Skip to main content
Top
Published in: Journal of Ovarian Research 1/2016

Open Access 01-12-2016 | Research

NPR2 is involved in FSH-mediated mouse oocyte meiotic resumption

Authors: Lei Yang, Qiang Wei, Wei Li, Qihui Xi, Xiaoe Zhao, Baohua Ma

Published in: Journal of Ovarian Research | Issue 1/2016

Login to get access

Abstract

Background

Previous studies have reported that follicle-stimulating hormone (FSH) is often added to culture media to induce oocyte meiotic resumption and maturation and to improve subsequent embryonic development during in vitro maturation (IVM). However, the underlying mechanisms remain unclear.

Methods

Cumulus-oocyte complexes (COCs) were collected from ovaries 46–48 h after the female mice were intraperitoneally injected with 8 IU equine chorionic gonadotropin (eCG) and then the COCs were cultured in different medium. qRT-PCR analysis was used to assess mRNA expression of EGF-like factors and natriuretic peptide receptor 2 (NPR2). Western Blot analysis was used to assess phosphorylation of mitogen-activated protein kinase 3/1 (MAPK3/1). The oocytes were morphologically assessed for meiotic resumption.

Results

FSH stimulated the expression of EGF-like factors, the activation of MAPK3/1, a decrease in NPR2 mRNA and oocyte meiotic resumption. Moreover, the FSH-induced decrease in NPR2 and oocyte meiotic resumption occurred via the MAPK3/1 singling pathway, which was activated by the epidermal growth factor receptor (EGFR) pathway.

Conclusions

NPR2 is involved in FSH-mediated oocyte meiotic resumption, and this process is associated with the EGFR and MAPK3/1 signaling pathways.
Literature
1.
go back to reference Kanatsu-Shinohara M, Schultz RM, Kopf GS. Acquisition of meiotic competence in mouse oocytes: absolute amounts of p34(cdc2), cyclin B1, cdc25C, and wee1 in meiotically incompetent and competent oocytes. Biol Reprod. 2000;63:1610–6.CrossRefPubMed Kanatsu-Shinohara M, Schultz RM, Kopf GS. Acquisition of meiotic competence in mouse oocytes: absolute amounts of p34(cdc2), cyclin B1, cdc25C, and wee1 in meiotically incompetent and competent oocytes. Biol Reprod. 2000;63:1610–6.CrossRefPubMed
2.
go back to reference Zhang M, Su Y-Q, 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.PubMedCentralCrossRefPubMed Zhang M, Su Y-Q, 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.PubMedCentralCrossRefPubMed
3.
go back to reference Shuhaibar LC, Egbert JR, Norris RP, Lampe PD, Nikolaev VO, Thunemann M, et al. Intercellular signaling via cyclic GMP diffusion through gap junctions restarts meiosis in mouse ovarian follicles. P Natl Acad Sci USA. 2015;112:5527–32.CrossRef Shuhaibar LC, Egbert JR, Norris RP, Lampe PD, Nikolaev VO, Thunemann M, et al. Intercellular signaling via cyclic GMP diffusion through gap junctions restarts meiosis in mouse ovarian follicles. P Natl Acad Sci USA. 2015;112:5527–32.CrossRef
4.
go back to reference 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
5.
go back to reference Zhang MJ, Hong OY, Xia GL. The signal pathway of gonadotrophins-induced mammalian oocyte meiotic resumption. Mol Hum Reprod. 2009;15:399–409.CrossRefPubMed Zhang MJ, Hong OY, Xia GL. The signal pathway of gonadotrophins-induced mammalian oocyte meiotic resumption. Mol Hum Reprod. 2009;15:399–409.CrossRefPubMed
6.
go back to reference Gonzalez-Bulnes A, Garcia-Garcia RM, Souza CJH, Santiago-Moreno J, Lopez-Sebastian A, Cocero MJ, et al. Patterns of follicular growth in superovulated sheep and influence on endocrine and ovarian response. Reprod Domest Anim. 2002;37:357–61.CrossRefPubMed Gonzalez-Bulnes A, Garcia-Garcia RM, Souza CJH, Santiago-Moreno J, Lopez-Sebastian A, Cocero MJ, et al. Patterns of follicular growth in superovulated sheep and influence on endocrine and ovarian response. Reprod Domest Anim. 2002;37:357–61.CrossRefPubMed
7.
go back to reference Grondahl C, Breinholt J, Wahl P, Murray A, Hansen TH, Faerge I, et al. Physiology of meiosis-activating sterol: endogenous formation and mode of action. Hum Reprod. 2003;18:122–9.CrossRefPubMed Grondahl C, Breinholt J, Wahl P, Murray A, Hansen TH, Faerge I, et al. Physiology of meiosis-activating sterol: endogenous formation and mode of action. Hum Reprod. 2003;18:122–9.CrossRefPubMed
8.
go back to reference Trounson A, Anderiesz C, Jones G. Maturation of human oocytes in vitro and their developmental competence. Reproduction. 2001;121:51–75.CrossRefPubMed Trounson A, Anderiesz C, Jones G. Maturation of human oocytes in vitro and their developmental competence. Reproduction. 2001;121:51–75.CrossRefPubMed
9.
go back to reference Jurema MW, Nogueira D. In vitro maturation of human oocytes for assisted reproduction. Fertil Steril. 2006;86:1277–91.CrossRefPubMed Jurema MW, Nogueira D. In vitro maturation of human oocytes for assisted reproduction. Fertil Steril. 2006;86:1277–91.CrossRefPubMed
10.
go back to reference Sirard M-A. Follicle environment and quality of in vitro matured oocytes. J Assist Reprod Gen. 2011;28:483–8.CrossRef Sirard M-A. Follicle environment and quality of in vitro matured oocytes. J Assist Reprod Gen. 2011;28:483–8.CrossRef
11.
go back to reference Rossi G, Macchiarelli G, Palmerini MG, Canipari R, Cecconi S. Meiotic spindle configuration is differentially influenced by FSH and epidermal growth factor during in vitro maturation of mouse oocytes. Hum Reprod. 2006;21:1765–70.CrossRefPubMed Rossi G, Macchiarelli G, Palmerini MG, Canipari R, Cecconi S. Meiotic spindle configuration is differentially influenced by FSH and epidermal growth factor during in vitro maturation of mouse oocytes. Hum Reprod. 2006;21:1765–70.CrossRefPubMed
12.
go back to reference Junk SM, Dharmarajan A, Yovich JL. FSH priming improves oocyte maturation, but priming with FSH or hCG has no effect on subsequent embryonic development in an in vitro maturation program. Theriogenology. 2003;59:1741–9.CrossRefPubMed Junk SM, Dharmarajan A, Yovich JL. FSH priming improves oocyte maturation, but priming with FSH or hCG has no effect on subsequent embryonic development in an in vitro maturation program. Theriogenology. 2003;59:1741–9.CrossRefPubMed
13.
go back to reference Li J, Mao G, Xia G. FSH modulates PKAI and GPR3 activities in mouse oocyte of COC in a gap junctional communication (GJC)-dependent manner to initiate meiotic resumption. PLoS One. 2012;7, e37835.PubMedCentralCrossRefPubMed Li J, Mao G, Xia G. FSH modulates PKAI and GPR3 activities in mouse oocyte of COC in a gap junctional communication (GJC)-dependent manner to initiate meiotic resumption. PLoS One. 2012;7, e37835.PubMedCentralCrossRefPubMed
14.
go back to reference Singh B, Barbe GJ, Armstrong DT. Factors influencing resumption of meiotic maturation and cumulus expansion of porcine oocyte-cumulus cell complexes in vitro. Mol Reprod Dev. 1993;36:113–9.CrossRefPubMed Singh B, Barbe GJ, Armstrong DT. Factors influencing resumption of meiotic maturation and cumulus expansion of porcine oocyte-cumulus cell complexes in vitro. Mol Reprod Dev. 1993;36:113–9.CrossRefPubMed
15.
go back to reference Younis A, Brackett B, Fayrer‐Hosken R. Influence of serum and hormones on bovine oocyte maturation and fertilization in vitro. Gamete Research. 1989;23:189–201.CrossRefPubMed Younis A, Brackett B, Fayrer‐Hosken R. Influence of serum and hormones on bovine oocyte maturation and fertilization in vitro. Gamete Research. 1989;23:189–201.CrossRefPubMed
16.
go back to reference Tremoleda JL, Tharasanit T, Van Tol HTA, Stout TAE, Colenbrander B, Bevers MM. Effects of follicular cells and FSH on the resumption of meiosis in equine oocytes matured in vitro. Reproduction. 2003;125:565–77.CrossRefPubMed Tremoleda JL, Tharasanit T, Van Tol HTA, Stout TAE, Colenbrander B, Bevers MM. Effects of follicular cells and FSH on the resumption of meiosis in equine oocytes matured in vitro. Reproduction. 2003;125:565–77.CrossRefPubMed
17.
go back to reference Bolamba D, Russ KD, Harper SA, Sandler JL, Durrant BS. Effects of epidermal growth factor and hormones on granulosa expansion and nuclear maturation of dog oocytes in vitro. Theriogenology. 2006;65:1037–47.CrossRefPubMed Bolamba D, Russ KD, Harper SA, Sandler JL, Durrant BS. Effects of epidermal growth factor and hormones on granulosa expansion and nuclear maturation of dog oocytes in vitro. Theriogenology. 2006;65:1037–47.CrossRefPubMed
18.
go back to reference Lindner HR, Tsafriri A, Lieberman ME, Zor U, Koch Y, Bauminger S, et al. Gonadotropin action on cultured Graafian follicles: induction of maturation division of the mammalian oocyte and differentiation of the luteal cell. Recent Prog Horm Res. 1974;30:79–138.PubMed Lindner HR, Tsafriri A, Lieberman ME, Zor U, Koch Y, Bauminger S, et al. Gonadotropin action on cultured Graafian follicles: induction of maturation division of the mammalian oocyte and differentiation of the luteal cell. Recent Prog Horm Res. 1974;30:79–138.PubMed
19.
go back to reference Eppig JJ. The participation of cyclic adenosine monophosphate (cAMP) in the regulation of meiotic maturation of oocytes in the laboratory mouse. J Reprod Fertil. 1989;38 Suppl 1:3–8. Eppig JJ. The participation of cyclic adenosine monophosphate (cAMP) in the regulation of meiotic maturation of oocytes in the laboratory mouse. J Reprod Fertil. 1989;38 Suppl 1:3–8.
20.
go back to reference Sun Q Y, Miao Y L, Schatten H. Towards a new understanding on the regulation of mammalian oocyte meiosis resumption. Cell Cycle. 2009;8:2741–7.CrossRefPubMed Sun Q Y, Miao Y L, Schatten H. Towards a new understanding on the regulation of mammalian oocyte meiosis resumption. Cell Cycle. 2009;8:2741–7.CrossRefPubMed
21.
go back to reference Liang CG, Huo LJ, Zhong ZS, Chen DY, Schatten H, Sun QY. Cyclic adenosine 3′, 5′-monophosphate-dependent activation of mitogen-activated protein kinase in cumulus cells is essential for germinal vesicle breakdown of porcine cumulus-enclosed oocytes. Endocrinology. 2005;146:4437–44.CrossRefPubMed Liang CG, Huo LJ, Zhong ZS, Chen DY, Schatten H, Sun QY. Cyclic adenosine 3′, 5′-monophosphate-dependent activation of mitogen-activated protein kinase in cumulus cells is essential for germinal vesicle breakdown of porcine cumulus-enclosed oocytes. Endocrinology. 2005;146:4437–44.CrossRefPubMed
22.
go back to reference Meinecke B, Krischek C. MAPK/ERK kinase (MEK) signalling is required for resumption of meiosis in cultured cumulus-enclosed pig oocytes. Zygote. 2003;11:7–16.CrossRefPubMed Meinecke B, Krischek C. MAPK/ERK kinase (MEK) signalling is required for resumption of meiosis in cultured cumulus-enclosed pig oocytes. Zygote. 2003;11:7–16.CrossRefPubMed
23.
go back to reference Richani D, Ritter L, Thompson J, Gilchrist R. Mode of oocyte maturation affects EGF-like peptide function and oocyte competence. Mol Hum Reprod. 2013;19:500–9.CrossRefPubMed Richani D, Ritter L, Thompson J, Gilchrist R. Mode of oocyte maturation affects EGF-like peptide function and oocyte competence. Mol Hum Reprod. 2013;19:500–9.CrossRefPubMed
24.
go back to reference Caixeta ES, Machado MF, Ripamonte P, Price C, Buratini J. Effects of FSH on the expression of receptors for oocyte-secreted factors and members of the EGF-like family during in vitro maturation in cattle. Reprod Fertil Dev. 2013;25:890–9.CrossRefPubMed Caixeta ES, Machado MF, Ripamonte P, Price C, Buratini J. Effects of FSH on the expression of receptors for oocyte-secreted factors and members of the EGF-like family during in vitro maturation in cattle. Reprod Fertil Dev. 2013;25:890–9.CrossRefPubMed
25.
go back to reference Yamashita Y, ShiMADA M. The release of EGF domain from EGF-like factors by a specific cleavage enzyme activates the EGFR-MAPK3/1 pathway in both granulosa cells and cumulus cells during the ovulation process. J Reprod Develop. 2012;58:510–4.CrossRef Yamashita Y, ShiMADA M. The release of EGF domain from EGF-like factors by a specific cleavage enzyme activates the EGFR-MAPK3/1 pathway in both granulosa cells and cumulus cells during the ovulation process. J Reprod Develop. 2012;58:510–4.CrossRef
26.
go back to reference Blaha M, Nemcova L, Prochazka R. Cyclic guanosine monophosphate does not inhibit gonadotropin-induced activation of mitogen-activated protein kinase 3/1 in pig cumulus-oocyte complexes. Reprod Biol Endocrin. 2015;13:1.CrossRef Blaha M, Nemcova L, Prochazka R. Cyclic guanosine monophosphate does not inhibit gonadotropin-induced activation of mitogen-activated protein kinase 3/1 in pig cumulus-oocyte complexes. Reprod Biol Endocrin. 2015;13:1.CrossRef
27.
go back to reference 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
28.
29.
go back to reference Fan H-Y, Huo L-J, Chen D-Y, Schatten H, Sun Q-Y. Protein kinase C and mitogen-activated protein kinase cascade in mouse cumulus cells: cross talk and effect on meiotic resumption of oocyte. Biol Reprod. 2004;70:1178–87.CrossRefPubMed Fan H-Y, Huo L-J, Chen D-Y, Schatten H, Sun Q-Y. Protein kinase C and mitogen-activated protein kinase cascade in mouse cumulus cells: cross talk and effect on meiotic resumption of oocyte. Biol Reprod. 2004;70:1178–87.CrossRefPubMed
30.
go back to reference Sugiura K, Su YQ, Eppig JJ. Targeted suppression of Has2 mRNA in mouse cumulus cell–oocyte complexes by adenovirus‐mediated short‐hairpin RNA expression. Mol Reprod Dev. 2009;76:537–47.PubMedCentralCrossRefPubMed Sugiura K, Su YQ, Eppig JJ. Targeted suppression of Has2 mRNA in mouse cumulus cell–oocyte complexes by adenovirus‐mediated short‐hairpin RNA expression. Mol Reprod Dev. 2009;76:537–47.PubMedCentralCrossRefPubMed
31.
go back to reference Chen X, Zhou B, Yan J, Xu B, Tai P, Li J, et al. Epidermal growth factor receptor activation by protein kinase C is necessary for FSH-induced meiotic resumption in porcine cumulus–oocyte complexes. J Endocrinol. 2008;197:409–19.CrossRefPubMed Chen X, Zhou B, Yan J, Xu B, Tai P, Li J, et al. Epidermal growth factor receptor activation by protein kinase C is necessary for FSH-induced meiotic resumption in porcine cumulus–oocyte complexes. J Endocrinol. 2008;197:409–19.CrossRefPubMed
32.
go back to reference Prochazka R, Blaha M, Nemcova L. Signaling pathways regulating FSH-and amphiregulin-induced meiotic resumption and cumulus cell expansion in the pig. Reproduction. 2012;144:535–46.CrossRefPubMed Prochazka R, Blaha M, Nemcova L. Signaling pathways regulating FSH-and amphiregulin-induced meiotic resumption and cumulus cell expansion in the pig. Reproduction. 2012;144:535–46.CrossRefPubMed
33.
go back to reference Su Y-Q, Wigglesworth K, Pendola FL, O’Brien MJ, Eppig JJ. Mitogen-activated protein kinase activity in cumulus cells is essential for gonadotropin-induced oocyte meiotic resumption and cumulus expansion in the mouse. Endocrinology. 2002;143:2221–32.CrossRefPubMed Su Y-Q, Wigglesworth K, Pendola FL, O’Brien MJ, Eppig JJ. Mitogen-activated protein kinase activity in cumulus cells is essential for gonadotropin-induced oocyte meiotic resumption and cumulus expansion in the mouse. Endocrinology. 2002;143:2221–32.CrossRefPubMed
34.
go back to reference 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
35.
go back to reference Lee K-B, Zhang M, Sugiura K, Wigglesworth K, Uliasz T, Jaffe LA, et al. Hormonal coordination of natriuretic peptide type C and natriuretic peptide receptor 3 expression in mouse granulosa cells. Biol Reprod. 2013;88:42.PubMedCentralCrossRefPubMed Lee K-B, Zhang M, Sugiura K, Wigglesworth K, Uliasz T, Jaffe LA, et al. Hormonal coordination of natriuretic peptide type C and natriuretic peptide receptor 3 expression in mouse granulosa cells. Biol Reprod. 2013;88:42.PubMedCentralCrossRefPubMed
36.
go back to reference Zhang M, Su Y-Q, 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.PubMedCentralCrossRefPubMed Zhang M, Su Y-Q, 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.PubMedCentralCrossRefPubMed
37.
38.
go back to reference Zhang J, Wei Q, Cai J, Zhao X, Ma B. Effect of C-type natriuretic peptide on maturation and developmental competence of goat oocytes matured in vitro. PLoS One. 2015;10. Zhang J, Wei Q, Cai J, Zhao X, Ma B. Effect of C-type natriuretic peptide on maturation and developmental competence of goat oocytes matured in vitro. PLoS One. 2015;10.
39.
go back to reference Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Bio. 2001;2:127–37.CrossRef Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Bio. 2001;2:127–37.CrossRef
40.
go back to reference Holbro T, Hynes NE. ErbB receptors: directing key signaling networks throughout life. Annu Rev Pharmacol. 2004;44:195–217.CrossRef Holbro T, Hynes NE. ErbB receptors: directing key signaling networks throughout life. Annu Rev Pharmacol. 2004;44:195–217.CrossRef
41.
go back to reference Dedieu T, Gall L, Crozet N, Sevellec C, Ruffini S. Mitogen‐activated protein kinase activity during goat oocyte maturation and the acquisition of meiotic competence. Mol Reprod Dev. 1996;45:351–8.CrossRefPubMed Dedieu T, Gall L, Crozet N, Sevellec C, Ruffini S. Mitogen‐activated protein kinase activity during goat oocyte maturation and the acquisition of meiotic competence. Mol Reprod Dev. 1996;45:351–8.CrossRefPubMed
42.
go back to reference Schmitt A, Nebreda AR. Signalling pathways in oocyte meiotic maturation. J Cell Sci. 2002;115:2457–9.PubMed Schmitt A, Nebreda AR. Signalling pathways in oocyte meiotic maturation. J Cell Sci. 2002;115:2457–9.PubMed
43.
go back to reference Su Y-Q, Denegre JM, Wigglesworth K, Pendola FL, O’Brien MJ, Eppig JJ. Oocyte-dependent activation of mitogen-activated protein kinase (ERK1/2) in cumulus cells is required for the maturation of the mouse oocyte–cumulus cell complex. Dev Biol. 2003;263:126–38.CrossRefPubMed Su Y-Q, Denegre JM, Wigglesworth K, Pendola FL, O’Brien MJ, Eppig JJ. Oocyte-dependent activation of mitogen-activated protein kinase (ERK1/2) in cumulus cells is required for the maturation of the mouse oocyte–cumulus cell complex. Dev Biol. 2003;263:126–38.CrossRefPubMed
44.
go back to reference 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
Metadata
Title
NPR2 is involved in FSH-mediated mouse oocyte meiotic resumption
Authors
Lei Yang
Qiang Wei
Wei Li
Qihui Xi
Xiaoe Zhao
Baohua Ma
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Ovarian Research / Issue 1/2016
Electronic ISSN: 1757-2215
DOI
https://doi.org/10.1186/s13048-016-0218-y

Other articles of this Issue 1/2016

Journal of Ovarian Research 1/2016 Go to the issue