Hostname: page-component-76fb5796d-2lccl Total loading time: 0 Render date: 2024-04-26T23:30:03.730Z Has data issue: false hasContentIssue false

Supplementation of c-type natriuretic peptide during in vitro growth period benefits the development of murine preantral follicles

Published online by Cambridge University Press:  25 November 2020

Li Ang
Affiliation:
Department of Biochemistry, Shanxi Medical University, Taiyuan030001, China
Cao Haixia
Affiliation:
Shanxi Provincial Key Laboratory of Cell Regeneration and Birth Defects, Taiyuan, 030001, China
Li Hongxia
Affiliation:
Shanxi Provincial Key Laboratory of Cell Regeneration and Birth Defects, Taiyuan, 030001, China
Li Ruijiao
Affiliation:
Shanxi Provincial Key Laboratory of Cell Regeneration and Birth Defects, Taiyuan, 030001, China
Guo Xingping
Affiliation:
Shanxi Provincial Key Laboratory of Cell Regeneration and Birth Defects, Taiyuan, 030001, China
Wang Huaixiu*
Affiliation:
Shanxi Provincial Key Laboratory of Cell Regeneration and Birth Defects, Taiyuan, 030001, China Beijing Perfect Family Hospital, Beijing, 100034, China
*
Author for correspondence: Wang Huaixiu. Shanxi Provincial Key Laboratory of Cell Regeneration and Birth Defects, Taiyuan, 030001, China. E-mail: 976378008@qq.com

Summary

The present study investigated the effects of c-type natriuretic peptide (CNP) on the development of murine preantral follicles during in vitro growth (IVG). Preantral follicles isolated from ovaries of Kunming mice were cultured in vitro. In the culture system, CNP was supplemented in the experimental groups and omitted in the control groups. In Experiment 1, CNP was only supplemented at the early stage and follicle development was evaluated. In Experiments 2 and 3, CNP was supplemented during the whole period of in vitro culture. In Experiment 2, follicle development and oocyte maturity were evaluated. In Experiment 3, follicle development and embryo cleavage after in vitro fertilization (IVF) were assessed. The results showed that in the control groups in all three experiments, granulosa cells migrated from within the follicle and the follicles could not reach the antral stage. In the experimental groups in all three experiments, no migration of granulosa cells was observed and follicle development was assessed as attaining the antral stage, which was significantly superior to that of the control group (P < 0.0001). Oocyte meiotic arrest was effectively maintained, hence giving good developmental competence. In conclusion, CNP supplementation in the culture system during IVG benefited the development of murine preantral follicles.

Type
Research Article
Copyright
© The Author(s), 2020. Published by Cambridge University Press

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Alexander, B, Coppola, G, DiBerardino, D, Rho, GJ, StJohn, E, Betts, DH and King, WA (2006). The effect of 6-dimethylaminopurine (6-DMAP) and cycloheximide (CHX) on the development and chromosomal complement of sheep parthenogenetic and nuclear transfer embryos. Mol Reprod Dev 73, 2030.CrossRefGoogle ScholarPubMed
Appeltant, R, Somfai, T, Maes, D, Van, S and Kikuchi, K (2016). Porcine oocyte maturation in vitro: role of CAMP and oocyte-secreted factors-a practical approach. J Reprod Dev 62, 439–49.CrossRefGoogle ScholarPubMed
Bouhelan, R, Bockaert, J, Mermet-Bauvier, R, Guillon, G and Homburger, V (1987). Heavy isotope labeling study of the turnover of forskolin-stimulated adenylate-cyclase in bc3h1 cell-line. J Biol Chem 262, 8470–5.CrossRefGoogle Scholar
Brito, IR, Lima, IMT, Xu, M, Shea, LD, Woodruff, TK and Figueiredo, JR (2014). Three-dimensional systems for in vitro follicular culture: overview of alginate-based matrices. Reprod Fertil. Dev 26, 915–30.CrossRefGoogle ScholarPubMed
Cheng, Y, Fan, HY, Wen, DC, Tong, C, Zhu, ZY, Lei, L, Sun, QY and Chen, DY (2003). Asynchronous cytoplast and karyoplast transplantation reveals that the cytoplasm determines the developmental fate of the nucleus in mouse oocytes. Mol Reprod Dev 65, 278–82.CrossRefGoogle ScholarPubMed
Choi, JK, Agarwal P an He X (2013). In vitro culture of early secondary preantral follicles in hanging drop of ovarian cell-conditioned medium to obtain MII oocytes from out-bred deer mice. Tissue Eng Part A 19, 12626–37.CrossRefGoogle Scholar
Eppig, JJ and O’Brien, MJ (1996). Development in vitro of mouse oocytes from primordial follicles. Biol Reprod 54, 197207.CrossRefGoogle ScholarPubMed
Eppig, JJ and Schroeder, AC (1977). Mouse oocyte development in vitro with various culture systems. Dev Biol 60, 371–88.CrossRefGoogle ScholarPubMed
Eppig, JJ and Schroeder, AC (1989). Capacity of mouse oocytes from preantral follicles to undergo embryogenesis and development to live young after growth maturation and fertilization in vitro . Biol Reprod 41, 268–76.CrossRefGoogle ScholarPubMed
Follin-Arbelet, V, Misund, K, Hallan, NE, Ugland, H, Sundan, A and Kiil, BH (2015). The natural compound forskolin synergized with dexamethasone to induce cell death in myeloma cells via BIM. Scirep-UK 5, 13001.Google Scholar
Franciosi, F, Coticchio, G, Lodda, V, Tessaro, I, Modina, SC, Fadini, R, Dal, C, Mariabeatrice, , Mignini, RM, Albertini, DF and Luciano, AM (2014). Natriuretic peptide precursor c delays meiotic resumptions and sustains gap junction-mediated communication in bovine cumulus-enclosed oocytes. Biol Reprod 91, 61.CrossRefGoogle Scholar
Higuchi, CM, Maeda, Y, Horiuchi, T and Yamazaki, Y (2015). A simplified method for three-dimensional (3-D) ovarian tissue culture yielding oocytes competent to produce full-term offspring in mice. PLoS One 10, e0143114.CrossRefGoogle ScholarPubMed
Hiradate, Y, Hosshino, Y, Tanemura, K and Sato, E (2013). C-type natriuretic peptide inhibits porcine oocyte meiotic resumption. Zygote 22, 372–7.CrossRefGoogle ScholarPubMed
Hsueh, AJW, Kawamura, K, Cheng, Y and Fauser, BCJM (2015). Intraovarian control of early folliculogenesis. Endocr Rev 36, 124.CrossRefGoogle ScholarPubMed
Jin, SY, Lei, L, Shikanov, A, Shea, LD and Woodruff, TK (2010). A novel two-step strategy for in vitro culture of early stage ovarian follicles in the mouse. Fertil Steril 93, 2633–9.CrossRefGoogle ScholarPubMed
Kreegar, PK, Deck, JW, Woodruff, TK and Shea, LD (2016). The in vitro regulation of ovarian follicle development using alginate extracellular matrix gels. Biomaterials 27, 714–23.CrossRefGoogle Scholar
Machaty, Z, Miller, AR and Zhang, L (2017). Egg activation at fertilization. Adv Exp Med Biol 953, 147.CrossRefGoogle ScholarPubMed
Mainigi, MA, Ord, T and Schultz, RM (2011). Meiotic and developmental competence in mice are compromised following follicle development in vitro using an alginate-based culture system. Biol Reprod 85, 269–76.CrossRefGoogle ScholarPubMed
Mochida, N and Akatani-Hasegawa, A (2013). Live births from isolated primary/early secondary follicles following a multistep culture without organ culture in mice. Reproduction 146, 3747.CrossRefGoogle ScholarPubMed
O’Brien, MJ, Pendola, JK and Eppig, JJ (2003). A revised protocol for in vitro development of mouse oocytes from primordial follicles dramatically improves their developmental competence. Biol Reprod 68, 1682–6.CrossRefGoogle ScholarPubMed
Romero, S, Sanchez, F, Lolicato, F, Van, RH and Smitz, J (2016). Immature oocytes from unprimed juvenile mice become a valuable source for embryo production when using c-type natriuretic peptide as essential component of culture medium. Biol Reprod 95, 6473.CrossRefGoogle ScholarPubMed
Samake, S and Smith, LC (1997). Synchronization of cell division in eight-cell bovine embryos produced in vitro: effects of 6-dimethylaminopurine. J Reprod Fertil 110, 21–7.CrossRefGoogle ScholarPubMed
Sato, Y, Cheng, Y, Kawamura, K, Takae, S and Hsueh, AJW (2012). C-type natriuretic peptide stimulates ovarian follicle development. Mol Endocrinol 26, 1158–66.CrossRefGoogle ScholarPubMed
Shikanov, A, Xu, M, Woodruff, TK and Shea, LD (2009). Interpenetrating fibrin-alginate matrices for in vitro ovarian follicle development. Biomaterials 30, 5476–85.CrossRefGoogle ScholarPubMed
Simli, M, Pellerano, P, Pignllo, S, Tavosanis, G, Ottaggio, L, de Saint-Georges, L and Bonatti, S (1997). 6-DMAP inhibition of early cell cycle events and induction of mitotic abnormalities. Mutagenesis 12, 313–9.CrossRefGoogle Scholar
Telfer, EE and Zelinski, MB (2013). Ovarian follicle culture: advances and challenges for human and nonhuman primates. Fertil. Steril 99, 1523–33.CrossRefGoogle ScholarPubMed
Tiwari, M, Prasad, S, Tripathi, A, Pandey, AN, Ali, I, Singh, AK, Shrivastav, TG and Chaube, SK (2015). Apoptosis in mammalian oocytes: a review. Apoptosis 20, 1019–25.CrossRefGoogle ScholarPubMed
Wang, JJ, Ge, W, Liu, JC, Klinger, FG, Dyce, PW, De, FM and Shen, W (2017). Complete in vitro oogenesis: retrospects and prospects. Cell Death Differ 24, 1845–52.CrossRefGoogle ScholarPubMed
Wei, Q, Zhou, C, Yuan, M, Miao, YY, Zhao, XE and Ma, BH (2017). Effect of c-type natriuretic peptide on maturation and developmental competence of immature mouse oocytes in vitro . Reprod Fertil Dev 29, 319–24.CrossRefGoogle ScholarPubMed
Wycheley, G, Downey, D, Kane, MT and Hynes, AC (2004). A novel follicle culture system markedly increases follicle volume, cell number and oestradiol secretion. Reproduction 127, 669–77.CrossRefGoogle Scholar
Xi, GY, Wang, WJ, Sarfaraz, AF and Yao, FS (2019). C-type natriuretic peptide enhances mouse preantral follicle growth. Reproduction 157, 445–55.CrossRefGoogle ScholarPubMed
Xu, M, Kreeger, PK, Shea, LD and Woodruff, TK (2006). Tissue-engineered follicles produce live, fertile offspring. Tissue Eng 12, 2739–46.CrossRefGoogle ScholarPubMed
Yang, L, Wei, Q, Li, W, Ge, JB, Zhao, XE and Ma, BH (2016). C-type natriuretic peptide improved vitrified-warmed mouse cumulus–oocyte complexes developmental competence. Cryobiology 72, 161–4.CrossRefGoogle ScholarPubMed
Zhang, MJ, Su, YQ, Sugiura, K, Xia, GL and Eppig, JJ (2010). Granulosa cell ligand NPPC and its receptor NPR2 maintain meiotic arrest in mouse oocytes. Science 330, 366–9.CrossRefGoogle ScholarPubMed
Zhang, T, Zhang, CQ, Fan, XM, Li, RL and Zhang, TX (2017a). Effect of c-type natriuretic peptide pretreatment on in vitro bovine maturation. In Vitro Cell Dev Biol 53, 199206.CrossRefGoogle ScholarPubMed
Zhang, YH, Wang, HR, Liu, W, Yang, Y, Wang, X, Zhang, ZY, Guo, QR, Wang, C and Xia, GL (2017b). Natriuretic peptides improve the developmental competence of in vitro cultured porcine oocytes. Reprod Biol Endocrinol 15, 4152.CrossRefGoogle ScholarPubMed
Zhong, T, Fan, XM, Li, RL, Zhang, CQ and Zhang, JX (2018) Effects of pre-incubation with c-type natriuretic peptide on nuclear maturation, mitochondrial behavior and developmental competence of sheep oocytes. Oogenesis 497, 200–6.Google Scholar