubertal mouse ovaries and treated them with of CNP to promote preantral follicle development to
0, 0.1, 1, 10, 100 or 500 nM CNP during in vitro culture. the early antral stage, thereby significantly enhancing
Follicle growth dynamics monitoring showed that the outcome of superovulation in both adult and
>10 nM CNP increased follicle diameter significantly prepubertal mice.
(Fig. 2A and B), as revealed by the significantly increased
follicle sizes without flattened morphology (Fig.  2A).
Therefore, the role of CNP in promoting preantral follicle ODGFs stimulated Nppc and Npr2 expression
growth was confirmed using the in vitro culture model To understand the mechanism responsible for Nppc and
of mouse preantral follicles. Npr2 expression regulation during early folliculogenesis,
we examined whether ODGFs modulate the two genes
in preantral follicles. Mouse preantral follicles and
CNP and 8-Br-cGMP enhanced preantral GC viability
preantral GCs were cultured in medium containing
Based on the above observations, we speculated that BMP15, GDF9, FGF8 or combinations thereof, for
the promoting effect of CNP on preantral follicle growth 24 h. Real-time RT-PCR revealed that the ODGFs had
may largely be due to its effect on preantral GC viability. promoting effects on Nppc expression in the preantral
Accordingly, we tested the effect of 0, 0.1, 10, 100 and follicles (Fig.  5A). These effects were also observed in
500 nM CNP on the viability of in vitro-cultured preantral the preantral GCs, except those cultured with FGF8,
https://rep.bioscientifica.com Reproduction (2019) 157 445–455

Downloaded from Bioscientifica.com at 11/05/2020 04:09:28PM


via free access
450 G Xi, W Wang, S A Fazlani and others

A C DAPI NPR2 Merge


EPFs LPFs EPFs LPFs
OO OO OO
pAF

OO OO OO
CCs
AF CCs CCs

GCs GCs GCs

DAPI CNP Merge


B GCs
OO
GCs
OO
GCs
OO

pAF
Nppc 250 bp
100 bp
Npr2 250 bp
100 bp GCs GCs GCs

Gapdh 250 bp AF OO OO OO
100 bp CCs CCs CCs

DAPI IgG Merge


D
** ** OO
* * pAF OO
4 5
Relative Nppc mRNA expression

Relative Npr2 mRNA expression

** **
4
3
3 E EPFs LPFs GCs
2 CNP
2
GAPDH
1 1 NPR2
0 0 GAPDH
EPFs LPFs GCs

Figure 1 Nppc and Npr2 expression in follicles of different diameters. (A) Representative morphology of EPFs and LPFs. Scale bar = 100 μm. (B)
RT-PCR detection of Nppc and Npr2 expression in EPFs and LPFs. For the blank control reaction, water was used as the template. For the no
reverse transcriptase (NRT) control, 100 ng RNA was used as the template to confirm the absence of genomic DNA contamination. (C)
Representative immunofluorescence images of CNP and NPR2 expression. Scale bar = 100 μm. AF, antral follicle; pAF, preantral follicle. (D)
Comparisons of relative expression levels of Nppc and Npr2 in EPFs and LPFs. Antral GCs and CCs were used as positive controls for detecting
Nppc and Npr2, respectively. Values are the means ± s.e.m. of four independent replicates. Four samples prepared from individual animals were
used. P values for qRT-PCR were obtained using one-way ANOVA, followed by a least significant difference test. *P < 0.05, **P < 0.01. (E)
Western blotting detection of protein abundance of CNP and NPR2 in EPFs, LPFs and GCs. Antral GCs were used as the positive control for
detecting NPR2.

which only increased Nppc expression mildly, but not the preantral follicles and GCs (Fig.  5B and D). These
significantly (Fig. 5C). It is worth noting that BMP15 plus findings demonstrate that ODGFs, which are important
GDF9 had a dramatic additive effect on Nppc expression regulators of preantral follicle development, can also
in the preantral GCs (Fig. 5C). Only BMP15 significantly manipulate CNP–NPR2 signaling in mouse preantral
enhanced the expression levels of Npr2 mRNA in both GCs at transcriptional level.

Reproduction (2019) 157 445–455 https://rep.bioscientifica.com

Downloaded from Bioscientifica.com at 11/05/2020 04:09:28PM


via free access
CNP stimulates ovarian GC viability 451

A B A 2
B 1.6
DMEM/F12+5%FBS
DMEM/F12+10%FBS
1.8
CNP Day 0 Day4

OD Value (450 nm)


190 ** 1.6

OD Value (450 nm)


1.2
1.4

0 nM
180 ** **
1.2 CNP CNP
1 0 nM 0.8 0 nM
0.8 500 nM 500 nM
170 **

Follicular Diameter/µm
0.6 100 nM 100 nM
** ** 10 nM 0.4 10 nM
0.4 1 nM
160 1 nM
0.1 nM 0.2 0.1 nM 0.1 nM
* 0 0
150 D0 D1 D2 D3 D0 D1 D2 D3
Time of Days Time of Days
140
1 nM C D
130 0.8 DMEM/F12+1%FBS ** 0.7
DMEM/F12+1%FBS
0.7 ** **
** 0.6

OD Value (450 nm)


120 * **

OD Value (450 nm)


0.6
0.5 **
D0 D1 D2 D3 D4 D5 ** **
10 nM 0.5 **
Time of Days CNP 0.4 cGMP
0.4 0 nM * 0 mM
0.3 **
0 nM 0.1 nM 1 nM 0.3 500 nM 1 mM
10 nM 100 nM 500 nM 100 nM 500 μM
0.2 0.2
10 nM 100 μM
100 nM 1 nM 0.1 50 μM
0.1
0.1 nM 10 μM
0 0
D0 D1 D2 D3 D0 D1 D2 D3
Time of Days Time of Days
500 nM

Figure 3 Effect of CNP and cGMP on GC viability in vitro. (A and B)


GC proliferation in CNP-treated high-serum culture medium (5 and
Figure 2 CNP promoted preantral follicle growth in vitro. (A) 10% FBS–DMEM/F12 medium) was not significantly different. (C) At
Representative images at day 0 and day 4 of cultured preantral 10, 100 and 500 nM, CNP significantly increased preantral GC
follicles with or without CNP treatment. Scale bar = 100 μm. (B) viability in low-serum (1% FBS) supplementation conditions. (D)
Diameters of preantral follicles with or without CNP treatment Preantral GCs cultured with 100 µM, 500 µM or 1 mM cGMP had
monitored daily from day 0 to day 4. Values are the means ± s.e.m. highly significant proliferation rates. Comparison was only performed
Four independent culture experiments were performed; there were at between the treatment groups and the control group. Five
least 100 follicles per group. Comparisons were performed between independent experiments were performed. Values are the
the treatment groups and the control group. Values are the means ± s.e.m. P values were obtained using Dunnett’s t-test.
means ± s.e.m. P values were obtained using Dunnett’s t-test. *P < 0.05, **P < 0.01.
*P < 0.05, **P < 0.01.
Discussion
The effect of CNP on the expression of key ovarian In female gametogenesis-related events, CNP is a well-
genes and steroidogenesis in mouse preantral follicles
established factor that plays a central role in regulating
To further elucidate the molecular mechanisms oocyte meiotic progress in growing follicles among
underlying the promoting effect of CNP on preantral different species (Zhang et al. 2010, Hiradate et al. 2014,
follicle development, we selected a set of key ovarian Zhong et al. 2015, Xi et al. 2018). In the present study, we
genes important for follicle growth and steroidogenesis focused on the function of CNP in promoting preantral
and detected the effect of CNP on their expression in follicle growth via enhanced GC viability. CNP–NPR2
cultured mouse preantral follicles. Preantral follicles were signaling was gradually activated upon early-to-late
treated with or without 200 nM CNP for 24 h before qRT- preantral follicle development (Fig.  1). The CNP and
PCR analyses of transcript levels. It should be noted that NPR2 expression dynamics during follicle development
the preantral growth promotion of 100 and 500 nM CNP provide a molecular basis for the promoting effect of
supplementation was not significantly different (Fig. 2). CNP, as an autocrine factor, on preantral follicle growth.
Therefore, an intermediate concentration (200 nM) was CNP treatment led to dose- and-time-dependent
used in the subsequent follicle culture. CNP increased increases in follicle size (Sato et al. 2012). Similarly, we
the expression of paracrine or autocrine factors that found that CNP stimulated preantral follicle growth in
contribute to follicle growth or GC viability (Wnt2b mice independent of FSH (Fig. 2). Correspondingly, LPFs
and Wnt5a) and a key steroidogenic enzyme (Cyp11a1) had significantly higher Nppc and Npr2 mRNA levels
(Fig.  6A). In contrast, CNP significantly repressed the than the EPFs (Fig.  1D). Considering GC viability is
expression of estrogen metabolic enzymes (Cyp1a1) essential for preantral follicle development (Zhang et al.
and cyclin-dependent kinase inhibitor (Cdkn1a/P21). 2011), we isolated preantral GCs and treated them with
Furthermore, the CNP-treated group had significantly CNP in vitro. CNP significantly promoted cell viability
increased 17β-estrogen concentrations in the medium (Fig. 3), and it is worth noting that this effect was also
at 4  days of culture compared with the control group independent of FSH or any other gonadotropins.
(Fig.  6B). These results demonstrate that CNP might CNP acts through guanylyl cyclase receptors, which
promote preantral follicle development by regulating activate intracellular cGMP production, one of the
WNT signaling and enhancing estrogen synthesis. most important second messengers. We show that, like

https://rep.bioscientifica.com Reproduction (2019) 157 445–455

Downloaded from Bioscientifica.com at 11/05/2020 04:09:28PM


via free access
452 G Xi, W Wang, S A Fazlani and others

A Viable oocytes Nonviable oocytes A Nppc B Npr2


a a
6 2

Relative mRNA expression


Relative mRNA expression
ab
5

Preantral follicles
b b
4 b b b
b b
3 c c 1
c
2 d d
d
1
0 0
BMP15 BMP15
GDF9 GDF9
FGF8 FGF8

C Nppc D
Npr2
4 a
3
B C

Relative mRNA expression

Relative mRNA expression


Adult mice Prepubertal mice a

3 b

Granulosa cells
2
* * b
Mean total number of oocytes per mouse

b b
35
n=20
* PBS 35 n=21
2 b b
b
Mean number of ooc y tes per mous e

c c b b b
c 1 b
n=20 CNP
30 30 1
n=20
0 0
25 n=20 25 n=22
BMP15 BMP15
GDF9 GDF9
20 20 FGF8 FGF8

15 15 Figure 5 The effects of ODGFs on Nppc and Npr2 expression in


preantral follicles and GCs. (A) Nppc and (B) Npr2 mRNA expression
10 10
in ODGF-treated preantral follicles. (C) Nppc and (D) Npr2 mRNA
5 5 expression in cultured GCs treated with combination ODGFs. Four
independent experiments were performed. The P values for qRT-PCR
0 0 were obtained using one-way ANOVA followed by a least significant
Total Viable PBS CNP difference test. Values are the means ± s.e.m. Lowercase letters (a, b, c)
indicate significant differences (P < 0.05).
Figure 4 Effects of CNP pre-treatment on superovulation in mice.
(A) Representative images of viable (arrows) and nonviable et  al. 2012). Preantral follicles gradually establish the
(arrowheads) ovulated oocytes. Scale bar = 100 μm. (B) The number of full ability to respond to gonadotropins during follicular
total and viable oocytes from adult mice with or without CNP antrum formation, so we speculate that CNP can respond
pre-treatment were significantly different. (C) The number of total to gonadotropins by accelerating the development of
oocytes from prepubertal mice with or without CNP pre-treatment
was significantly different. The number of mice per group is
more preantral follicles. Accordingly, we tested the
indicated. Values are the means ± s.e.m. The two-tailed Student t-test effect of CNP on the ovulation rate in both adult and
was used to evaluate the statistical significance. *P < 0.05. prepubertal mice using the standard superovulation

CNP, cGMP remarkably increased GC viability in vitro A B


** Control 25
(Fig.  3D). 8-Br-cGMP suppressed rat preantral follicle 4
200nM CNP
*
apoptosis cultured in serum-free conditions (Mcgee
Relative mRNA expression

et  al. 1997). The authors hypothesized that a potential 20


3
Estrogen (pg/ml)

activator of the cGMP pathway may be essential for *


** 15
preantral follicle survival and development (Mcgee
2
et al. 1997). We speculate that CNP may be an upstream
10
factor that controls cGMP content in mouse preantral ** **
follicles. Nonetheless, how CNP increases GC viability 1
5
and whether it can inhibit GC apoptosis remains to
be studied. 0 0
In addition to the evidence from the in vitro Control 200nM
CNP
experiments, the in vivo studies using 13-day-old
juvenile mice also demonstrated that CNP may
promote preantral follicle development and enhance Figure 6 CNP regulates the transcription of ovarian developmental
preantral follicle response to subsequent exogenous key genes and increases estrogen levels. (A) Expression levels of
gonadotropins and lead to the satisfactory outcome of representative genes involved in follicle development (Wnt2b,
Wnt5a, Cdkn1a) and estrogen metabolism (Cyp1a1 and Cyp11a1) in
ovulation induction (Fig.  4). Similarly, CNP treatment
preantral follicles without (Control) or with 200 nM CNP treatment in
of 21-day-old prepubertal mice facilitated early antral vitro. (B) Estrogen concentration in control and CNP groups on day 4.
follicle development to the preovulatory stage, thereby Four independent experiments were performed. Values are the
allowing LH/hCG ovulation induction to generate means ± s.e.m. The two-tailed Student t-test was used to evaluate the
fertilizable oocytes and successful pregnancy (Sato statistical significance. *P < 0.05, **P < 0.01.

Reproduction (2019) 157 445–455 https://rep.bioscientifica.com

Downloaded from Bioscientifica.com at 11/05/2020 04:09:28PM


via free access
CNP stimulates ovarian GC viability 453

method. CNP pre-treatment significantly improved (Abedini et  al. 2016). Therefore, Wnt5a expression is
the effect of superovulation in both groups of mice, as critical for normal preantral follicle development in
indicated by the increase in total ovulated and viable mice. Cdkn1a encodes the potent cyclin-dependent
oocytes. Based on the profound inhibitory effect of CNP kinase inhibitor P21, which can directly bind to cyclin/
on meiotic resumption, we have established a natural cyclin-dependent kinase 2 or 4 complexes and further
factor synchronized in vitro oocyte maturation system inhibit their activity, resulting in G1 arrest (Dutto
in our laboratory, which can significantly improve the et  al. 2015). More recently, it was reported that p21
developmental competence of matured oocytes, thereby inhibits ovarian GC proliferation (Jiang et  al. 2015).
resulting in higher in vitro bovine embryo production CNP treatment of preantral follicles may regulate
efficiency (Xi et  al. 2018). In view of the role of CNP the GC cell cycle by affecting p21 expression. In the
in promoting follicle growth, it may be used to replace future, it is necessary to focus on how CNP regulates
gonadotropin for treating patients with infertility. As p21 expression and its effect on the cell cycle. We
CNP is a natural small-molecule polypeptide present in observed that adding CNP to cultured preantral
the ovary, it may be possible to avoid adverse effects follicles enhanced Cyp11a1 expression, which is
stemming from the long half-life of gonadotropins, involved in estrogen synthesis. Consistent with our
such as ovarian hyperstimulation syndrome. Therefore, observations, estrogen concentration was increased
our findings may provide a new strategy for improving after the 4-day CNP treatment (Fig.  6B). The elevated
current superovulation technology in human infertility estrogen levels were characteristic of preantral follicle
treatment or farm animal reproductive management in growth in vitro, and estrogen supplementation benefits
the future, if no adverse effects of CNP are found. the reorganization of cultured follicles (Gore-Langton
ODGFs are prominent paracrine regulators et al. 1990, Adriaens et al. 2004).
of preantral follicle development, modulating Antrum initiation, the transition from preantral to
transcription in preantral follicles (Hayashi et al. 1999, early antral follicle, is the determinant for normal
Celestino et al. 2011, Lima et al. 2012, Fenwick et al. folliculogenesis. In humans, developmental disorder of
2013). We examined whether ODGFs can modulate preantral follicles, including impaired antrum formation
Nppc and Npr2 expression in preantral follicles and and poor gonadotropin response, is a main cause of
GCs. Generally, GDF9, BMP15 and FGF8 alone or female infertility. In short, our study not only highlights
combined enhanced Nppc and Npr2 expression in the role of CNP in promoting preantral follicle growth by
preantral follicles and GCs (Fig. 5). Given the essential enhancing follicular GC viability, but may also provide
role of CYP17A1 expression, GDF9 promotes rat a strategy for improving poor gonadotropin response in
preantral follicle growth by upregulating follicular human medicine.
androgen biosynthesis (Orisaka et al. 2009). Recently,
it has been found that BMP15 maintains in vitro culture
caprine preantral follicle integrity and promotes their Declaration of interest
growth (Celestino et  al. 2011, Lima et  al. 2012). The The authors declare that there is no conflict of interest that
expression of FGF8 and its cognate receptors were could be perceived as prejudicing the impartiality of the
discovered in fetal bovine preantral follicles (Buratini research reported.
et  al. 2005). However, their regulation of Nppc
and Npr2 in mouse preantral follicle has not been
reported. Our results show that ODGFs are involved Funding
in regulating Nppc and Npr2 expression in mouse This work was supported by grants from the National Key R&D
preantral follicle and imply that CNP may be a critical Program (2017YFD0501901 and 2017YFD0501905), the
mediator of the regulatory role of oocytes in preantral National Natural Science Foundation of China (No. 3167246
follicle growth and survival, as oocytes secrete GDF9, and 31472092) and the Earmarked Fund for the Innovative
BMP15 and FGF8. Teams of Beijing Swine Industrialization Research Program.
We investigated the effect of CNP on gene expression
in mouse preantral follicle and found that it affects
the expression of key ovarian genes involved in cell Author contribution statement
growth and key steroidogenic enzymes (Fig.  6). CNP G X, J T and L A were responsible for experimental design.
enhanced Wnt2b, Wnt5a and Cyp11a1 expression G X, L A, S A F and W W were responsible for writing the
and reduced Cyp1a1 and Cdkn1a mRNA levels in paper and for data analysis. G X, W W and S A F were
cultured preantral follicles (Fig.  6). Using conditional responsible for RNA isolation, qRT-PCR and western blotting.
gene targeting, Abedini et  al. found that GC-specific M Y was responsible for cell viability analysis. F Y and J H
inactivation of Wnt5a in mouse preantral follicle were responsible for sample collection. J T and L A recruited
results in female subfertility associated with increased the subjects and supervised the experiments and revised
follicular atresia and decreased ovulation rates the manuscript.

https://rep.bioscientifica.com Reproduction (2019) 157 445–455

Downloaded from Bioscientifica.com at 11/05/2020 04:09:28PM


via free access
454 G Xi, W Wang, S A Fazlani and others

References Geister  KA, Brinkmeier  ML, Hsieh  M, Faust  SM, Karolyi  IJ, Perosky  JE,
Kozloff  KM, Conti  M & Camper  SA 2013 A novel loss-of-function
Abedini  A, Zamberlam  G, Lapointe  E, Tourigny  C, Boyer  A, Paquet  M, mutation in Npr2 clarifies primary role in female reproduction and
Hayashi K, Honda H, Kikuchi A, Price C et al. 2016 WNT5a is required reveals a potential therapy for acromesomelic dysplasia, Maroteaux
for normal ovarian follicle development and antagonizes gonadotropin type. Human Molecular Genetics 22 345–357. (https://doi.org/10.1093/
responsiveness in granulosa cells by suppressing canonical WNT hmg/dds432)
signaling. FASEB Journal 30 1534–1547. (https://doi.org/10.1096/fj.15- Gore-Langton  RE & Daniel  SA 1990 Follicle-stimulating hormone and
280313) estradiol regulate antrum-like reorganization of granulosa cells in rat
Adriaens  I, Cortvrindt  R & Smitz  J 2004 Differential FSH exposure in preantral follicle cultures. Biology of Reproduction 43 65–72. (https://
preantral follicle culture has marked effects on folliculogenesis and doi.org/10.1095/biolreprod43.1.65)
oocyte developmental competence. Human Reproduction 19 398–408. Halpin  DM, Charlton  HM & Faddy  MJ 1986 Effects of gonadotrophin
(https://doi.org/10.1093/humrep/deh074) deficiency on follicular development in hypogonadal (hpg) mice. Journal
Balaban  B & Urman  B 2006 Effect of oocyte morphology on embryo of Reproduction and Fertility 78 119–125. (https://doi.org/10.1530/
development and implantation. Reproductive Biomedicine Online 12 jrf.0.0780119)
608–615. (https://doi.org/10.1016/S1472-6483(10)61187-X) Hayashi  M, Mcgee  EA, Min  G, Klein  C, Rose  UM, van Duin  M &
Boyer A, Goff AK & Boerboom D 2010 WNT signaling in ovarian follicle Hsueh  AJ 1999 Recombinant growth differentiation factor-9 (GDF-
biology and tumorigenesis. Trends in Endocrinology and Metabolism 21 9) enhances growth and differentiation of cultured early ovarian
25–32. (https://doi.org/10.1016/j.tem.2009.08.005) follicles. Endocrinology 140 1236–1244. (https://doi.org/10.1210/
Buratini JJ, Glapinski VF, Giometti IC, Teixeira AB, Costa IB, Avellar MC, endo.140.3.6548)
Barros  CM & Price  CA 2005 Expression of fibroblast growth factor-8 Hiradate  Y, Hoshino  Y, Tanemura  K & Sato  E 2014 C-type natriuretic
and its cognate receptors, fibroblast growth factor receptor (FGFR)-3c peptide inhibits porcine oocyte meiotic resumption. Zygote 22
and-4, in fetal bovine preantral follicles. Molecular Reproduction and 372–377. (https://doi.org/10.1017/S0967199412000615)
Development 70 255–261. (https://doi.org/10.1002/mrd.20205) Huang  DH, Zhang  SW, Zhao  H & Zhang  L 2011 The role of C-type
Cattanach  BM, Iddon  CA, Charlton  HM, Chiappa  SA & Fink  G natriuretic peptide in rat testes during spermatogenesis. Asian Journal of
1977 Gonadotrophin-releasing hormone deficiency in a mutant Andrology 13 275–280. (https://doi.org/10.1038/aja.2010.147)
mouse with hypogonadism. Nature 269 338–340. (https://doi. Hunter  MG, Robinson  RS, Mann  GE & Webb  R 2004 Endocrine and
org/10.1038/269338a0) paracrine control of follicular development and ovulation rate in farm
Celestino JJ, Lima-Verde IB, Bruno JB, Matos MH, Chaves RN, Saraiva MV, species. Animal Reproduction Science 82–83 461–477. (https://doi.
Silva  CM, Faustino  LR, Rossetto  R, Lopes  CA et  al. 2011 Steady-state org/10.1016/j.anireprosci.2004.05.013)
level of bone morphogenetic protein-15 in goat ovaries and its influence Jiang L, Huang J, Li L, Chen Y, Chen X, Zhao X & Yang D 2015 MicroRNA-93
on in vitro development and survival of preantral follicles. Molecular promotes ovarian granulosa cells proliferation through targeting CDKN1A
and Cellular Endocrinology 338 1–9. (https://doi.org/10.1016/j. in polycystic ovarian syndrome. Journal of Clinical Endocrinology and
mce.2011.02.007) Metabolism 100 E729–E738. (https://doi.org/10.1210/jc.2014-3827)
Cheng Y, Kawamura K, Takae S, Deguchi M, Yang Q, Kuo C & Hsueh AJ 2013 Kake T, Kitamura H, Adachi Y, Yoshioka T, Watanabe T, Matsushita H, Fujii T,
Oocyte-derived R-spondin2 promotes ovarian follicle development. Kondo E, Tachibe T, Kawase Y et al. 2009 Chronically elevated plasma
FASEB Journal 27 2175–2184. (https://doi.org/10.1096/fj.12-223412) C-type natriuretic peptide level stimulates skeletal growth in transgenic
Chinkers M, Garbers DL, Chang MS, Lowe DG, Chin HM, Goeddel DV mice. American Journal of Physiology. Endocrinology and Metabolism
& Schulz  S 1989 A membrane form of guanylate cyclase is an 297 E1339–E1348. (https://doi.org/10.1152/ajpendo.00272.2009)
atrial natriuretic peptide receptor. Nature 338 78–83. (https://doi. Kawamura  K, Cheng  Y, Kawamura  N, Takae  S, Okada  A, Kawagoe  Y,
org/10.1038/338078a0) Mulders  S, Terada  Y & Hsueh  AJ 2011 Pre-ovulatory LH/hCG surge
Chusho  H, Tamura  N, Ogawa  Y, Yasoda  A, Suda  M, Miyazawa  T, decreases C-type natriuretic peptide secretion by ovarian granulosa
Nakamura  K, Nakao  K, Kurihara  T, Komatsu  Y et  al. 2001 Dwarfism cells to promote meiotic resumption of pre-ovulatory oocytes. Human
and early death in mice lacking C-type natriuretic peptide. PNAS 98 Reproduction 26 3094–3101. (https://doi.org/10.1093/humrep/der282)
4016–4021. (https://doi.org/10.1073/pnas.071389098) Kobayashi  N, Orisaka  M, Cao  M, Kotsuji  F, Leader  A, Sakuragi  N &
Cortvrindt  R, Smitz  J & Van Steirteghem  AC 1996 In-vitro maturation, Tsang  BK 2009 Growth differentiation factor-9 mediates follicle-
fertilization and embryo development of immature oocytes from stimulating hormone-thyroid hormone interaction in the regulation of
early preantral follicles from prepuberal mice in a simplified culture rat preantral follicular development. Endocrinology 150 5566–5574.
system. Human Reproduction 11 2656–2666. (https://doi.org/10.1093/ (https://doi.org/10.1210/en.2009-0262)
oxfordjournals.humrep.a019188) Latham  KE, Wigglesworth  K, Mcmenamin  M & Eppig  JJ 2004 Stage-
Cortvrindt  R, Hu  Y & Smitz  J 1998 Recombinant luteinizing hormone dependent effects of oocytes and growth differentiation factor 9
as a survival and differentiation factor increases oocyte maturation on mouse granulosa cell development: advance programming and
in recombinant follicle stimulating hormone-supplemented mouse subsequent control of the transition from preantral secondary follicles
preantral follicle culture. Human Reproduction 13 1292–1302. (https:// to early antral tertiary follicles. Biology of Reproduction 70 1253–1262.
doi.org/10.1093/humrep/13.5.1292) (https://doi.org/10.1095/biolreprod.103.023937)
Craig J, Orisaka M, Wang H, Orisaka S, Thompson W, Zhu C, Kotsuji F & Lee KB, Zhang M, Sugiura K, Wigglesworth K, Uliasz T, Jaffe LA & Eppig JJ
Tsang BK 2007 Gonadotropin and intra-ovarian signals regulating follicle 2013 Hormonal coordination of natriuretic peptide type C and natriuretic
development and atresia: the delicate balance between life and death. peptide receptor 3 expression in mouse granulosa cells. Biology of
Frontiers in Bioscience 12 3628–3639. (https://doi.org/10.2741/2339) Reproduction 88 42. (https://doi.org/10.1095/biolreprod.112.104810)
Dutto  I, Tillhon  M, Cazzalini  O, Stivala  LA & Prosperi  E 2015 Biology Lima  IM, Brito  IR, Rossetto  R, Duarte  AB, Rodrigues  GQ, Saraiva  MV,
of the cell cycle inhibitor p21(CDKN1A): molecular mechanisms and Costa  JJ, Donato  MA, Peixoto  CA, Silva  JR et  al. 2012 BMPRIB and
relevance in chemical toxicology. Archives of Toxicology 89 155–178. BMPRII mRNA expression levels in goat ovarian follicles and the in vitro
(https://doi.org/10.1007/s00204-014-1430-4) effects of BMP-15 on preantral follicle development. Cell and Tissue
Eppig  JJ 2001 Oocyte control of ovarian follicular development and Research 348 225–238. (https://doi.org/10.1007/s00441-012-1361-4)
function in mammals. Reproduction 122 829–838. (https://doi. Livak KJ & Schmittgen TD 2001 Analysis of relative gene expression data
org/10.1530/rep.0.1220829) using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method.
Eppig  JJ, Wigglesworth  K & Pendola  FL 2002 The mammalian oocyte Methods 25 402–408. (https://doi.org/10.1006/meth.2001.1262)
orchestrates the rate of ovarian follicular development. PNAS 99 Lumsden  NG, Khambata  RS & Hobbs  AJ 2010 C-type natriuretic
2890–2894. (https://doi.org/10.1073/pnas.052658699) peptide (CNP): cardiovascular roles and potential as a therapeutic
Fenwick MA, Mora JM, Mansour YT, Baithun C, Franks S & Hardy K 2013 target. Current Pharmaceutical Design 16 4080–4088. (https://doi.
Investigations of TGF-beta signaling in preantral follicles of female org/10.2174/138161210794519237)
mice reveal differential roles for bone morphogenetic protein 15. Machado MF, Caixeta ES, Sudiman J, Gilchrist RB, Thompson JG, Lima PF,
Endocrinology 154 3423–3436. (https://doi.org/10.1210/en.2012-2251) Price  CA & Buratini  J 2015 Fibroblast growth factor 17 and bone

Reproduction (2019) 157 445–455 https://rep.bioscientifica.com

Downloaded from Bioscientifica.com at 11/05/2020 04:09:28PM


via free access
CNP stimulates ovarian GC viability 455

morphogenetic protein 15 enhance cumulus expansion and improve Sato  Y, Cheng  Y, Kawamura  K, Takae  S & Hsueh  AJ 2012 C-type
quality of in vitro-produced embryos in cattle. Theriogenology 84 natriuretic peptide stimulates ovarian follicle development. Molecular
390–398. (https://doi.org/10.1016/j.theriogenology.2015.03.031) Endocrinology 26 1158–1166. (https://doi.org/10.1210/me.2012-1027)
Mcgee E, Spears N, Minami S, Hsu SY, Chun SY, Billig H & Hsueh AJ 1997 Su YQ, Denegre JM, Wigglesworth K, Pendola FL, O’Brien MJ & Eppig JJ
Preantral ovarian follicles in serum-free culture: suppression of apoptosis 2003 Oocyte-dependent activation of mitogen-activated protein kinase
after activation of the cyclic guanosine 3',5'-monophosphate pathway (ERK1/2) in cumulus cells is required for the maturation of the mouse
and stimulation of growth and differentiation by follicle-stimulating oocyte-cumulus cell complex. Developmental Biology 263 126–138.
hormone. Endocrinology 138 2417–2424. (https://doi.org/10.1210/ (https://doi.org/10.1016/S0012-1606(03)00437-8)
endo.138.6.5164) Xi  G, An  L, Jia  Z, Tan  K, Zhang  J, Wang  Z, Zhang  C, Miao  K, Wu  Z &
Mcnatty KP, Reader K, Smith P, Heath DA & Juengel JL 2007 Control of Tian J 2018 Natriuretic peptide receptor 2 (NPR2) localized in bovine
ovarian follicular development to the gonadotrophin-dependent phase: oocyte underlies a unique mechanism for C-type natriuretic peptide
a 2006 perspective. Society of Reproduction and Fertility Supplement 64 (CNP)-induced meiotic arrest. Theriogenology 106 198–209. (https://
55–68. (https://doi.org/10.5661/RDR-VI-55) doi.org/10.1016/j.theriogenology.2017.09.003)
Miyoshi T, Otsuka F, Nakamura E, Inagaki K, Ogura-Ochi K, Tsukamoto N, Xu B, Gao L, Cui Y, Gao L, Dai X, Li M, Zhang Y, Ma X, Diao F & Liu J
Takeda M & Makino H 2012 Regulatory role of kit ligand-c-kit interaction 2013 SET protein up-regulated testosterone production in the cultured
and oocyte factors in steroidogenesis by rat granulosa cells. Molecular preantral follicles. Reproductive Biology and Endocrinology 11 9.
and Cellular Endocrinology 358 18–26. (https://doi.org/10.1016/j. (https://doi.org/10.1186/1477-7827-11-9)
mce.2012.02.011) Yagci R, Totan Y, Ozluk E, Ozyurt H, Akbay G & Eksioglu M 2008 Serum
Nishikimi T, Kuwahara K & Nakao K 2011 Current biochemistry, molecular levels of natriuretic peptides in patients with Behcet’s disease. Clinical
biology, and clinical relevance of natriuretic peptides. Journal of Rheumatology 27 1153–1158. (https://doi.org/10.1007/s10067-008-
Cardiology 57 131–140. (https://doi.org/10.1016/j.jjcc.2011.01.002) 0886-0)
Orisaka  M, Orisaka  S, Jiang  JY, Craig  J, Wang  Y, Kotsuji  F & Tsang  BK Yan  W, Zhou  S, Shen  W, Cheng  J, Yuan  S, Ye  S, Jin  Y, Luo  A & Wang  S
2006 Growth differentiation factor 9 is antiapoptotic during 2019 Suppression of SEMA6C promotes preantral follicles atresia with
follicular development from preantral to early antral stage. Molecular decreased cell junctions in mice ovaries. Journal of Cellular Physiology
Endocrinology 20 2456–2468. (https://doi.org/10.1210/me.2005-0357) 234 4934–4943. (https://doi.org/10.1002/jcp.27294)
Orisaka  M, Jiang  JY, Orisaka  S, Kotsuji  F & Tsang  BK 2009 Growth Zhang M, Su YQ, Sugiura K, Xia G & Eppig JJ 2010 Granulosa cell ligand
differentiation factor 9 promotes rat preantral follicle growth by NPPC and its receptor NPR2 maintain meiotic arrest in mouse oocytes.
up-regulating follicular androgen biosynthesis. Endocrinology 150 Science 330 366–369. (https://doi.org/10.1126/science.1193573)
2740–2748. (https://doi.org/10.1210/en.2008-1536) Zhang CP, Yang JL, Zhang J, Li L, Huang L, Ji SY, Hu ZY, Gao F & Liu YX
Orisaka  M, Hattori  K, Fukuda  S, Mizutani  T, Miyamoto  K, Sato  T, 2011 Notch signaling is involved in ovarian follicle development by
Tsang BK, Kotsuji F & Yoshida Y 2013 Dysregulation of ovarian follicular regulating granulosa cell proliferation. Endocrinology 152 2437–2447.
development in female rat: LH decreases FSH sensitivity during (https://doi.org/10.1210/en.2010-1182)
preantral-early antral transition. Endocrinology 154 2870–2880. (https:// Zhang Y, Wang H, Liu W, Yang Y, Wang X, Zhang Z, Guo Q, Wang C & Xia G
doi.org/10.1210/en.2012-2173) 2017 Natriuretic peptides improve the developmental competence of in
Potter LR, Abbey-Hosch S & Dickey DM 2006 Natriuretic peptides, their vitro cultured porcine oocytes. Reproductive Biology and Endocrinology
receptors, and cyclic guanosine monophosphate-dependent signaling 15 41. (https://doi.org/10.1186/s12958-017-0258-1)
functions. Endocrine Reviews 27 47–72. (https://doi.org/10.1210/ Zhong Y, Lin J, Liu X, Hou J, Zhang Y & Zhao X 2015 C-Type natriuretic
er.2005-0014) peptide maintains domestic cat oocytes in meiotic arrest. Reproduction,
Ren L, Wang Z, An L, Zhang Z, Tan K, Miao K, Tao L, Cheng L, Zhang Z, Fertility, and Development 28 1553–1559. (https://doi.org/10.1071/
Yang M et al. 2015 Dynamic comparisons of high-resolution expression RD14425)
profiles highlighting mitochondria-related genes between in vivo
and in vitro fertilized early mouse embryos. Human Reproduction 30
2892–2911. (https://doi.org/10.1093/humrep/dev228)
Sadr  SZ, Ebrahimi  B, Shahhoseini  M, Fatehi  R & Favaedi  R 2015 Received 4 September 2018
Mouse preantral follicle development in two-dimensional and three-
First decision 10 October 2018
dimensional culture systems after ovarian tissue vitrification. European
Journal of Obstetrics, Gynecology, and Reproductive Biology 194 Revised manuscript received 11 February 2019
206–211. (https://doi.org/10.1016/j.ejogrb.2015.09.028) Accepted 25 February 2019

https://rep.bioscientifica.com Reproduction (2019) 157 445–455

Downloaded from Bioscientifica.com at 11/05/2020 04:09:28PM


via free access

You might also like