Skip to main content
Top
Published in: Reproductive Biology and Endocrinology 1/2019

Open Access 01-12-2019 | Review

The art of oocyte meiotic arrest regulation

Authors: Bo Pan, Julang Li

Published in: Reproductive Biology and Endocrinology | Issue 1/2019

Login to get access

Abstract

A central dogma of mammalian reproductive biology is that the size of the primordial follicle pool represents reproductive capacity in females. The assembly of the primordial follicle starts after the primordial germ cells (PGCs)-derived oocyte releases from the synchronously dividing germline cysts. PGCs initiate meiosis during fetal development. However, after synapsis and recombination of homologous chromosomes, they arrest at the diplotene stage of the first meiotic prophase (MI). The diplotene-arrested oocyte, together with the surrounding of a single layer of flattened granulosa cells, forms a basic unit of the ovary, the primordial follicle. At the start of each estrous (animal) or menstrual cycle (human), in response to a surge of luteinizing hormone (LH) from the pituitary gland, a limited number of primordial follicles are triggered to develop into primary follicles, preantral follicles, antral follicles and reach to preovulatory follicle stage. During the transition from the preantral to antral stages, the enclosed oocyte gradually acquires the capacity to resume meiosis. Meiotic resumption from the prophase of MI is morphologically characterized by the dissolution of the oocyte nuclear envelope, which is generally termed the “germinal vesicle breakdown” (GVBD). Following GVBD and completion of MI, the oocyte enters meiosis II without an obvious S-phase and arrests at metaphase phase II (MII) until fertilization. The underlying mechanism of meiotic arrest has been widely explored in numerous studies. Many studies indicated that two cellular second messengers, cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) play an essential role in maintaining oocyte meiotic arrest. This review will discuss how these two cyclic nucleotides regulate oocyte maturation by blocking or initiating meiotic processes, and to provide an insight in future research.
Literature
1.
go back to reference Jones KT. Turning it on and off: M-phase promoting factor during meiotic maturation and fertilization. Mol Hum Reprod. 2004;10:1–5.PubMedCrossRef Jones KT. Turning it on and off: M-phase promoting factor during meiotic maturation and fertilization. Mol Hum Reprod. 2004;10:1–5.PubMedCrossRef
2.
go back to reference Hashimoto N, Kishimoto T. Regulation of meiotic metaphase by a cytoplasmic maturation-promoting factor during mouse oocyte maturation. Dev Biol. 1988;126:242–52.PubMedCrossRef Hashimoto N, Kishimoto T. Regulation of meiotic metaphase by a cytoplasmic maturation-promoting factor during mouse oocyte maturation. Dev Biol. 1988;126:242–52.PubMedCrossRef
3.
go back to reference Gautier J, Norbury C, Lohka M, Nurse P, Maller J. Purified maturation-promoting factor contains the product of a Xenopus homolog of the fission yeast cell cycle control gene cdc2+. Cell. 1988;54:433–9.PubMedCrossRef Gautier J, Norbury C, Lohka M, Nurse P, Maller J. Purified maturation-promoting factor contains the product of a Xenopus homolog of the fission yeast cell cycle control gene cdc2+. Cell. 1988;54:433–9.PubMedCrossRef
4.
go back to reference Han SJ, Conti M. New pathways from PKA to the Cdc2/cyclin B complex in oocytes: Wee1B as a potential PKA substrate. Cell Cycle. 2006;5:227–31.PubMedCrossRef Han SJ, Conti M. New pathways from PKA to the Cdc2/cyclin B complex in oocytes: Wee1B as a potential PKA substrate. Cell Cycle. 2006;5:227–31.PubMedCrossRef
5.
go back to reference Oh JS, Han SJ, Conti M. Wee1B, Myt1, and Cdc25 function in distinct compartments of the mouse oocyte to control meiotic resumption. J Cell Biol. 2010;188:199–207.PubMedPubMedCentralCrossRef Oh JS, Han SJ, Conti M. Wee1B, Myt1, and Cdc25 function in distinct compartments of the mouse oocyte to control meiotic resumption. J Cell Biol. 2010;188:199–207.PubMedPubMedCentralCrossRef
6.
go back to reference Lincoln AJ, Wickramasinghe D, Stein P, Schultz RM, Palko ME, De Miguel MP, Tessarollo L, Donovan PJ. Cdc25b phosphatase is required for resumption of meiosis during oocyte maturation. Nat Genet. 2002;30:446–9.PubMedCrossRef Lincoln AJ, Wickramasinghe D, Stein P, Schultz RM, Palko ME, De Miguel MP, Tessarollo L, Donovan PJ. Cdc25b phosphatase is required for resumption of meiosis during oocyte maturation. Nat Genet. 2002;30:446–9.PubMedCrossRef
7.
go back to reference Solc P, Saskova A, Baran V, Kubelka M, Schultz RM, Motlik J. CDC25A phosphatase controls meiosis I progression in mouse oocytes. Dev Biol. 2008;317:260–9.PubMedPubMedCentralCrossRef Solc P, Saskova A, Baran V, Kubelka M, Schultz RM, Motlik J. CDC25A phosphatase controls meiosis I progression in mouse oocytes. Dev Biol. 2008;317:260–9.PubMedPubMedCentralCrossRef
8.
go back to reference Ray D, Terao Y, Nimbalkar D, Hirai H, Osmundson EC, Zou X, Franks R, Christov K, Kiyokawa H. Hemizygous disruption of Cdc25A inhibits cellular transformation and mammary tumorigenesis in mice. Cancer Res. 2007;67:6605–11.PubMedCrossRef Ray D, Terao Y, Nimbalkar D, Hirai H, Osmundson EC, Zou X, Franks R, Christov K, Kiyokawa H. Hemizygous disruption of Cdc25A inhibits cellular transformation and mammary tumorigenesis in mice. Cancer Res. 2007;67:6605–11.PubMedCrossRef
9.
go back to reference Dekel N, Lawrence TS, Gilula NB, Beers WH. Modulation of cell-to-cell communication in the cumulus-oocyte complex and the regulation of oocyte maturation by LH. Dev Biol. 1981;86:356–62.PubMedCrossRef Dekel N, Lawrence TS, Gilula NB, Beers WH. Modulation of cell-to-cell communication in the cumulus-oocyte complex and the regulation of oocyte maturation by LH. Dev Biol. 1981;86:356–62.PubMedCrossRef
10.
go back to reference Webb RJ, Marshall F, Swann K, Carroll J. Follicle-stimulating hormone induces a gap junction-dependent dynamic change in [cAMP] and protein kinase a in mammalian oocytes. Dev Biol. 2002;246:441–54.PubMedCrossRef Webb RJ, Marshall F, Swann K, Carroll J. Follicle-stimulating hormone induces a gap junction-dependent dynamic change in [cAMP] and protein kinase a in mammalian oocytes. Dev Biol. 2002;246:441–54.PubMedCrossRef
11.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
12.
go back to reference Eppig JJ, Freter RR, Ward-Bailey PF, Schultz RM. Inhibition of oocyte maturation in the mouse: participation of cAMP, steroid hormones, and a putative maturation-inhibitory factor. Dev Biol. 1983;100:39–49.PubMedCrossRef Eppig JJ, Freter RR, Ward-Bailey PF, Schultz RM. Inhibition of oocyte maturation in the mouse: participation of cAMP, steroid hormones, and a putative maturation-inhibitory factor. Dev Biol. 1983;100:39–49.PubMedCrossRef
13.
go back to reference Bornslaeger EA, Schultz RM. Regulation of mouse oocyte maturation: effect of elevating cumulus cell cAMP on oocyte cAMP levels. Biol Reprod. 1985;33:698–704.PubMedCrossRef Bornslaeger EA, Schultz RM. Regulation of mouse oocyte maturation: effect of elevating cumulus cell cAMP on oocyte cAMP levels. Biol Reprod. 1985;33:698–704.PubMedCrossRef
14.
go back to reference Salustri A, Petrungaro S, De Felici M, Conti M, Siracusa G. Effect of follicle-stimulating hormone on cyclic adenosine monophosphate level and on meiotic maturation in mouse cumulus cell-enclosed oocytes cultured in vitro. Biol Reprod. 1985;33:797–802.PubMedCrossRef Salustri A, Petrungaro S, De Felici M, Conti M, Siracusa G. Effect of follicle-stimulating hormone on cyclic adenosine monophosphate level and on meiotic maturation in mouse cumulus cell-enclosed oocytes cultured in vitro. Biol Reprod. 1985;33:797–802.PubMedCrossRef
15.
go back to reference Sherizly I, Galiani D, Dekel N. Regulation of oocyte maturation: communication in the rat cumulus-oocyte complex. Hum Reprod. 1988;3:761–6.PubMedCrossRef Sherizly I, Galiani D, Dekel N. Regulation of oocyte maturation: communication in the rat cumulus-oocyte complex. Hum Reprod. 1988;3:761–6.PubMedCrossRef
16.
go back to reference Yoshimura Y, Nakamura Y, Ando M, Jinno M, Oda T, Karube M, Koyama N, Nanno T. Stimulatory role of cyclic adenosine-monophosphate as a mediator of meiotic resumption in rabbit oocytes. Endocrinology. 1992;131:351–6.PubMedCrossRef Yoshimura Y, Nakamura Y, Ando M, Jinno M, Oda T, Karube M, Koyama N, Nanno T. Stimulatory role of cyclic adenosine-monophosphate as a mediator of meiotic resumption in rabbit oocytes. Endocrinology. 1992;131:351–6.PubMedCrossRef
17.
go back to reference Thomas RE, Armstrong DT, Gilchrist RB. Differential effects of specific phosphodiesterase isoenzyme inhibitors on bovine oocyte meiotic maturation. Dev Biol. 2002;244:215–25.PubMedCrossRef Thomas RE, Armstrong DT, Gilchrist RB. Differential effects of specific phosphodiesterase isoenzyme inhibitors on bovine oocyte meiotic maturation. Dev Biol. 2002;244:215–25.PubMedCrossRef
18.
go back to reference Burghardt RC, Barhoumi R, Sewall TC, Bowen JA. Cyclic AMP induces rapid increases in gap junction permeability and changes in the cellular distribution of connexin43. J Membr Biol. 1995;148:243–53.PubMedCrossRef Burghardt RC, Barhoumi R, Sewall TC, Bowen JA. Cyclic AMP induces rapid increases in gap junction permeability and changes in the cellular distribution of connexin43. J Membr Biol. 1995;148:243–53.PubMedCrossRef
19.
go back to reference Sandberg K, Ji H, Iida T, Catt KJ. Intercellular communication between follicular angiotensin receptors and Xenopus laevis oocytes: medication by an inositol 1,4,5-trisphosphate-dependent mechanism. J Cell Biol. 1992;117:157–67.PubMedCrossRef Sandberg K, Ji H, Iida T, Catt KJ. Intercellular communication between follicular angiotensin receptors and Xenopus laevis oocytes: medication by an inositol 1,4,5-trisphosphate-dependent mechanism. J Cell Biol. 1992;117:157–67.PubMedCrossRef
20.
go back to reference Chesnel F, Wigglesworth K, Eppig JJ. Acquisition of meiotic competence by denuded mouse oocytes: participation of somatic-cell product(s) and cAMP. Dev Biol. 1994;161:285–95.PubMedCrossRef Chesnel F, Wigglesworth K, Eppig JJ. Acquisition of meiotic competence by denuded mouse oocytes: participation of somatic-cell product(s) and cAMP. Dev Biol. 1994;161:285–95.PubMedCrossRef
21.
go back to reference Liu J, Erlichman B, Weinstein LS. The stimulatory G protein α-subunit Gsα is imprinted in human thyroid glands: implications for thyroid function in pseudohypoparathyroidism types 1A and 1B. J Clin Endocrinol Metab. 2003;88:4336–41.PubMedCrossRef Liu J, Erlichman B, Weinstein LS. The stimulatory G protein α-subunit Gsα is imprinted in human thyroid glands: implications for thyroid function in pseudohypoparathyroidism types 1A and 1B. J Clin Endocrinol Metab. 2003;88:4336–41.PubMedCrossRef
22.
go back to reference Mehlmann LM, Jones TL, Jaffe LA. Meiotic arrest in the mouse follicle maintained by a Gs protein in the oocyte. Science. 2002;297:1343–5.PubMedCrossRef Mehlmann LM, Jones TL, Jaffe LA. Meiotic arrest in the mouse follicle maintained by a Gs protein in the oocyte. Science. 2002;297:1343–5.PubMedCrossRef
23.
go back to reference Kalinowski RR, Berlot CH, Jones TL, Ross LF, Jaffe LA, Mehlmann LM. Maintenance of meiotic prophase arrest in vertebrate oocytes by a Gs protein-mediated pathway. Dev Biol. 2004;267:1–13.PubMedCrossRef Kalinowski RR, Berlot CH, Jones TL, Ross LF, Jaffe LA, Mehlmann LM. Maintenance of meiotic prophase arrest in vertebrate oocytes by a Gs protein-mediated pathway. Dev Biol. 2004;267:1–13.PubMedCrossRef
24.
go back to reference Freudzon L, Norris RP, Hand AR, Tanaka S, Saeki Y, Jones TL, Rasenick MM, Berlot CH, Mehlmann LM, Jaffe LA. Regulation of meiotic prophase arrest in mouse oocytes by GPR3, a constitutive activator of the Gs G protein. J Cell Biol. 2005;171:255–65.PubMedPubMedCentralCrossRef Freudzon L, Norris RP, Hand AR, Tanaka S, Saeki Y, Jones TL, Rasenick MM, Berlot CH, Mehlmann LM, Jaffe LA. Regulation of meiotic prophase arrest in mouse oocytes by GPR3, a constitutive activator of the Gs G protein. J Cell Biol. 2005;171:255–65.PubMedPubMedCentralCrossRef
25.
26.
go back to reference Yang CR, Wei YC, Qi ST, Chen L, Zhang QH, Ma JY, Luo YB, Wang YP, Hou Y, Schatten H, Liu ZH, Sun QY. The G Protein coupled receptor 3 is involved in cAMP and cGMP signaling and maintenance of meiotic arrest in porcine oocytes. PLoS One. 2012;7:e38807.PubMedPubMedCentralCrossRef Yang CR, Wei YC, Qi ST, Chen L, Zhang QH, Ma JY, Luo YB, Wang YP, Hou Y, Schatten H, Liu ZH, Sun QY. The G Protein coupled receptor 3 is involved in cAMP and cGMP signaling and maintenance of meiotic arrest in porcine oocytes. PLoS One. 2012;7:e38807.PubMedPubMedCentralCrossRef
27.
go back to reference Deng J, Lang S, Wylie C, Hammes SR. The Xenopus laevis isoform of G protein-coupled receptor 3 (GPR3) is a constitutively active cell surface receptor that participates in maintaining meiotic arrest in X-laevis oocytes. Mol Endocrinol. 2008;22:1853–65.PubMedPubMedCentralCrossRef Deng J, Lang S, Wylie C, Hammes SR. The Xenopus laevis isoform of G protein-coupled receptor 3 (GPR3) is a constitutively active cell surface receptor that participates in maintaining meiotic arrest in X-laevis oocytes. Mol Endocrinol. 2008;22:1853–65.PubMedPubMedCentralCrossRef
28.
go back to reference Pang YF, Thomas P. Involvement of estradiol-17 beta and its membrane receptor, G protein coupled receptor 30 (GPR30) in regulation of oocyte maturation in zebrafish, Danio rario. Gen Comp Endocr. 2009;161:58–61.PubMedCrossRef Pang YF, Thomas P. Involvement of estradiol-17 beta and its membrane receptor, G protein coupled receptor 30 (GPR30) in regulation of oocyte maturation in zebrafish, Danio rario. Gen Comp Endocr. 2009;161:58–61.PubMedCrossRef
29.
go back to reference Hinckley M, Vaccari S, Horner K, Chen R, Conti M. The G-protein-coupled receptors GPR3 and GPR12 are involved in cAMP signaling and maintenance of meiotic arrest in rodent oocytes. Dev Biol. 2005;287:249–61.PubMedCrossRef Hinckley M, Vaccari S, Horner K, Chen R, Conti M. The G-protein-coupled receptors GPR3 and GPR12 are involved in cAMP signaling and maintenance of meiotic arrest in rodent oocytes. Dev Biol. 2005;287:249–61.PubMedCrossRef
30.
go back to reference Mehlmann LM, Saeki Y, Tanaka S, Brennan TJ, Evsikov AV, Pendola FL, Knowles BB, Eppig JJ, Jaffe LA. The Gs-linked receptor GPR3 maintains meiotic arrest in mammalian oocytes. Science. 2004;306:1947–50.PubMedCrossRef Mehlmann LM, Saeki Y, Tanaka S, Brennan TJ, Evsikov AV, Pendola FL, Knowles BB, Eppig JJ, Jaffe LA. The Gs-linked receptor GPR3 maintains meiotic arrest in mammalian oocytes. Science. 2004;306:1947–50.PubMedCrossRef
31.
go back to reference Sasseville M, Albuz FK, Cote N, Guillemette C, Gilchrist RB, Richard FJ. Characterization of novel phosphodiesterases in the bovine ovarian follicle. Biol Reprod. 2009;81:415–25.PubMedPubMedCentralCrossRef Sasseville M, Albuz FK, Cote N, Guillemette C, Gilchrist RB, Richard FJ. Characterization of novel phosphodiesterases in the bovine ovarian follicle. Biol Reprod. 2009;81:415–25.PubMedPubMedCentralCrossRef
32.
go back to reference Horner K, Livera G, Hinckley M, Trinh K, Storm D, Conti M. Rodent oocytes express an active adenylyl cyclase required for meiotic arrest. Dev Biol. 2003;258:385–96.PubMedCrossRef Horner K, Livera G, Hinckley M, Trinh K, Storm D, Conti M. Rodent oocytes express an active adenylyl cyclase required for meiotic arrest. Dev Biol. 2003;258:385–96.PubMedCrossRef
33.
go back to reference Finidorilepicard J, Schorderetslatkine S, Hanoune J, Baulieu EE. Progesterone inhibits membrane-bound adenylate-cyclase in Xenopus-Laevis oocytes. Nature. 1981;292:255–7.CrossRef Finidorilepicard J, Schorderetslatkine S, Hanoune J, Baulieu EE. Progesterone inhibits membrane-bound adenylate-cyclase in Xenopus-Laevis oocytes. Nature. 1981;292:255–7.CrossRef
34.
go back to reference Sadler SE, Maller JL. Progesterone inhibits adenylate-cyclase in Xenopus oocytes - action on the guanine-nucleotide regulatory protein. J Biol Chem. 1981;256:6368–73.PubMed Sadler SE, Maller JL. Progesterone inhibits adenylate-cyclase in Xenopus oocytes - action on the guanine-nucleotide regulatory protein. J Biol Chem. 1981;256:6368–73.PubMed
36.
go back to reference Buck J, Sinclair ML, Schapal L, Cann MJ, Levin LR. Cytosolic adenylyl cyclase defines a unique signaling molecule in mammals. Proc Natl Acad Sci U S A. 1999;96:79–84.PubMedPubMedCentralCrossRef Buck J, Sinclair ML, Schapal L, Cann MJ, Levin LR. Cytosolic adenylyl cyclase defines a unique signaling molecule in mammals. Proc Natl Acad Sci U S A. 1999;96:79–84.PubMedPubMedCentralCrossRef
37.
go back to reference Chen YQ, Cann MJ, Litvin TN, Iourgenko V, Sinclair ML, Levin LR, Buck J. Soluble adenylyl cyclase as an evolutionarily conserved bicarbonate sensor. Science. 2000;289:625–8.PubMedCrossRef Chen YQ, Cann MJ, Litvin TN, Iourgenko V, Sinclair ML, Levin LR, Buck J. Soluble adenylyl cyclase as an evolutionarily conserved bicarbonate sensor. Science. 2000;289:625–8.PubMedCrossRef
38.
go back to reference Esposito G, Jaiswal BS, Xie F, Krajnc-Franken MAM, Robben TJAA, Strik AM, Kuil C, Philipsen RLA, van Duin M, Conti M, Gossen JA. Mice deficient for soluble adenylyl cyclase are infertile because of a severe sperm-motility defect (vol 101, pg 2993, 2004). P Natl Acad Sci USA 2004; 101:5180–5180. Esposito G, Jaiswal BS, Xie F, Krajnc-Franken MAM, Robben TJAA, Strik AM, Kuil C, Philipsen RLA, van Duin M, Conti M, Gossen JA. Mice deficient for soluble adenylyl cyclase are infertile because of a severe sperm-motility defect (vol 101, pg 2993, 2004). P Natl Acad Sci USA 2004; 101:5180–5180.
39.
go back to reference Xie F, Garcia MA, Carlson AE, Schuh SM, Babcock DF, Jaiswal BS, Gossen JA, Esposito G, van Duin M, Conti M. Soluble adenylyl cyclase (sAC) is indispensable for sperm function and fertilization. Dev Biol. 2006;296:353–62.PubMedCrossRef Xie F, Garcia MA, Carlson AE, Schuh SM, Babcock DF, Jaiswal BS, Gossen JA, Esposito G, van Duin M, Conti M. Soluble adenylyl cyclase (sAC) is indispensable for sperm function and fertilization. Dev Biol. 2006;296:353–62.PubMedCrossRef
40.
go back to reference Conti M, Jin SL. The molecular biology of cyclic nucleotide phosphodiesterases. Prog Nucleic Acid Res Mol Biol. 1999;63:1–38.PubMedCrossRef Conti M, Jin SL. The molecular biology of cyclic nucleotide phosphodiesterases. Prog Nucleic Acid Res Mol Biol. 1999;63:1–38.PubMedCrossRef
41.
go back to reference Kenan Y, Murata T, Shakur Y, Degerman E, Manganiello VC. Functions of the N-terminal region of cyclic nucleotide phosphodiesterase 3 (PDE 3) isoforms. J Biol Chem. 2000;275:12331–8.PubMedCrossRef Kenan Y, Murata T, Shakur Y, Degerman E, Manganiello VC. Functions of the N-terminal region of cyclic nucleotide phosphodiesterase 3 (PDE 3) isoforms. J Biol Chem. 2000;275:12331–8.PubMedCrossRef
42.
go back to reference Richard FJ, Tsafriri A, Conti M. Role of phosphodiesterase type 3A in rat oocyte maturation. Biol Reprod. 2001;65:1444–51.PubMedCrossRef Richard FJ, Tsafriri A, Conti M. Role of phosphodiesterase type 3A in rat oocyte maturation. Biol Reprod. 2001;65:1444–51.PubMedCrossRef
43.
go back to reference Reinhardt RR, Chin E, Zhou J, Taira M, Murata T, Manganiello VC, Bondy CA. Distinctive anatomical patterns of gene expression for cGMP-inhibited cyclic nucleotide phosphodiesterases (vol 95, pg 1528, 1995). J Clin Invest 1997; 99:551–551. Reinhardt RR, Chin E, Zhou J, Taira M, Murata T, Manganiello VC, Bondy CA. Distinctive anatomical patterns of gene expression for cGMP-inhibited cyclic nucleotide phosphodiesterases (vol 95, pg 1528, 1995). J Clin Invest 1997; 99:551–551.
44.
go back to reference Tsafriri A, Chun SY, Zhang R, Hsueh AJW, Conti M. Oocyte maturation involves compartmentalization and opposing changes of cAMP levels in follicular somatic and germ cells: studies using selective phosphodiesterase inhibitors. Dev Biol. 1996;178:393–402.PubMedCrossRef Tsafriri A, Chun SY, Zhang R, Hsueh AJW, Conti M. Oocyte maturation involves compartmentalization and opposing changes of cAMP levels in follicular somatic and germ cells: studies using selective phosphodiesterase inhibitors. Dev Biol. 1996;178:393–402.PubMedCrossRef
45.
go back to reference Shitsukawa K, Andersen CB, Richard FJ, Horner AK, Wiersma A, van Duin M, Conti M. Cloning and characterization of the cyclic guanosine monophosphate-inhibited phosphodiesterase PDE3A expressed in mouse oocyte. Biol Reprod. 2001;65:188–96.PubMedCrossRef Shitsukawa K, Andersen CB, Richard FJ, Horner AK, Wiersma A, van Duin M, Conti M. Cloning and characterization of the cyclic guanosine monophosphate-inhibited phosphodiesterase PDE3A expressed in mouse oocyte. Biol Reprod. 2001;65:188–96.PubMedCrossRef
46.
go back to reference Mayes MA, Sirard MA. Effect of type 3 and type 4 phosphodiesterase inhibitors on the maintenance of bovine oocytes in meiotic arrest. Biol Reprod. 2002;66:180–4.PubMedCrossRef Mayes MA, Sirard MA. Effect of type 3 and type 4 phosphodiesterase inhibitors on the maintenance of bovine oocytes in meiotic arrest. Biol Reprod. 2002;66:180–4.PubMedCrossRef
47.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
48.
go back to reference Shu YM, Zeng HT, Ren Z, Zhuang GL, Liang XY, Shen HW, Yao SZ, Ke PQ, Wang NN. Effects of cilostamide and forskolin on the meiotic resumption and embryonic development of immature human oocytes. Hum Reprod. 2008;23:504–13.PubMedCrossRef Shu YM, Zeng HT, Ren Z, Zhuang GL, Liang XY, Shen HW, Yao SZ, Ke PQ, Wang NN. Effects of cilostamide and forskolin on the meiotic resumption and embryonic development of immature human oocytes. Hum Reprod. 2008;23:504–13.PubMedCrossRef
49.
go back to reference Vanhoutte L, De Sutter P, Nogueira D, Gerris J, Dhont M, Van der Elst J. Nuclear and cytoplasmic maturation of in vitro matured human oocytes after temporary nuclear arrest by phosphodiesterase 3-inhibitor. Hum Reprod. 2007;22:1239–46.PubMedCrossRef Vanhoutte L, De Sutter P, Nogueira D, Gerris J, Dhont M, Van der Elst J. Nuclear and cytoplasmic maturation of in vitro matured human oocytes after temporary nuclear arrest by phosphodiesterase 3-inhibitor. Hum Reprod. 2007;22:1239–46.PubMedCrossRef
50.
go back to reference Coticchio G, Rossi G, Borini A, Grondahl C, Macchiarelli G, Flamigni C, Fleming S, Cecconi S. Mouse oocyte meiotic resumption and polar body extrusion in vitro are differentially influenced by FSH, epidermal growth factor and meiosis-activating sterol. Hum Reprod. 2004;19:2913–8.PubMedCrossRef Coticchio G, Rossi G, Borini A, Grondahl C, Macchiarelli G, Flamigni C, Fleming S, Cecconi S. Mouse oocyte meiotic resumption and polar body extrusion in vitro are differentially influenced by FSH, epidermal growth factor and meiosis-activating sterol. Hum Reprod. 2004;19:2913–8.PubMedCrossRef
51.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
52.
go back to reference Masciarelli S, Horner K, Liu CY, Park SH, Hinckley M, Hockman S, Nedachi T, Jin C, Conti M, Manganiello V. Cyclic nucleotide phosphodiesterase 3A-deficient mice as a model of female infertility. J Clin Invest. 2004;114:196–205.PubMedPubMedCentralCrossRef Masciarelli S, Horner K, Liu CY, Park SH, Hinckley M, Hockman S, Nedachi T, Jin C, Conti M, Manganiello V. Cyclic nucleotide phosphodiesterase 3A-deficient mice as a model of female infertility. J Clin Invest. 2004;114:196–205.PubMedPubMedCentralCrossRef
53.
54.
go back to reference Ignatov A, Lintzel J, Hermans-Borgmeyer I, Kreienkamp H-J, Joost P, Thomsen S, Methner A, Schaller HC. Role of the G-protein-coupled receptor GPR12 as high-affinity receptor for sphingosylphosphorylcholine and its expression and function in brain development. J Neurosci. 2003;23:907–14.PubMedCrossRef Ignatov A, Lintzel J, Hermans-Borgmeyer I, Kreienkamp H-J, Joost P, Thomsen S, Methner A, Schaller HC. Role of the G-protein-coupled receptor GPR12 as high-affinity receptor for sphingosylphosphorylcholine and its expression and function in brain development. J Neurosci. 2003;23:907–14.PubMedCrossRef
55.
go back to reference Ye X. Lysophospholipid signaling in the function and pathology of the reproductive system. Hum Reprod Update. 2008;14:519–36.PubMedCrossRef Ye X. Lysophospholipid signaling in the function and pathology of the reproductive system. Hum Reprod Update. 2008;14:519–36.PubMedCrossRef
56.
go back to reference Hinokio K, Yamano S, Nakagawa K, Iraharaa M, Kamada M, Tokumura A, Aono T. Lysophosphatidic acid stimulates nuclear and cytoplasmic maturation of golden hamster immature oocytes in vitro via cumulus cells. Life Sci. 2002;70:759–67.PubMedCrossRef Hinokio K, Yamano S, Nakagawa K, Iraharaa M, Kamada M, Tokumura A, Aono T. Lysophosphatidic acid stimulates nuclear and cytoplasmic maturation of golden hamster immature oocytes in vitro via cumulus cells. Life Sci. 2002;70:759–67.PubMedCrossRef
57.
go back to reference Komatsu J, Yamano S, Kuwahara A, Tokumura A, Irahara M. The signaling pathways linking to lysophosphatidic acid-promoted meiotic maturation in mice. Life Sci. 2006;79:506–11.PubMedCrossRef Komatsu J, Yamano S, Kuwahara A, Tokumura A, Irahara M. The signaling pathways linking to lysophosphatidic acid-promoted meiotic maturation in mice. Life Sci. 2006;79:506–11.PubMedCrossRef
58.
go back to reference Liang CG, Su YQ, Fan HY, Schatten H, Sun QY. Mechanisms regulating oocyte meiotic resumption: roles of mitogen-activated protein kinase. Mol Endocrinol. 2007;21:2037–55.PubMedCrossRef Liang CG, Su YQ, Fan HY, Schatten H, Sun QY. Mechanisms regulating oocyte meiotic resumption: roles of mitogen-activated protein kinase. Mol Endocrinol. 2007;21:2037–55.PubMedCrossRef
59.
go back to reference Turko IV, Ballard SA, Francis SH, Corbin JD. Inhibition of cyclic GMP-binding cyclic GMP-specific phosphodiesterase (type 5) by sildenafil and related compounds. Mol Pharmacol. 1999;56:124–30.PubMedCrossRef Turko IV, Ballard SA, Francis SH, Corbin JD. Inhibition of cyclic GMP-binding cyclic GMP-specific phosphodiesterase (type 5) by sildenafil and related compounds. Mol Pharmacol. 1999;56:124–30.PubMedCrossRef
60.
go back to reference Shuhaibar LC, Egbert JR, Norris RP, Lampe PD, Nikolaev VO, Thunemann M, Wen L, Feil R, Jaffe LA. 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, Wen L, Feil R, Jaffe LA. 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
61.
go back to reference Jaffe LA, Egbert JR. Regulation of mammalian oocyte meiosis by intercellular communication within the ovarian follicle. Annu Rev Physiol. 2017;79:237–60.PubMedCrossRef Jaffe LA, Egbert JR. Regulation of mammalian oocyte meiosis by intercellular communication within the ovarian follicle. Annu Rev Physiol. 2017;79:237–60.PubMedCrossRef
62.
go back to reference Zhang M, Su YQ, Sugiura K, Xia G, Eppig JJ. Granulosa cell ligand NPPC and its receptor NPR2 maintain meiotic arrest in mouse oocytes. Science. 2010;330:366–9.PubMedPubMedCentralCrossRef Zhang M, Su YQ, Sugiura K, Xia G, Eppig JJ. Granulosa cell ligand NPPC and its receptor NPR2 maintain meiotic arrest in mouse oocytes. Science. 2010;330:366–9.PubMedPubMedCentralCrossRef
63.
go back to reference Saulnier PJ, Roussel R, Halimi JM, Lebrec J, Dardari D, Maimaitiming S, Guilloteau G, Prugnard X, Marechaud R, Ragot S, Marre M, Hadjadj S, Surdiagene DN, Groups Ds. Impact of natriuretic peptide clearance receptor (NPR3) gene variants on blood pressure in type 2 diabetes. Diabetes Care. 2011;34:1199–204.PubMedPubMedCentralCrossRef Saulnier PJ, Roussel R, Halimi JM, Lebrec J, Dardari D, Maimaitiming S, Guilloteau G, Prugnard X, Marechaud R, Ragot S, Marre M, Hadjadj S, Surdiagene DN, Groups Ds. Impact of natriuretic peptide clearance receptor (NPR3) gene variants on blood pressure in type 2 diabetes. Diabetes Care. 2011;34:1199–204.PubMedPubMedCentralCrossRef
64.
go back to reference Hiradate Y, Hoshino Y, Tanemura K, Sato E. C-type natriuretic peptide inhibits porcine oocyte meiotic resumption. Zygote. 2014;22:372–7.PubMedCrossRef Hiradate Y, Hoshino Y, Tanemura K, Sato E. C-type natriuretic peptide inhibits porcine oocyte meiotic resumption. Zygote. 2014;22:372–7.PubMedCrossRef
65.
go back to reference Kawamura K, Cheng Y, Kawamura N, Takae S, Okada A, Kawagoe Y, Mulders S, Terada Y, Hsueh AJ. 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.PubMedCrossRef Kawamura K, Cheng Y, Kawamura N, Takae S, Okada A, Kawagoe Y, Mulders S, Terada Y, Hsueh AJ. 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.PubMedCrossRef
66.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
67.
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.PubMedCrossRef 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.PubMedCrossRef
68.
go back to reference Zhang Y, Wang H, Liu W, Yang Y, Wang X, Zhang Z, Guo Q, Wang C, Xia G. Natriuretic peptides improve the developmental competence of in vitro cultured porcine oocytes. Reprod Biol Endocrinol. 2017;15:41.PubMedPubMedCentralCrossRef Zhang Y, Wang H, Liu W, Yang Y, Wang X, Zhang Z, Guo Q, Wang C, Xia G. Natriuretic peptides improve the developmental competence of in vitro cultured porcine oocytes. Reprod Biol Endocrinol. 2017;15:41.PubMedPubMedCentralCrossRef
69.
go back to reference Ogawa Y, Itoh H, Yoshitake Y, Inoue M, Yoshimasa T, Serikawa T, Nakao K. Molecular cloning and chromosomal assignment of the mouse C-type natriuretic peptide (CNP) gene (Nppc): comparison with the human CNP gene (NPPC). Genomics. 1994;24:383–7.PubMedCrossRef Ogawa Y, Itoh H, Yoshitake Y, Inoue M, Yoshimasa T, Serikawa T, Nakao K. Molecular cloning and chromosomal assignment of the mouse C-type natriuretic peptide (CNP) gene (Nppc): comparison with the human CNP gene (NPPC). Genomics. 1994;24:383–7.PubMedCrossRef
70.
go back to reference Zhang Y, Hao X, Xiang X, Wei K, Xia G, Zhang M. Porcine natriuretic peptide type B (pNPPB) maintains mouse oocyte meiotic arrest via natriuretic peptide receptor 2 (NPR2) in cumulus cells. Mol Reprod Dev. 2014;81:462–9.PubMedCrossRef Zhang Y, Hao X, Xiang X, Wei K, Xia G, Zhang M. Porcine natriuretic peptide type B (pNPPB) maintains mouse oocyte meiotic arrest via natriuretic peptide receptor 2 (NPR2) in cumulus cells. Mol Reprod Dev. 2014;81:462–9.PubMedCrossRef
71.
go back to reference De Cesaro MP, Macedo MP, Santos JT, Rosa PR, Ludke CA, Rissi VB, Gasperin BG, Goncalves PB. Natriuretic peptides stimulate oocyte meiotic resumption in bovine. Anim Reprod Sci. 2015;159:52–9.PubMedCrossRef De Cesaro MP, Macedo MP, Santos JT, Rosa PR, Ludke CA, Rissi VB, Gasperin BG, Goncalves PB. Natriuretic peptides stimulate oocyte meiotic resumption in bovine. Anim Reprod Sci. 2015;159:52–9.PubMedCrossRef
72.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
73.
go back to reference Franciosi F, Coticchio G, Lodde V, Tessaro I, Modina SC, Fadini R, Dal Canto M, Renzini MM, Albertini DF, Luciano AM. Natriuretic peptide precursor C delays meiotic resumption and sustains gap junction-mediated communication in bovine cumulus-enclosed oocytes. Biol Reprod. 2014;91:61.PubMedCrossRef Franciosi F, Coticchio G, Lodde V, Tessaro I, Modina SC, Fadini R, Dal Canto M, Renzini MM, Albertini DF, Luciano AM. Natriuretic peptide precursor C delays meiotic resumption and sustains gap junction-mediated communication in bovine cumulus-enclosed oocytes. Biol Reprod. 2014;91:61.PubMedCrossRef
74.
go back to reference Xi G, An L, Jia Z, Tan K, Zhang J, Wang Z, Zhang C, Miao K, Wu Z, Tian J. Natriuretic peptide receptor 2 (NPR2) localized in bovine oocyte underlies a unique mechanism for C-type natriuretic peptide (CNP)-induced meiotic arrest. Theriogenology. 2018;106:198–209.PubMedCrossRef Xi G, An L, Jia Z, Tan K, Zhang J, Wang Z, Zhang C, Miao K, Wu Z, Tian J. Natriuretic peptide receptor 2 (NPR2) localized in bovine oocyte underlies a unique mechanism for C-type natriuretic peptide (CNP)-induced meiotic arrest. Theriogenology. 2018;106:198–209.PubMedCrossRef
75.
go back to reference Wang X, Wang H, Liu W, Zhang Z, Zhang Y, Zhang W, Chen Z, Xia G, Wang C. High level of C-type natriuretic peptide induced by hyperandrogen-mediated anovulation in polycystic ovary syndrome mice. Clin Sci (Lond). 2018;132:759–76.CrossRef Wang X, Wang H, Liu W, Zhang Z, Zhang Y, Zhang W, Chen Z, Xia G, Wang C. High level of C-type natriuretic peptide induced by hyperandrogen-mediated anovulation in polycystic ovary syndrome mice. Clin Sci (Lond). 2018;132:759–76.CrossRef
77.
go back to reference Downs SM, Eppig JJ. Induction of mouse oocyte maturation in vivo by perturbants of purine metabolism. Biol Reprod. 1987;36:431–7.PubMedCrossRef Downs SM, Eppig JJ. Induction of mouse oocyte maturation in vivo by perturbants of purine metabolism. Biol Reprod. 1987;36:431–7.PubMedCrossRef
78.
go back to reference Downs SM. Induction of meiotic maturation in vivo in the mouse by IMP dehydrogenase inhibitors: effects on the developmental capacity of ova. Mol Reprod Dev. 1994;38:293–302.PubMedCrossRef Downs SM. Induction of meiotic maturation in vivo in the mouse by IMP dehydrogenase inhibitors: effects on the developmental capacity of ova. Mol Reprod Dev. 1994;38:293–302.PubMedCrossRef
79.
go back to reference Wigglesworth K, Lee KB, O'Brien MJ, Peng J, Matzuk MM, Eppig JJ. Bidirectional communication between oocytes and ovarian follicular somatic cells is required for meiotic arrest of mammalian oocytes. P Natl Acad Sci USA. 2013;110:E3723–9.CrossRef Wigglesworth K, Lee KB, O'Brien MJ, Peng J, Matzuk MM, Eppig JJ. Bidirectional communication between oocytes and ovarian follicular somatic cells is required for meiotic arrest of mammalian oocytes. P Natl Acad Sci USA. 2013;110:E3723–9.CrossRef
80.
go back to reference Eppig JJ, Ward-Bailey PF, Coleman DL. Hypoxanthine and adenosine in murine ovarian follicular fluid: concentrations and activity in maintaining oocyte meiotic arrest. Biol Reprod. 1985;33:1041–9.PubMedCrossRef Eppig JJ, Ward-Bailey PF, Coleman DL. Hypoxanthine and adenosine in murine ovarian follicular fluid: concentrations and activity in maintaining oocyte meiotic arrest. Biol Reprod. 1985;33:1041–9.PubMedCrossRef
81.
go back to reference Downs SM, Coleman DL, Eppig JJ. Maintenance of murine oocyte meiotic arrest: uptake and metabolism of hypoxanthine and adenosine by cumulus cell-enclosed and denuded oocytes. Dev Biol. 1986;117:174–83.PubMedCrossRef Downs SM, Coleman DL, Eppig JJ. Maintenance of murine oocyte meiotic arrest: uptake and metabolism of hypoxanthine and adenosine by cumulus cell-enclosed and denuded oocytes. Dev Biol. 1986;117:174–83.PubMedCrossRef
82.
go back to reference Downs SM, Coleman DL, Ward-Bailey PF, Eppig JJ. Hypoxanthine is the principal inhibitor of murine oocyte maturation in a low molecular weight fraction of porcine follicular fluid. Proc Natl Acad Sci U S A. 1985;82:454–8.PubMedPubMedCentralCrossRef Downs SM, Coleman DL, Ward-Bailey PF, Eppig JJ. Hypoxanthine is the principal inhibitor of murine oocyte maturation in a low molecular weight fraction of porcine follicular fluid. Proc Natl Acad Sci U S A. 1985;82:454–8.PubMedPubMedCentralCrossRef
83.
go back to reference Russwurm M, Mullershausen F, Friebe A, Jager R, Russwurm C, Koesling D. Design of fluorescence resonance energy transfer (FRET)-based cGMP indicators: a systematic approach. Biochem J. 2007;407:69–77.PubMedPubMedCentralCrossRef Russwurm M, Mullershausen F, Friebe A, Jager R, Russwurm C, Koesling D. Design of fluorescence resonance energy transfer (FRET)-based cGMP indicators: a systematic approach. Biochem J. 2007;407:69–77.PubMedPubMedCentralCrossRef
84.
go back to reference Norris RP, Ratzan WJ, Freudzon M, Mehlmann LM, Krall J, Movsesian MA, Wang H, Ke H, Nikolaev VO, Jaffe LA. Cyclic GMP from the surrounding somatic cells regulates cyclic AMP and meiosis in the mouse oocyte. Development. 2009;136:1869–78.PubMedPubMedCentralCrossRef Norris RP, Ratzan WJ, Freudzon M, Mehlmann LM, Krall J, Movsesian MA, Wang H, Ke H, Nikolaev VO, Jaffe LA. Cyclic GMP from the surrounding somatic cells regulates cyclic AMP and meiosis in the mouse oocyte. Development. 2009;136:1869–78.PubMedPubMedCentralCrossRef
85.
go back to reference Downs SM, Daniel SA, Bornslaeger EA, Hoppe PC, Eppig JJ. Maintenance of meiotic arrest in mouse oocytes by purines: modulation of cAMP levels and cAMP phosphodiesterase activity. Gamete research. 1989;23:323–34.PubMedCrossRef Downs SM, Daniel SA, Bornslaeger EA, Hoppe PC, Eppig JJ. Maintenance of meiotic arrest in mouse oocytes by purines: modulation of cAMP levels and cAMP phosphodiesterase activity. Gamete research. 1989;23:323–34.PubMedCrossRef
86.
go back to reference Celik O, Celik N, Gungor S, Haberal ET, Aydin S. Selective regulation of oocyte meiotic events enhances Progress in fertility preservation methods. Biochem Insights. 2015;8:11–21.PubMedPubMedCentralCrossRef Celik O, Celik N, Gungor S, Haberal ET, Aydin S. Selective regulation of oocyte meiotic events enhances Progress in fertility preservation methods. Biochem Insights. 2015;8:11–21.PubMedPubMedCentralCrossRef
87.
go back to reference Ponsioen B, Zhao J, Riedl J, Zwartkruis F, van der Krogt G, Zaccolo M, Moolenaar WH, Bos JL, Jalink K. Detecting cAMP-induced Epac activation by fluorescence resonance energy transfer: Epac as a novel cAMP indicator. EMBO Rep. 2004;5:1176–80.PubMedPubMedCentralCrossRef Ponsioen B, Zhao J, Riedl J, Zwartkruis F, van der Krogt G, Zaccolo M, Moolenaar WH, Bos JL, Jalink K. Detecting cAMP-induced Epac activation by fluorescence resonance energy transfer: Epac as a novel cAMP indicator. EMBO Rep. 2004;5:1176–80.PubMedPubMedCentralCrossRef
89.
go back to reference Young JM, McNeilly AS. Theca: the forgotten cell of the ovarian follicle. Reproduction. 2010;140:489–504.PubMedCrossRef Young JM, McNeilly AS. Theca: the forgotten cell of the ovarian follicle. Reproduction. 2010;140:489–504.PubMedCrossRef
90.
go back to reference Robinson JW, Zhang M, Shuhaibar LC, Norris RP, Geerts A, Wunder F, Eppig JJ, Potter LR, Jaffe LA. 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.PubMedPubMedCentralCrossRef Robinson JW, Zhang M, Shuhaibar LC, Norris RP, Geerts A, Wunder F, Eppig JJ, Potter LR, Jaffe LA. 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.PubMedPubMedCentralCrossRef
91.
go back to reference Lee KB, Zhang M, Sugiura K, Wigglesworth K, Uliasz T, Jaffe LA, Eppig JJ. Hormonal coordination of natriuretic peptide type C and natriuretic peptide receptor 3 expression in mouse granulosa cells. Biol Reprod. 2013;88:42.PubMed Lee KB, Zhang M, Sugiura K, Wigglesworth K, Uliasz T, Jaffe LA, Eppig JJ. Hormonal coordination of natriuretic peptide type C and natriuretic peptide receptor 3 expression in mouse granulosa cells. Biol Reprod. 2013;88:42.PubMed
92.
go back to reference Wang Y, Kong N, Li N, Hao X, Wei K, Xiang X, Xia G, Zhang M. 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.PubMedCrossRef Wang Y, Kong N, Li N, Hao X, Wei K, Xiang X, Xia G, Zhang M. 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.PubMedCrossRef
93.
go back to reference Hsieh M, Lee D, Panigone S, Homer K, Chen R, Theologis A, Lee DC, Threadgill DW, Conti M. Luteinizing hormone-dependent activation of the epidermal growth factor network is essential for ovulation. Mol Cell Biol. 2007;27:1914–24.PubMedCrossRef Hsieh M, Lee D, Panigone S, Homer K, Chen R, Theologis A, Lee DC, Threadgill DW, Conti M. Luteinizing hormone-dependent activation of the epidermal growth factor network is essential for ovulation. Mol Cell Biol. 2007;27:1914–24.PubMedCrossRef
94.
go back to reference Park JY, Su YQ, Ariga M, Law E, Jin SLC, Conti M. EGF-like growth factors as mediators of LH action in the ovulatory follicle. Science. 2004;303:682–4.CrossRef Park JY, Su YQ, Ariga M, Law E, Jin SLC, Conti M. EGF-like growth factors as mediators of LH action in the ovulatory follicle. Science. 2004;303:682–4.CrossRef
95.
go back to reference K Pahwa A, Arbab-Zadeh A, J Brotman D, S Feldman L. Potential role of coronary computed tomography-angiography for guiding perioperative cardiac management for non-cardiac surgery. Heart Int. 2013;8:e1.PubMedPubMedCentralCrossRef K Pahwa A, Arbab-Zadeh A, J Brotman D, S Feldman L. Potential role of coronary computed tomography-angiography for guiding perioperative cardiac management for non-cardiac surgery. Heart Int. 2013;8:e1.PubMedPubMedCentralCrossRef
96.
go back to reference Egbert JR, Shuhaibar LC, Edmund AB, Van Helden DA, Robinson JW, Uliasz TF, Baena V, Geerts A, Wunder F, Potter LR. 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.PubMedPubMedCentralCrossRef Egbert JR, Shuhaibar LC, Edmund AB, Van Helden DA, Robinson JW, Uliasz TF, Baena V, Geerts A, Wunder F, Potter LR. 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.PubMedPubMedCentralCrossRef
97.
go back to reference Norris RP, Freudzon M, Mehlmann LM, Cowan AE, Simon AM, Paul DL, Lampe PD, Jaffe LA. Luteinizing hormone causes MAP kinase-dependent phosphorylation and closure of connexin 43 gap junctions in mouse ovarian follicles: one of two paths to meiotic resumption. Development. 2008;135:3229–38.PubMedPubMedCentralCrossRef Norris RP, Freudzon M, Mehlmann LM, Cowan AE, Simon AM, Paul DL, Lampe PD, Jaffe LA. Luteinizing hormone causes MAP kinase-dependent phosphorylation and closure of connexin 43 gap junctions in mouse ovarian follicles: one of two paths to meiotic resumption. Development. 2008;135:3229–38.PubMedPubMedCentralCrossRef
98.
go back to reference Shuhaibar LC, Egbert JR, Edmund AB, Uliasz TF, Dickey DM, Yee SP, Potter LR, Jaffe LA. Dephosphorylation of juxtamembrane serines and threonines of the NPR2 guanylyl cyclase is required for rapid resumption of oocyte meiosis in response to luteinizing hormone. Dev Biol. 2016;409:194–201.PubMedCrossRef Shuhaibar LC, Egbert JR, Edmund AB, Uliasz TF, Dickey DM, Yee SP, Potter LR, Jaffe LA. Dephosphorylation of juxtamembrane serines and threonines of the NPR2 guanylyl cyclase is required for rapid resumption of oocyte meiosis in response to luteinizing hormone. Dev Biol. 2016;409:194–201.PubMedCrossRef
99.
go back to reference Egbert JR, Uliasz TF, Shuhaibar LC, Geerts A, Wunder F, Kleiman RJ, Humphrey JM, Lampe PD, Artemyev NO, Rybalkin SD, Beavo JA, Movsesian MA, Jaffe LA. Luteinizing hormone causes phosphorylation and activation of the cGMP phosphodiesterase PDE5 in rat ovarian follicles, contributing, together with PDE1 activity, to the resumption of meiosis. Biol Reprod. 2016;94:110.PubMedPubMedCentralCrossRef Egbert JR, Uliasz TF, Shuhaibar LC, Geerts A, Wunder F, Kleiman RJ, Humphrey JM, Lampe PD, Artemyev NO, Rybalkin SD, Beavo JA, Movsesian MA, Jaffe LA. Luteinizing hormone causes phosphorylation and activation of the cGMP phosphodiesterase PDE5 in rat ovarian follicles, contributing, together with PDE1 activity, to the resumption of meiosis. Biol Reprod. 2016;94:110.PubMedPubMedCentralCrossRef
100.
go back to reference Egbert JR, Yee S-P, Jaffe LA. Luteinizing hormone signaling phosphorylates and activates the cyclic GMP phosphodiesterase PDE5 in mouse ovarian follicles, contributing an additional component to the hormonally induced decrease in cyclic GMP that reinitiates meiosis. Dev Biol. 2018;435:6–14.PubMedCrossRef Egbert JR, Yee S-P, Jaffe LA. Luteinizing hormone signaling phosphorylates and activates the cyclic GMP phosphodiesterase PDE5 in mouse ovarian follicles, contributing an additional component to the hormonally induced decrease in cyclic GMP that reinitiates meiosis. Dev Biol. 2018;435:6–14.PubMedCrossRef
101.
go back to reference Sasseville M, Cote N, Gagnon MC, Richard FJ. Up-regulation of 3′5'-cyclic guanosine monophosphate-specific phosphodiesterase in the porcine cumulus-oocyte complex affects steroidogenesis during in vitro maturation. Endocrinology. 2008;149:5568–76.PubMedCrossRef Sasseville M, Cote N, Gagnon MC, Richard FJ. Up-regulation of 3′5'-cyclic guanosine monophosphate-specific phosphodiesterase in the porcine cumulus-oocyte complex affects steroidogenesis during in vitro maturation. Endocrinology. 2008;149:5568–76.PubMedCrossRef
102.
go back to reference Bergeron A, Guillemette C, Sirard M-A, Richard FJ. Active 3′–5′ cyclic nucleotide phosphodiesterases are present in detergent-resistant membranes of mural granulosa cells. Reprod Fertil Dev. 2017;29:778–90.PubMedCrossRef Bergeron A, Guillemette C, Sirard M-A, Richard FJ. Active 3′–5′ cyclic nucleotide phosphodiesterases are present in detergent-resistant membranes of mural granulosa cells. Reprod Fertil Dev. 2017;29:778–90.PubMedCrossRef
103.
go back to reference Liu W, Xin Q, Wang X, Wang S, Wang H, Zhang W, Yang Y, Zhang Y, Zhang Z, Wang C, Xu Y, Duan E, Xia G. Estrogen receptors in granulosa cells govern meiotic resumption of pre-ovulatory oocytes in mammals. Cell Death Dis. 2017;8:e2662.PubMedPubMedCentralCrossRef Liu W, Xin Q, Wang X, Wang S, Wang H, Zhang W, Yang Y, Zhang Y, Zhang Z, Wang C, Xu Y, Duan E, Xia G. Estrogen receptors in granulosa cells govern meiotic resumption of pre-ovulatory oocytes in mammals. Cell Death Dis. 2017;8:e2662.PubMedPubMedCentralCrossRef
104.
go back to reference Hambleton R, Krall J, Tikishvili E, Honeggar M, Ahmad F, Manganiello VC, Movsesian MA. Isoforms of cyclic nucleotide phosphodiesterase PDE3 and their contribution to cAMP hydrolytic activity in subcellular fractions of human myocardium. J Biol Chem. 2005;280:39168–74.PubMedCrossRef Hambleton R, Krall J, Tikishvili E, Honeggar M, Ahmad F, Manganiello VC, Movsesian MA. Isoforms of cyclic nucleotide phosphodiesterase PDE3 and their contribution to cAMP hydrolytic activity in subcellular fractions of human myocardium. J Biol Chem. 2005;280:39168–74.PubMedCrossRef
105.
go back to reference Wigglesworth K, Lee K-B, O’Brien MJ, Peng J, Matzuk MM, Eppig JJ. Bidirectional communication between oocytes and ovarian follicular somatic cells is required for meiotic arrest of mammalian oocytes. Proc Natl Acad Sci. 2013;110:E3723–9.PubMedCrossRef Wigglesworth K, Lee K-B, O’Brien MJ, Peng J, Matzuk MM, Eppig JJ. Bidirectional communication between oocytes and ovarian follicular somatic cells is required for meiotic arrest of mammalian oocytes. Proc Natl Acad Sci. 2013;110:E3723–9.PubMedCrossRef
106.
go back to reference Hao X, Wang Y, Kong N, Zhang Y, Zhao Y, Xia G, Zhang M. Epidermal growth factor-mobilized intracellular calcium of cumulus cells decreases natriuretic peptide receptor 2 affinity for natriuretic peptide type C and induces oocyte meiotic resumption in the mouse. Biol Reprod. 2016;95:45.PubMedCrossRef Hao X, Wang Y, Kong N, Zhang Y, Zhao Y, Xia G, Zhang M. Epidermal growth factor-mobilized intracellular calcium of cumulus cells decreases natriuretic peptide receptor 2 affinity for natriuretic peptide type C and induces oocyte meiotic resumption in the mouse. Biol Reprod. 2016;95:45.PubMedCrossRef
108.
go back to reference Kim SM, Kim SH, Cho KW, Young S. Expression of C-type Natriuretic Peptide and its Specific Guanylyl Cyclase-Coupled Receptor in Pig Ovarian Granulosa Cells; 2018. Kim SM, Kim SH, Cho KW, Young S. Expression of C-type Natriuretic Peptide and its Specific Guanylyl Cyclase-Coupled Receptor in Pig Ovarian Granulosa Cells; 2018.
110.
go back to reference Tamura N, Doolittle LK, Hammer RE, Shelton JM, Richardson JA, Garbers DL. Critical roles of the guanylyl cyclase B receptor in endochondral ossification and development of female reproductive organs. Proc Natl Acad Sci U S A. 2004;101:17300–5.PubMedPubMedCentralCrossRef Tamura N, Doolittle LK, Hammer RE, Shelton JM, Richardson JA, Garbers DL. Critical roles of the guanylyl cyclase B receptor in endochondral ossification and development of female reproductive organs. Proc Natl Acad Sci U S A. 2004;101:17300–5.PubMedPubMedCentralCrossRef
111.
go back to reference Geister KA, Brinkmeier ML, Hsieh M, Faust SM, Karolyi IJ, Perosky JE, Kozloff KM, Conti M, Camper SA. A novel loss-of-function mutation in Npr2 clarifies primary role in female reproduction and reveals a potential therapy for acromesomelic dysplasia, Maroteaux type. Hum Mol Genet. 2013;22:345–57.PubMedCrossRef Geister KA, Brinkmeier ML, Hsieh M, Faust SM, Karolyi IJ, Perosky JE, Kozloff KM, Conti M, Camper SA. A novel loss-of-function mutation in Npr2 clarifies primary role in female reproduction and reveals a potential therapy for acromesomelic dysplasia, Maroteaux type. Hum Mol Genet. 2013;22:345–57.PubMedCrossRef
112.
go back to reference Kim SH, Cho KW, Lim SH, Hwang YH, Ryu H, Oh SH, Seul KH, Jeong GB, Yoon S. Presence and release of immunoreactive atrial natriuretic peptide in granulosa cells of the pig ovarian follicle. Regul Pept. 1992;42:153–62.PubMedCrossRef Kim SH, Cho KW, Lim SH, Hwang YH, Ryu H, Oh SH, Seul KH, Jeong GB, Yoon S. Presence and release of immunoreactive atrial natriuretic peptide in granulosa cells of the pig ovarian follicle. Regul Pept. 1992;42:153–62.PubMedCrossRef
113.
go back to reference Peng JY, Xin HY, Han P, Zhao HB, Bai L, An XP, Cao BY. Identification and gene expression analyses of natriuretic peptide system in the ovary of goat (Capra hircus). Gene. 2013;524:105–13.PubMedCrossRef Peng JY, Xin HY, Han P, Zhao HB, Bai L, An XP, Cao BY. Identification and gene expression analyses of natriuretic peptide system in the ovary of goat (Capra hircus). Gene. 2013;524:105–13.PubMedCrossRef
114.
go back to reference Vollmar AM, Mytzka C, Arendt RM, Schulz R. Atrial natriuretic peptide in bovine corpus luteum. Endocrinology. 1988;123:762–7.PubMedCrossRef Vollmar AM, Mytzka C, Arendt RM, Schulz R. Atrial natriuretic peptide in bovine corpus luteum. Endocrinology. 1988;123:762–7.PubMedCrossRef
Metadata
Title
The art of oocyte meiotic arrest regulation
Authors
Bo Pan
Julang Li
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Reproductive Biology and Endocrinology / Issue 1/2019
Electronic ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-018-0445-8

Other articles of this Issue 1/2019

Reproductive Biology and Endocrinology 1/2019 Go to the issue