Skip to main content
Erschienen in: Journal of Ovarian Research 1/2017

Open Access 01.12.2017 | Research

Macrophage colony-stimulating factor (M-CSF) is an intermediate in the process of luteinizing hormone-induced decrease in natriuretic peptide receptor 2 (NPR2) and resumption of oocyte meiosis

verfasst von: Wenchao Sun, Chang Liu, Ying Feng, Guangchao Zhuo, Wenjing Zhou, Xiaoyang Fei, Zhifen Zhang

Erschienen in: Journal of Ovarian Research | Ausgabe 1/2017

Abstract

Background

Luteinizing hormone (LH) regulation of the ligand, natriuretic peptide precursor type C, and its receptor, natriuretic peptide receptor 2 (NPR2), is critical for oocyte maturation; however, the mechanism is not fully understood. Macrophage colony-stimulating factor (M-CSF) has recently been shown to be involved in oocyte maturation and ovulation. In the present study we determined whether or not M-CSF plays a role in the intermediate signal that mediates LH regulation of NPR2 in resumption of oocyte meiosis.

Methods

Immature female C57BL/6 mice were injected i.p. with 5 IU of equine chorionic gonadotropin (eCG) to stimulate follicle development. After 44–48 h, the eCG-stimulated mice were injected i.p. with an ovulatory dose of 5 IU of human chorionic gonadotropin (hCG). The ovaries were excised at selected times. Pre-ovulatory follicles (POFs) and cumulus-oocyte complexes were cultured in different media. Immunohistochemical and quantitative real-time PCR analyses were used to assess the expression of M-CSF, M-CSF receptor (M-CSF-R), and NPR2. The presence of germinal vesicle breakdown (GVBD) was examined under a stereomicroscope to morphologically evaluate resumption of oocyte meiosis.

Results

NPR2 was mainly expressed in cumulus cells of pre-ovulatory follicles, while M-CSF and M-CSF-R were expressed in both mural granulosa and cumulus cells. The levels of M-CSF/M-CSF-R and NPR2 decreased within 4 h after treatment of hCG. M-CSF not only reduced the expression of NPR2 mRNA via its receptor (M-CSF-R), but also increased the proportion of GVBD in oocytes.

Conclusion

M-CSF serves as an intermediate signal, thus inducing a vital decrease in the NPR2 levels in cumulus cells, and regulates the process of LH-induced resumption of meiosis.
Abkürzungen
cAMP
cyclic adenosine 3′,5′-monophosphate
cGMP
cyclic guanosine 3′,5′-monophosphate
COCs
Cumulus-oocyte complexes
E2
Estradiol
eCG
Equine chorionic gonadotropin
EGFR
Epidermal growth factor receptor
FBS
Fetal bovine serum
GCs
Granulosa cells
GVBD
Germinal vesicle breakdown
hCG
Human chorionic gonadotropin
IOD
Integrated optical density
LH
Luteinizing hormone
M-CSF
Macrophage colony-stimulating factor
M-CSF-R
M-CSF receptor
MEM
Minimum essential media
NPPC
Natriuretic peptide precursor type C
NPR2
Natriuretic peptide receptor 2
PDE
Phosphodiesterase
POFs
Pre-ovulatory follicles

Background

In female mammals, oocytes grow and undergo meiosis over a prolonged period of time [1, 2]. Once the growing follicles reach the early antral stage, oocytes acquire meiotic competence [3]; however, oocytes are arrested at the diplotene stage of the first meiotic prophase because signals from the surrounding granulosa cells (GCs) prevent the machinery required for resumption of meiosis [3, 4]. Throughout prophase arrest, the oocyte is situated in a follicle where the oocyte is encircled by GCs (Fig. 1). The essence of signals maintaining meiotic arrest has been demonstrated as a complicated interaction between cyclic adenosine 3′,5′-monophosphate (cAMP) and cyclic guanosine 3′,5′-monophosphate (cGMP) signaling [510]. Cyclic AMP is generated by the oocyte via the activation of Gs G-protein by the G-protein-coupled receptor and adenylyl cyclases. Cyclic GMP, synthesized in surrounding GCs, diffuses into the oocyte through the network of gap junction communications, and inhibits oocyte cAMP-phosphodiesterase (PDE) 3A activity and hydrolysis of cAMP to maintain meiotic arrest [27, 9, 11, 12]. Subsequent studies have indicated that generation of cGMP is stimulated by a paracrine loop, which includes natriuretic peptide receptor 2 (NPR2) and the ligand, natriuretic peptide precursor type C (NPPC) [13]. NPPC, produced by mural GCs, activates NPR2, which is produced mainly by cumulus cells surrounding the oocyte, increases cAMP and cGMP levels in the oocyte, and prevents spontaneous (gonadotropin-independent) resumption of oocyte meiosis [13, 14]. In mice deficient in the ligand, NPPC, or its cognate receptor, NPR2, oocytes precociously re-enter the meiotic cell cycle as soon as the oocytes reach the early antral follicle stage [13, 15].
Oocytes resume meiosis when the luteinizing hormone (LH) surge causes dramatic changes in pre-ovulatory follicles (POFs) [16, 17]. It has been shown that LH causes a spectacular decrease in NPPC in the follicles of a wide variety of mammalian species, including mice, rats, pigs, and humans [4, 5, 13, 18, 19]. Decreased NPPC in turn reduces the amount of NPR2 and cGMP, and meiosis resumes in oocytes [4, 5]. Recently, when the kinetic curve of cytokines was further studied, NPPC was not shown to be decreased until 2 h after the LH surge, whereas the decrease in cGMP was first detected at 15–20 min [14]. NPR2 also undergoes a rapid decrease in activity within 10 min after LH exposure, when the NPPC concentration is constant [20, 21]. This phenomenon (that the receptor is motivated before ligand activation) gave rise to the hypothesis that multiple pathways mediate LH regulation of NPR2 and downstream cGMP signaling in the ovarian follicle [21]. These multiple pathways include the phosphoprotein phosphatase signaling pathway [20], which has recently been associated with ovulation process. Multiple pathways are known to include the epidermal growth factor receptor (EGFR) signaling pathway [2126], but recent findings have shown new relationships. The exact number of these multiple pathways is unknown.
Macrophage colony-stimulating factor (M-CSF), a hemopoietic growth factor with a classic function of controlling the proliferation and differentiation of macrophages, has recently been shown to be involved in oocyte maturation and ovulation [2730]. We have previously reported that M-CSF is implicated in follicular GC function [27], and M-CSF can modulate the generation of NPPC, which may regulate ovulation triggered by LH [31]. In the present study we determined whether or not M-CSF is included in the aforementioned “multiple pathways” that mediate LH regulation of NPR2 in ovarian follicles.

Methods

Animals and hormone treatments

Immature (22–25 days old) female C57BL/6 mice (Zhejiang Academy of Medical Sciences, Hangzhou, China) were injected i.p. with 5 IU of equine chorionic gonadotropin (eCG) to stimulate follicle development. After 44–48 h, the eCG-stimulated mice were injected i.p. with an ovulatory dose of human chorionic gonadotropin (hCG; 5 IU). The ovaries were excised at selected times after injection and processed for immunohistochemical analysis and quantitative real-time PCR. For cell culture of POFs and cumulus-oocyte complexes (COCs), the eCG-stimulated mice were euthanized and the ovaries were excised without hCG injection. All chemicals were purchased from Sigma-Aldrich (St. Louis, MO, USA) unless otherwise stated. All animal procedures were approved by the guidelines of the Nanjing Medical University Administrative Panel on Laboratory Animal Care.

Culture of POFs

The POFs were dissected stereomicroscopically from the ovaries of eCG-stimulated mice, as previously described [32, 33]. The POFs were placed in minimum essential media (MEM)-α supplemented with 100 mg/ml of fetal bovine serum (FBS), 100 U/ml of penicillin G, and 100 μg/ml of streptomycin sulfate. After equilibration, follicles (10–15 per group) were cultured at 37 °C in an atmosphere of 5% O2, 5% CO2, and 90% N2 for the indicated time in the presence or absence of hCG (5 IU/ml). At the end of the culture period, follicles were collected for quantitative RT-PCR analysis to measure hCG regulation of NPR2 transcript levels.

Culture of COCs

COCs were obtained by puncturing the POFs in the ovaries from eCG-stimulated mice. After isolation, COCs were washed in the medium and cultured for 2 h. The culture medium was MEM-α supplemented with 100 mg/ml of FBS, 100 U/ml of penicillin G, and 100 μg/ml of streptomycin sulfate with or without 30 nM NPPC. At least 10 COCs per treatment group were cultured. Cultures were maintained under a controlled atmosphere of 5% O2, 5% CO2, and 90% N2 at 37 °C. After culture, COCs were collected for quantitative RT-PCR to calculate the NPR2 transcript levels. The presence of germinal vesicle breakdown (GVBD) was examined under a stereomicroscope.

RNA isolation, reverse transcription, and quantitative real-time PCR

Mouse ovaries, cultured follicles, and COCs were collected in 350 μl of RNeasy lysis buffer. The tissues and cells were stored at −80 °C until analysis for mRNA expression. Total RNA was isolated from frozen samples using the RNeasy micro-RNA isolation kit (Qiagen, Valencia, CA, USA), as recommended by the manufacturer’s instructions. Reverse transcription and real-time PCR was then carried out to quantify the steady-state mRNA levels of NPPC, NPR2, M-CSF, and M-CSF-R using an ABI 7500 real-time PCR instrument (Applied Biosystems, Foster City, CA, USA). The housekeeping gene, Rpl19, was considered the internal control. The primers for real-time PCR of NPPC, NPR2, M-CSF, M-CSF-R, and Rpl19 are listed in Table 1. The levels of NPPC, NPR2, M-CSF, and M-CSF-R mRNA were first normalized to the level of Rpl19 expression, then demonstrated relative to a control group in which the level of expression was set at 1. Each experiment was repeated independently at least three times.
Table 1
Primer sequences, forward (F) or reverse (R), used for quantitative RT-PCR
Gene
F or R
Primer sequence
NPPC
F
GGGAGCCAATCTCAAGGGAG
 
R
GTTGCCGCCTTTGTATTTGC
NPR2
F
GCATTGTCACCGAGTATTGTCC
 
R
CAGACCGTAATCTGTTATTTTGAGC
M-CSF
F
TGATTGGGAATGGACACCTG
 
R
AAAGGCAATCTGGCATGAAGT
M-CSF-R
F
GGTGGCTGTGAAGATGCTAAAG
 
R
AGGCTCCCAAGAGGTTGACTAT
Rpl19
F
CCGCTGCGGGAAAAAGAAG
 
R
CAGCCCATCCTTGATCAGCTT

Immunohistochemistry

The excised ovaries were fixed in 10% formalin. After dehydration, the fixed ovaries were embedded in paraplast, then sectioned at 5-μm intervals onto Superfrost Plus microscope slides. For immunohistochemical staining, sections of ovaries were deparaffinized and rehydrated, treated with 3% H2O2 for 20 min to inactivate intrinsic peroxidase activity, and incubated with ethylene dinitrilo tetraacetic acid buffer for 10 min for antigen retrieval. Washes were carried out with automation phosphate buffer. Sections were incubated for 2 h at 4 °C with rabbit anti-mouse antibody diluted 1:200 in buffer containing 5% bovine serum albumin. Sections were next incubated with horseradish peroxidase-labeled goat anti-rabbit antibody for 50 min at room temperature. Staining was achieved using diaminobenzidine chromogen. The staining reactions were stopped with distilled water, and sections were dehydrated and mounted with neutral balsam.

Image analysis of densitometry

Slides were examined under a microscope with a 200 × objective. The obtained images were captured and examined by Image Pro Plus 6.0 software (Media Cybernetics, Inc., Washington, USA). The integrated optical density (IOD) was calibrated, and the area of interest was set. The mean optical density was defined as the IOD divided by the total area examined.

Statistical analysis

Statistical analyses were carried out using SPSS software (version 16.0: SPSS, Inc., Chicago, IL, USA). Data are presented as the mean ± SEM. Differences between experimental and control groups were analyzed by ANOVA test. Statistical significance was set at P value of less than 0.05.

Results

Localization of M-CSF, M-CSF-R, and NPR2 in POFs

To investigate if M-CSF, M-CSF-R, and NPR2 signaling is involved in regulation of oocyte meiosis in POFs. We analyzed M-CSF, M-CSF-R, and NPR2 localization in ovarian sections from eCG-stimulated mice. Immunohistochemistry analysis revealed that M-CSF and M-CSF-R are expressed in both mural GCs and cumulus cells (Fig. 2a-d), while the expression of NPR2 in POFs was mainly detected in cumulus cells and was also observed in peri-antral mural GCs (GCs located on the lining of antral spaces, [Fig. 2e and f]).

Changes in expression of M-CSF, M-CSF-R, and NPR2 in the ovaries of mice injected with gonadotropin

We further elucidated the importance of M-CSF, M-CSF-R, and NPR2 signaling in LH-induced resumption of oocyte meiosis. Using immunohistochemical techniques, we determined the changes in expression of M-CSF, M-CSF-R, and NPR2 in the ovaries of eCG-stimulated mice at 0 (48 h after eCG treatment), 0.5, 1, 2, and 4 h after injection with hCG (Fig. 3a). The mean optical density was calculated and showed that M-CSF and M-CSF-R expression was gradually decreased within 4 h after hCG treatment, which was administered 48 h after eCG injection (Fig. 3b). NPR2 expression peaked at 1 h, but an obvious reduction in expression was detected at 4 h (Fig. 3b). Expression of M-CSF, M-CSF-R, and NPR2 mRNA was also detected (Fig. 3c) and was consistent with the results of immunohistochemistry and densitometry analysis.

Kinetic curve of NPR2 mRNA levels controlled by hCG in vitro

To study the effect of hCG on the kinetic curve of NPR2 mRNA levels in POFs, we examined the expression of NPR2 mRNA after 0, 0.5, 1, 2, and 4 h of culture after hCG treatment. The hCG (5 IU/ml) significantly decreased NPR2 mRNA levels by one-half at 0.5 h, and nearly 90% at 4 h of culture. In the control group without hCG treatment, the levels of NPR2 mRNA in POFs were slightly increased after POFs were cultured for 1 h. Then, the NPR2 mRNA levels were only slightly decreased after POFs were cultured for 4 h (Fig. 4a). The image of cultured POFs is presented in Fig. 4b. The results are in agreement with the speculation that hCG regulates NPR2.

M-CSF/M-CSF-R signaling decreases the level of NPR2 mRNA in cumulus cells and contributes to resumption of meiosis in oocytes

The effect of M-CSF/M-CSF-R signaling on NPR2 mRNA expression and oocyte maturation was determined using cultured COCs isolated from POFs of eCG-stimulated mice. Isolated COCs spontaneously resume meiosis because mural GCs containing the inhibitory molecule, NPPC, is removed. Therefore, a culture system supplemented with 30 nM NPPC was adopted to maintain meiotic arrest in oocytes [23]. M-CSF (200 ng/ml) significantly decreased the level of NPR2 mRNA when COCs were cultured for 2 h (Fig. 5a). M-CSF stimulated GVBD in approximately 65% of oocytes at 2 h of culture (Fig. 5b and c). GVBD marks the onset of meiotic resumption and is the key event in oocyte maturation (Fig. 5d). Furthermore, the M-CSF-induced decrease in NPR2 and resumption of oocyte meiosis was partially inhibited by GW2580 (a selective M-CSF-R inhibitor purchased from Selleckchem, Houston, TX, USA; Fig. 5a and b), suggesting that M-CSF functioned via the activity of the M-CSF receptor.

Discussion

In the present study, we focused on the effect of M-CSF in the regulation of oocyte meiosis, and identified some crucial roles, including: (a) NPR2 was mainly expressed in cumulus cells of POFs, while M-CSF and M-CSF-R were expressed in both mural GCs and cumulus cells; (b) the levels of M-CSF/M-CSF-R and NPR2 decreased within 4 h after hCG treatment; and (c) M-CSF not only reduced the expression of NPR2 mRNA via its receptor (M-CSF-R), but also increased the proportion of GVBD of oocytes, which indicates that M-CSF is an intermediate signal, inducing a vital decrease in NPR2 levels in cumulus cells, and regulates the process of LH-induced resumption of meiosis.
M-CSF extensively participates in the processes of ovulation [27, 29]. Female mice lacking the coding region for the M-CSF gene (M-CSF deficient) have remarkably lower ovulation rates compared to the wild-type counterparts. Indeed, administration of M-CSF from birth to reinstate circulating M-CSF levels could reverse these defects [27]. In humans, high serum concentrations of M-CSF were related to successful oocyte retrieval during in-vitro fertilization and embryo transfer cycles [28]. LH surge triggers dramatic changes in cytokines during ovulation [16]. Although the changes in M-CSF and M-CSF-R 24 h after a LH surge are clear [29], the alteration in M-CSF/M-CSF-R within a short period of time (4 h) following the LH surge is unknown. Therefore, we studied the changes in M-CSF/M-CSF-R expression in the ovaries of mice injected with gonadotropin. The data from our study showed that M-CSF/M-CSF-R, expressed in both mural GCs and cumulus cells, was gradually decreased within 4 h after hCG treatment. We hypothesized that this change is related to estradiol (E2). Reportedly, the expression of M-CSF was enhanced by E2 in luteinized GCs in a dose-dependent manner in vitro [27]. E2 maintains cumulus cell expression of NPR2 and inhibits the resumption of meiosis in mouse oocytes in vitro [34], and the level decreased during ovulation. Thus, decreased E2 has a certain role to the resumption of oocyte meiosis and ovulation. Therefore, the decreased level of M-CSF after hCG treatment may be due to the decreased levels of E2. Although the hypothesis is theoretically feasible, it is still necessary to further investigate whether or not a decrease in M-CSF during ovulation is related to a decrease in the E2 level.
Recent studies have revealed that cGMP stimulated by NPR2 from cumulus cells diffuses into oocytes via gap junctions and controls cAMP concentration through inhibition of PDE3A activity, indicating that higher NPR2 levels in cumulus cells is responsible for oocyte meiotic arrest by maintaining high cAMP levels in oocytes [1, 47]. Our results showed that NPR2 is primarily expressed in cumulus cells surrounding oocytes, which is consistent with the literature. The results presented herein support the thought that control of NPR2, which is expressed in cumulus cells, is essential for maintaining meiotic arrest in pre-ovulatory oocytes [4, 13]. LH reduces the activity of NPR2 during ovulation, and promotes resumption of meiosis in oocytes [14]. In particular, our findings demonstrated that NPR2 is also expressed in peri-antral mural GCs (GCs situated on the antral side), which substantiates the viewpoint that some mGCs are activated by NPPC in an autocrine process to raise cGMP levels [24, 34]. NPR2 was down-regulated following a specific time curve after hCG injection. The kinetic curve of NPR2 after hCG treatment in our study was similar to that reported in the literature [23], but there were some differences. In our research, NPR2 expression peaked 1 h after hCG treatment, and an obvious reduction of approximately 85% in expression was detected at 4 h in vivo. The rise in NPR2 expression within 2 h after LH treatment was different from that reported in the literature, likely due to the long half-life (40 ~ 120 h) of eCG administered 48 h before hCG injection to stimulate follicle development [22, 24]. The eCG promoted the up-regulation of NPR2 during follicle growth [33]. To clarify the NPR2 expression change within 2 h after LH treatment, cultured POFs were used to examine the kinetic curve of NPR2 mRNA levels controlled by hCG in vitro. The data from our study were consistent with the literature [23]. We did not observe a significant increase in expression of NPR2 in hCG-containing media in vitro, possibly because in vitro culture eliminated the residual effect of eCG. To further declare the effect of M-CSF/M-CSF-R signaling on NPR2 mRNA expression and oocyte maturation, COCs isolated from POFs were cultured. The results showed that the M-CSF/M-CSF-R signaling reduced levels of NPR2 mRNA in cumulus cells and promoted resumption of oocyte meiosis. Then, we added GW2580 to restrict the effect of M-CSF signaling in vitro. The results showed that the effect of M-CSF at 4 h was partially abolished.

Conclusion

We conclude that M-CSF is an intermediate signal, inducing a vital decrease in NPR2 levels in cumulus cells, and regulates the process of LH-induced resumption of meiosis. Although further research is needed, our findings bring forth powerful evidence to interpret “multiple pathways” that mediate LH regulation of NPR2 in the process of resuming oocyte meiosis.

Acknowledgements

We acknowledge core support provided by the National Natural Science Foundation of China (grant no. 31470078).

Funding

The study was funded by core support provided by the National Natural Science Foundation of China (grant no. 31470078).

Availability of data and materials

All data generated or analyzed during this study are included in this published article. The datasets used during the current study are available from the corresponding author on reasonable request.

Ethics approval

All animal procedures were approved by the guidelines of the Nanjing Medical University Administrative Panel on Laboratory Animal Care. The study does not include any human samples.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Mehlmann LM. Stops and starts in mammalian oocytes: recent advances in understanding the regulation of meiotic arrest and oocyte maturation. Reproduction. 2005;130:791–9.CrossRefPubMed Mehlmann LM. Stops and starts in mammalian oocytes: recent advances in understanding the regulation of meiotic arrest and oocyte maturation. Reproduction. 2005;130:791–9.CrossRefPubMed
2.
Zurück zum Zitat Solc P, Schultz RM, Motlik J, Prophase I. Arrest and progression to metaphase I in mouse oocytes: comparison of resumption of meiosis and recovery from G2-arrest in somatic cells. Mol Hum Reprod. 2010;16:654–64.CrossRefPubMedPubMedCentral Solc P, Schultz RM, Motlik J, Prophase I. Arrest and progression to metaphase I in mouse oocytes: comparison of resumption of meiosis and recovery from G2-arrest in somatic cells. Mol Hum Reprod. 2010;16:654–64.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Holt JE, Lane SI, Jones KT. The control of meiotic maturation in mammalian oocytes. Curr Top Dev Biol. 2013;102:207–26.CrossRefPubMed Holt JE, Lane SI, Jones KT. The control of meiotic maturation in mammalian oocytes. Curr Top Dev Biol. 2013;102:207–26.CrossRefPubMed
4.
Zurück zum Zitat Zhang M, Xia G. Hormonal control of mammalian oocyte meiosis at diplotene stage. Cell Mol Life Sci. 2012;69:1279–88.CrossRefPubMed Zhang M, Xia G. Hormonal control of mammalian oocyte meiosis at diplotene stage. Cell Mol Life Sci. 2012;69:1279–88.CrossRefPubMed
5.
Zurück zum Zitat Zhang M, Ouyang H, Xia G. The signal pathway of gonadotrophins-induced mammalian oocyte meiotic resumption. Mol Hum Reprod. 2009;15:399–409.CrossRefPubMed Zhang M, Ouyang H, Xia G. The signal pathway of gonadotrophins-induced mammalian oocyte meiotic resumption. Mol Hum Reprod. 2009;15:399–409.CrossRefPubMed
6.
Zurück zum Zitat Norris RP, Ratzan WJ, Freudzon M, Mehlmann LM, Krall J, Movsesian MA, et al. Cyclic GMP from the surrounding somatic cells regulates cyclic AMP and meiosis in the mouse oocyte. Development. 2009;136:1869–78.CrossRefPubMedPubMedCentral Norris RP, Ratzan WJ, Freudzon M, Mehlmann LM, Krall J, Movsesian MA, et al. Cyclic GMP from the surrounding somatic cells regulates cyclic AMP and meiosis in the mouse oocyte. Development. 2009;136:1869–78.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Vaccari S, Weeks JL 2nd, Hsieh M, Menniti FS, Conti M. Cyclic GMP Signaling is involved in the luteinizing hormone-dependent meiotic maturation of mouse oocytes. Biol Reprod. 2009;81:595–604.CrossRefPubMedPubMedCentral Vaccari S, Weeks JL 2nd, Hsieh M, Menniti FS, Conti M. Cyclic GMP Signaling is involved in the luteinizing hormone-dependent meiotic maturation of mouse oocytes. Biol Reprod. 2009;81:595–604.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Tripathi A, Kumar KV, Chaube SK. Meiotic cell cycle arrest in mammalian oocytes. J Cell Physiol. 2010;223:592–600.PubMed Tripathi A, Kumar KV, Chaube SK. Meiotic cell cycle arrest in mammalian oocytes. J Cell Physiol. 2010;223:592–600.PubMed
9.
Zurück zum Zitat Vaccari S, Horner K, Mehlmann LM, Conti M. Generation of mouse oocytes defective in cAMP synthesis and degradation: endogenous cyclic AMP is essential for meiotic arrest. Dev Biol. 2008;316:124–34.CrossRefPubMedPubMedCentral Vaccari S, Horner K, Mehlmann LM, Conti M. Generation of mouse oocytes defective in cAMP synthesis and degradation: endogenous cyclic AMP is essential for meiotic arrest. Dev Biol. 2008;316:124–34.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat LaPolt PS, Leung K, Ishimaru R, Tafoya MA, You-hsin Chen J. Roles of cyclic GMP in modulating ovarian functions. Reprod BioMed Online. 2003;6:15–23.CrossRefPubMed LaPolt PS, Leung K, Ishimaru R, Tafoya MA, You-hsin Chen J. Roles of cyclic GMP in modulating ovarian functions. Reprod BioMed Online. 2003;6:15–23.CrossRefPubMed
11.
Zurück zum Zitat Sasseville M, Cote N, Guillemette C, Richard FJ. New insight into the role of phosphodiesterase 3A in porcine oocyte maturation. BMC Dev Biol. 2006;6:47.CrossRefPubMedPubMedCentral Sasseville M, Cote N, Guillemette C, Richard FJ. New insight into the role of phosphodiesterase 3A in porcine oocyte maturation. BMC Dev Biol. 2006;6:47.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Richard FJ, Tsafriri A, Conti M. Role of phosphodiesterase type 3A in rat oocyte maturation. Biol Reprod. 2001;65:1444–51.CrossRefPubMed Richard FJ, Tsafriri A, Conti M. Role of phosphodiesterase type 3A in rat oocyte maturation. Biol Reprod. 2001;65:1444–51.CrossRefPubMed
13.
Zurück zum Zitat Zhang M, YQ S, 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.CrossRefPubMedPubMedCentral Zhang M, YQ S, 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.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Robinson JW, Zhang M, Shuhaibar LC, Norris RP, Geerts A, Wunder F, et al. Luteinizing hormone reduces the activity of the NPR2 guanylyl cyclase in mouse ovarian follicles, contributing to the cyclic GMP decrease that promotes resumption of meiosis in oocytes. Dev Biol. 2012;366:308–16.CrossRefPubMedPubMedCentral Robinson JW, Zhang M, Shuhaibar LC, Norris RP, Geerts A, Wunder F, et al. Luteinizing hormone reduces the activity of the NPR2 guanylyl cyclase in mouse ovarian follicles, contributing to the cyclic GMP decrease that promotes resumption of meiosis in oocytes. Dev Biol. 2012;366:308–16.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Kiyosu C, Tsuji T, Yamada K, Kajita S, Kunieda T. NPPC/NPR2 signaling is essential for oocyte meiotic arrest and cumulus oophorus formation during follicular development in the mouse ovary. Reproduction. 2012;144:187–93.CrossRefPubMed Kiyosu C, Tsuji T, Yamada K, Kajita S, Kunieda T. NPPC/NPR2 signaling is essential for oocyte meiotic arrest and cumulus oophorus formation during follicular development in the mouse ovary. Reproduction. 2012;144:187–93.CrossRefPubMed
16.
Zurück zum Zitat Coticchio G, Dal Canto M, Mignini Renzini M, Guglielmo MC, Brambillasca F, Turchi D, et al. Oocyte maturation: gamete-somatic cells interactions, meiotic resumption, cytoskeletal dynamics and cytoplasmic reorganization. Hum Reprod Update. 2015;21:427–54.CrossRefPubMed Coticchio G, Dal Canto M, Mignini Renzini M, Guglielmo MC, Brambillasca F, Turchi D, et al. Oocyte maturation: gamete-somatic cells interactions, meiotic resumption, cytoskeletal dynamics and cytoplasmic reorganization. Hum Reprod Update. 2015;21:427–54.CrossRefPubMed
17.
Zurück zum Zitat Conti M, Hsieh M, Zamah AM, Oh JS. Novel signaling mechanisms in the ovary during oocyte maturation and ovulation. Mol Cell Endocrinol. 2012;356:65–73.CrossRefPubMed Conti M, Hsieh M, Zamah AM, Oh JS. Novel signaling mechanisms in the ovary during oocyte maturation and ovulation. Mol Cell Endocrinol. 2012;356:65–73.CrossRefPubMed
18.
Zurück zum Zitat Kawamura K, Cheng Y, Kawamura N, Takae S, Okada A, Kawagoe Y, et al. Pre-ovulatory LH/hCG surge decreases C-type natriuretic peptide secretion by ovarian granulosa cells to promote meiotic resumption of pre-ovulatory oocytes. Hum Reprod. 2011;26:3094–101.CrossRefPubMed Kawamura K, Cheng Y, Kawamura N, Takae S, Okada A, Kawagoe Y, et al. Pre-ovulatory LH/hCG surge decreases C-type natriuretic peptide secretion by ovarian granulosa cells to promote meiotic resumption of pre-ovulatory oocytes. Hum Reprod. 2011;26:3094–101.CrossRefPubMed
19.
Zurück zum Zitat Zhang W, Chen Q, Yang Y, Liu W, Zhang M, Xia G, et al. Epidermal growth factor-network signaling mediates luteinizing hormone regulation of BNP and CNP and their receptor NPR2 during porcine oocyte meiotic resumption. Mol Reprod Dev. 2014;81:1030–41.CrossRefPubMed Zhang W, Chen Q, Yang Y, Liu W, Zhang M, Xia G, et al. Epidermal growth factor-network signaling mediates luteinizing hormone regulation of BNP and CNP and their receptor NPR2 during porcine oocyte meiotic resumption. Mol Reprod Dev. 2014;81:1030–41.CrossRefPubMed
20.
Zurück zum Zitat Egbert JR, Shuhaibar LC, Edmund AB, Van Helden DA, Robinson JW, Uliasz TF, et al. Dephosphorylation and inactivation of NPR2 guanylyl cyclase in granulosa cells contributes to the LH-induced decrease in cGMP that causes resumption of meiosis in rat oocytes. Development. 2014;141:3594–604.CrossRefPubMedPubMedCentral Egbert JR, Shuhaibar LC, Edmund AB, Van Helden DA, Robinson JW, Uliasz TF, et al. Dephosphorylation and inactivation of NPR2 guanylyl cyclase in granulosa cells contributes to the LH-induced decrease in cGMP that causes resumption of meiosis in rat oocytes. Development. 2014;141:3594–604.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Liu X, Xie F, Zamah AM, Cao B, Conti M. Multiple pathways mediate luteinizing hormone regulation of cGMP signaling in the mouse ovarian follicle. Biol Reprod. 2014;91:9.PubMedPubMedCentral Liu X, Xie F, Zamah AM, Cao B, Conti M. Multiple pathways mediate luteinizing hormone regulation of cGMP signaling in the mouse ovarian follicle. Biol Reprod. 2014;91:9.PubMedPubMedCentral
22.
Zurück zum Zitat Panigone S, Hsieh M, Fu M, Persani L, Conti M. Luteinizing hormone signaling in preovulatory follicles involves early activation of the epidermal growth factor receptor pathway. Mol Endocrinol. 2008;22:924–36.CrossRefPubMedPubMedCentral Panigone S, Hsieh M, Fu M, Persani L, Conti M. Luteinizing hormone signaling in preovulatory follicles involves early activation of the epidermal growth factor receptor pathway. Mol Endocrinol. 2008;22:924–36.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Wang Y, Kong N, Li N, Hao X, Wei K, Xiang X, et al. Epidermal growth factor receptor signaling-dependent calcium elevation in cumulus cells is required for npr2 inhibition and meiotic resumption in mouse oocytes. Endocrinology. 2013;154:3401–9.CrossRefPubMed Wang Y, Kong N, Li N, Hao X, Wei K, Xiang X, et al. Epidermal growth factor receptor signaling-dependent calcium elevation in cumulus cells is required for npr2 inhibition and meiotic resumption in mouse oocytes. Endocrinology. 2013;154:3401–9.CrossRefPubMed
24.
Zurück zum Zitat 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 follicles. Mol Reprod Dev. 2012;79:795–802.CrossRefPubMed 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 follicles. Mol Reprod Dev. 2012;79:795–802.CrossRefPubMed
25.
Zurück zum Zitat Park JY, YQ S, Ariga M, Law E, Jin SL, Conti M. EGF-like growth factors as mediators of LH action in the ovulatory follicle. Science. 2004;303:682–4.CrossRefPubMed Park JY, YQ S, Ariga M, Law E, Jin SL, Conti M. EGF-like growth factors as mediators of LH action in the ovulatory follicle. Science. 2004;303:682–4.CrossRefPubMed
26.
Zurück zum Zitat Norris RP, Freudzon M, Nikolaev VO, Jaffe LA. Epidermal growth factor receptor kinase activity is required for gap junction closure and for part of the decrease in ovarian follicle cGMP in response to LH. Reproduction. 2010;140:655–62.CrossRefPubMedPubMedCentral Norris RP, Freudzon M, Nikolaev VO, Jaffe LA. Epidermal growth factor receptor kinase activity is required for gap junction closure and for part of the decrease in ovarian follicle cGMP in response to LH. Reproduction. 2010;140:655–62.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Zhang Z, Fang Q, Wang J. Involvement of macrophage colony-stimulating factor (M-CSF) in the function of follicular granulosa cells. Fertil Steril. 2008;90:749–54.CrossRefPubMed Zhang Z, Fang Q, Wang J. Involvement of macrophage colony-stimulating factor (M-CSF) in the function of follicular granulosa cells. Fertil Steril. 2008;90:749–54.CrossRefPubMed
28.
Zurück zum Zitat Takasaki A, Ohba T, Okamura Y, Honda R, Seki M, Tanaka N, et al. Clinical use of colony-stimulating factor-1 in ovulation induction for poor responders. Fertil Steril. 2008;90:2287–90.CrossRefPubMed Takasaki A, Ohba T, Okamura Y, Honda R, Seki M, Tanaka N, et al. Clinical use of colony-stimulating factor-1 in ovulation induction for poor responders. Fertil Steril. 2008;90:2287–90.CrossRefPubMed
29.
Zurück zum Zitat Salmassi A, Zhang Z, Schmutzler AG, Koch K, Buck S, Jonat W, et al. Expression of mRNA and protein of macrophage colony-stimulating factor and its receptor in human follicular luteinized granulosa cells. Fertil Steril. 2005;83:419–25.CrossRefPubMed Salmassi A, Zhang Z, Schmutzler AG, Koch K, Buck S, Jonat W, et al. Expression of mRNA and protein of macrophage colony-stimulating factor and its receptor in human follicular luteinized granulosa cells. Fertil Steril. 2005;83:419–25.CrossRefPubMed
30.
Zurück zum Zitat Salmassi A, Mettler L, Jonat W, Buck S, Koch K, Schmutzler AG. Circulating level of macrophage colony-stimulating factor can be predictive for human in vitro fertilization outcome. Fertil Steril. 2010;93:116–23.CrossRefPubMed Salmassi A, Mettler L, Jonat W, Buck S, Koch K, Schmutzler AG. Circulating level of macrophage colony-stimulating factor can be predictive for human in vitro fertilization outcome. Fertil Steril. 2010;93:116–23.CrossRefPubMed
31.
Zurück zum Zitat Xu S, Zhang Z, Xia L-X, Huang J. Role of macrophage colony-stimulating factor (M-CSF) in human granulosa cells. Gynecol Endocrinol. 2016;32:1005–8.CrossRefPubMed Xu S, Zhang Z, Xia L-X, Huang J. Role of macrophage colony-stimulating factor (M-CSF) in human granulosa cells. Gynecol Endocrinol. 2016;32:1005–8.CrossRefPubMed
32.
33.
Zurück zum Zitat Sato Y, Cheng Y, Kawamura K, Takae S, Hsueh AJ. C-type natriuretic peptide stimulates ovarian follicle development. Mol Endocrinol. 2012;26:1158–66.CrossRefPubMedPubMedCentral Sato Y, Cheng Y, Kawamura K, Takae S, Hsueh AJ. C-type natriuretic peptide stimulates ovarian follicle development. Mol Endocrinol. 2012;26:1158–66.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Zhang M, Su YQ, Sugiura K, Wigglesworth K, Xia G, Eppig JJ. Estradiol promotes and maintains cumulus cell expression of natriuretic peptide receptor 2 (NPR2) and meiotic arrest in mouse oocytes in vitro. Endocrinology. 2011;152:4377–85.CrossRefPubMedPubMedCentral Zhang M, Su YQ, Sugiura K, Wigglesworth K, Xia G, Eppig JJ. Estradiol promotes and maintains cumulus cell expression of natriuretic peptide receptor 2 (NPR2) and meiotic arrest in mouse oocytes in vitro. Endocrinology. 2011;152:4377–85.CrossRefPubMedPubMedCentral
Metadaten
Titel
Macrophage colony-stimulating factor (M-CSF) is an intermediate in the process of luteinizing hormone-induced decrease in natriuretic peptide receptor 2 (NPR2) and resumption of oocyte meiosis
verfasst von
Wenchao Sun
Chang Liu
Ying Feng
Guangchao Zhuo
Wenjing Zhou
Xiaoyang Fei
Zhifen Zhang
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Journal of Ovarian Research / Ausgabe 1/2017
Elektronische ISSN: 1757-2215
DOI
https://doi.org/10.1186/s13048-017-0364-x

Weitere Artikel der Ausgabe 1/2017

Journal of Ovarian Research 1/2017 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.