Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
RESEARCH ARTICLE

Effects of brain-derived neurotrophic factor on oocyte maturation and embryonic development in a rat model of polycystic ovary syndrome

Qiaoli Zhang A , Dong Liu B , Meiling Zhang A , Na Li C , Shulan Lu D , Yanzhi Du A F and Zi-Jiang Chen A E
+ Author Affiliations
- Author Affiliations

A Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Lingshan Road 845, Shanghai 200135, China.

B Department of Obstetrics and Gynecology, The Second Hospital of Tianjin Medical University, Pingjiang Road 23, Tianjin 300211, China.

C Department of Clinical Laboratory, The First Affiliated Hospital of Xi’an Jiaotong University, Yanta West Road 277, Xi’an 710061, China.

D Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Yanta West Road 277, Xi’an 710061, China.

E National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Shandong Provincial Key Laboratory of Reproductive Medicine, Center for Reproductive Medicine, Shandong Provincial Hospital, Shandong University, Jingwu Road 324, Jinan 250021, China.

F Corresponding author. Email: yanzhidu@hotmail.com

Reproduction, Fertility and Development 28(12) 1904-1915 https://doi.org/10.1071/RD15131
Submitted: 5 April 2015  Accepted: 21 May 2015   Published: 25 June 2015

Abstract

Brain-derived neurotrophic factor (BDNF) is expressed extensively in the mammalian female reproductive system and has been implicated in the development of follicles and oocytes. However, BDNF expression patterns in the ovary and its effects on oocyte maturation and embryonic development in polycystic ovary syndrome (PCOS) have not been established. In the present study, we established a PCOS model by treating the rats with insulin and human chorionic gonadotropin (hCG). Rats treated with insulin + hCG had heavier bodyweight and ovarian weight, higher circulating concentrations of luteinising hormone (LH) and testosterone (T), and greater homeostatic model assessment of insulin resistance (HOMA-IR) values compared with control rats (P < 0.05). BDNF and its receptor tyrosine kinase type B (TrkB) were located in cyst walls, granulosa and theca cells, and BDNF protein levels were lower in ovaries of insulin + hCG-treated rats (P < 0.05). The rate of oocyte maturation and formation of blastocysts and morulae was greatest in rats treated with 5 ng mL–1 BDNF (P < 0.05) compared to other BDNF groups (1 and 10 ng mL–1) and the control. The control rats were also PCOS rats and were treated without BDNF. There were no significant differences in the rate of germinal vesicle breakdown (GVBD) and fertilisation among the various treatment groups (1, 5 and 10 ng mL–1) and the control group (P > 0.05). The results indicate that in vitro treatment with an appropriate concentration of BDNF not only promotes oocyte maturation, but also rescues embryonic development in rats treated with insulin + hCG as a model of PCOS.

Additional keywords: developmental potential, in vitro maturation, neurotrophin.


References

Adams, J., Polson, D. W., and Franks, S. (1986). Prevalence of polycystic ovaries in women with anovulation and idiopathic hirsutism. Br. Med. J. (Clin. Res. Ed.) 293, 355–359.
Prevalence of polycystic ovaries in women with anovulation and idiopathic hirsutism.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaL283ntFSitQ%3D%3D&md5=606daea49154fc00604da7b2bdbfe236CAS |

Anderson, R. A., Sciorio, R., Kinnell, H., Bayne, R. A., Thong, K. J., De Sousa, P. A., and Pickering, S. (2009). Cumulus gene expression as a predictor of human oocyte fertilisation, embryo development and competence to establish a pregnancy. Reproduction 138, 629–637.
Cumulus gene expression as a predictor of human oocyte fertilisation, embryo development and competence to establish a pregnancy.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhtlShur3L&md5=da490d576f4d2e7d0a4b1193182b44e6CAS | 19602522PubMed |

Anderson, R. A., Bayne, R. A., Gardner, J., and De Sousa, P. A. (2010). Brain-derived neurotrophic factor is a regulator of human oocyte maturation and early embryo development. Fertil. Steril. 93, 1394–1406.
Brain-derived neurotrophic factor is a regulator of human oocyte maturation and early embryo development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXkslyjt7Y%3D&md5=ea3052d26ec642ffb1f45e1a06489602CAS | 19463996PubMed |

Anger, D. L., Zhang, B., Boutross-Tadross, O., and Foster, W. G. (2007). Tyrosine receptor kinase B (TrkB) protein expression in the human endometrium. Endocrine 31, 167–173.
Tyrosine receptor kinase B (TrkB) protein expression in the human endometrium.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhtVSgtb%2FK&md5=4824547c4701050d90a1bb3e1fca6f94CAS | 17873329PubMed |

Azziz, R., Woods, K. S., Reyna, R., Key, T. J., Knochenhauer, E. S., and Yildiz, B. O. (2004). The prevalence and features of the polycystic ovary syndrome in an unselected population. J. Clin. Endocrinol. Metab. 89, 2745–2749.
The prevalence and features of the polycystic ovary syndrome in an unselected population.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXmtlGqurs%3D&md5=af95798510aa82b94f4ec08e6b585996CAS | 15181052PubMed |

Barde, Y. A., Edger, D., and Thoenen, H. (1982). Purification of a new neurotrophic factor from mammalian brain. EMBO J. 1, 549–553.
| 1:CAS:528:DyaL38XltVCgsLY%3D&md5=4f3165121556044254e48e813d0727f6CAS | 7188352PubMed |

Berga, S. L., and Yen, S. S. (1989). Opioidergic regulation of LH pulsatility in women with polycystic ovary syndrome. Clin. Endocrinol. (Oxf.) 30, 177–184.
Opioidergic regulation of LH pulsatility in women with polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK3c7isFWlsw%3D%3D&md5=9937a01f2abe841a283e21c67b6d1169CAS | 2532984PubMed |

Buyuk, E., and Seifer, D. B. (2008). Follicular-fluid neurotrophin levels in women undergoing assisted reproductive technology for different etiologies of infertility. Fertil. Steril. 90, 1611–1615.
Follicular-fluid neurotrophin levels in women undergoing assisted reproductive technology for different etiologies of infertility.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhsVOhur3K&md5=901405846aff721fd82e70c0741c21c7CAS | 18222435PubMed |

Buyuk, E., Santoro, N., Cohen, H. W., Charron, M. J., and Jindal, S. (2011). Reduced neurotrophin receptor tropomyosin-related kinase A expression in human granulosa cells: a novel marker of diminishing ovarian reserve. Fertil. Steril. 96, 474–478.e4.
Reduced neurotrophin receptor tropomyosin-related kinase A expression in human granulosa cells: a novel marker of diminishing ovarian reserve.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXptlygsLg%3D&md5=3f08a86df2a62cdaa7a06887855400fcCAS | 21645891PubMed |

Chang, R. J. (2004). A practical approach to the diagnosis of polycystic ovary syndrome. Am. J. Obstet. Gynecol. 191, 713–717.
A practical approach to the diagnosis of polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar | 15467530PubMed |

Chen, Z. J., Zhao, H., He, L., Shi, Y., Qin, Y., Shi, Y., Li, Z., You, L., Zhao, J., Liu, J., et al. (2011). Genome-wide association study identifies susceptibility loci for polycystic ovary syndrome on chromosome 2p16.3, 2p21 and 9q33.3. Nat. Genet. 43, 55–59.
Genome-wide association study identifies susceptibility loci for polycystic ovary syndrome on chromosome 2p16.3, 2p21 and 9q33.3.Crossref | GoogleScholarGoogle Scholar | 21151128PubMed |

Cheng, A., Coksaygan, T., Tang, H., Khatri, R., Balice-Gordon, R. J., Rao, M. S., and Mattson, M. P. (2007). Truncated tyrosine kinase B brain-derived neurotrophic factor receptor directs cortical neural stem cells to a glial cell fate by a novel signaling mechanism. J. Neurochem. 100, 1515–1530.
| 1:CAS:528:DC%2BD2sXjsF2rtrg%3D&md5=d8b86e886b194e610750a57f9ee5532fCAS | 17286628PubMed |

Craig, J., Zhu, H., Dyce, P. W., Petrik, J., and Li, J. (2004). Leptin enhances oocyte nuclear and cytoplasmic maturation via the mitogen-activated protein kinase pathway. Endocrinology 145, 5355–5363.
Leptin enhances oocyte nuclear and cytoplasmic maturation via the mitogen-activated protein kinase pathway.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXptVygs7o%3D&md5=31f1271681af53c16a23b78ddc4d0085CAS | 15284194PubMed |

De Sousa, P. A., Martins da Silva, S. J., and Anderson, R. A. (2004). Neurotrophin signaling in oocyte survival and developmental competence: a paradigm for cellular totipotency. Cloning Stem Cells 6, 375–385.
Neurotrophin signaling in oocyte survival and developmental competence: a paradigm for cellular totipotency.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXisFKmug%3D%3D&md5=44aee8195d788d6a4f5baa24199c0fdcCAS | 15671666PubMed |

Dissen, G. A., Garcia-Rudaz, C., and Ojeda, S. R. (2009). Role of neurotrophic factors in early ovarian development. Semin. Reprod. Med. 27, 24–31.
Role of neurotrophic factors in early ovarian development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhvVGntrw%3D&md5=a4bb648221e2167f40a07015309540b8CAS | 19197802PubMed |

Dorfman, M. D., Kerr, B., Garcia-Rudaz, C., Paredes, A. H., Dissen, G. A., and Ojeda, S. R. (2011). Neurotrophins acting via TRKB receptors activate the JAGGED1–NOTCH2 cell–cell communication pathway to facilitate early ovarian development. Endocrinology 152, 5005–5016.
Neurotrophins acting via TRKB receptors activate the JAGGED1–NOTCH2 cell–cell communication pathway to facilitate early ovarian development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXhs1GrtrbJ&md5=9cfb52c57bd0bd5a46933ca4d62904f3CAS | 22028443PubMed |

Dunaif, A., Graf, M., Mandeli, J., Laumas, V., and Dobrjansky, A. (1987). Characterization of groups of hyperandrogenic women with acanthosis nigricans, impaired glucose tolerance, and/or hyperinsulinemia. J. Clin. Endocrinol. Metab. 65, 499–507.
Characterization of groups of hyperandrogenic women with acanthosis nigricans, impaired glucose tolerance, and/or hyperinsulinemia.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaL2szgvVKnsw%3D%3D&md5=49136c39949f173ac436ac90068be6dbCAS | 3305551PubMed |

Ehrmann, D. A. (2005). Polycystic ovary syndrome. N. Engl. J. Med. 352, 1223–1236.
Polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXisFShtL8%3D&md5=84c17960fdcde6c328dd9b6bb7a505a3CAS | 15788499PubMed |

Eppig, J. J. (1996). Coordination of nuclear and cytoplasmic oocyte maturation in eutherian mammals. Reprod. Fertil. Dev. 8, 485–489.
Coordination of nuclear and cytoplasmic oocyte maturation in eutherian mammals.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK2s%2FitlOgtw%3D%3D&md5=64e25eaf4edc0806dbefa31ca5e2a381CAS | 8870074PubMed |

Farhi, J., Fisch, B., Garor, R., Peled, Y., Pinkas, H., and Abir, R. (2011). Neurotrophin 4 enhances in vitro follicular assembly in human fetal ovaries. Fertil. Steril. 95, 1267–1271.
Neurotrophin 4 enhances in vitro follicular assembly in human fetal ovaries.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXjs1Cksb8%3D&md5=59a4e721cdeb4d98545e0480681c7746CAS | 20447632PubMed |

Goud, P. T., Goud, A. P., Qian, C., Laverge, H., Van der Elst, J., De Sutter, P., and Dhont, M. (1998). In-vitro maturation of human germinal vesicle stage oocytes: role of cumulus cells and epidermal growth factor in the culture medium. Hum. Reprod. 13, 1638–1644.
In-vitro maturation of human germinal vesicle stage oocytes: role of cumulus cells and epidermal growth factor in the culture medium.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXkslOlurY%3D&md5=0b1c35b00058bd90eff79380873ae0e4CAS | 9688405PubMed |

Han, S. J., Vaccari, S., Nedachi, T., Andersen, C. B., Kovacina, K. S., Roth, R. A., and Conti, M. (2006). Protein kinase B/Akt phosphorylation of PDE3A and its role in mammalian oocyte maturation. EMBO J. 25, 5716–5725.
Protein kinase B/Akt phosphorylation of PDE3A and its role in mammalian oocyte maturation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XhtlShsbzO&md5=98d991b44d424e5371aad6e886612c0aCAS | 17124499PubMed |

Hardy, K., and Spanos, S. (2002). Growth factor expression and function in the human and mouse preimplantation embryo. J. Endocrinol. 172, 221–236.
Growth factor expression and function in the human and mouse preimplantation embryo.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38XhvVejt78%3D&md5=844a5282882c0dd1d11874619e96f8e3CAS | 11834440PubMed |

Harel, S., Jin, S., Fisch, B., Feldberg, D., Krissi, H., Felz, C., Freimann, S., Tan, S. L., Ao, A., and Abir, R. (2006). Tyrosine kinase B receptor and its activated neurotrophins in ovaries from human fetuses and adults. Mol. Hum. Reprod. 12, 357–365.
Tyrosine kinase B receptor and its activated neurotrophins in ovaries from human fetuses and adults.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XmsVeisrw%3D&md5=785008745acfd3cc9674dc11ba5a7e1cCAS | 16648150PubMed |

Hoshino, Y., and Sato, E. (2008). Protein kinase B (PKB/Akt) is required for the completion of meiosis in mouse oocytes. Dev. Biol. 314, 215–223.
Protein kinase B (PKB/Akt) is required for the completion of meiosis in mouse oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhtV2mtr8%3D&md5=f25e408b6309540d9c1b026fc7e2ca69CAS | 18177853PubMed |

Jensen, T., and Johnson, A. L. (2001). Expression and function of brain-derived neurotrophin factor and its receptor, TrkB, in ovarian follicles from the domestic hen (Gallus gallus domesticus). J. Exp. Biol. 204, 2087–2095.
| 1:CAS:528:DC%2BD3MXls1ekt7k%3D&md5=72d74b74646c90cc3af1c2b72debd014CAS | 11441050PubMed |

Kawamura, K., Kawamura, N., Mulders, S. M., Sollewijn Gelpke, M. D., and Hsueh, A. J. (2005). Ovarian brain-derived neurotrophic factor (BDNF) promotes the development of oocytes into preimplantation embryos. Proc. Natl Acad. Sci. USA 102, 9206–9211.
Ovarian brain-derived neurotrophic factor (BDNF) promotes the development of oocytes into preimplantation embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXlvF2ku7g%3D&md5=df656d25b9807d1f33b8f6b2a685fd6cCAS | 15967989PubMed |

Krizsan-Agbas, D., Pedchenko, T., Hasan, W., and Smith, P. G. (2003). Oestrogen regulates sympathetic neurite outgrowth by modulating brain derived neurotrophic factor synthesis and release by the rodent uterus. Eur. J. Neurosci. 18, 2760–2768.
Oestrogen regulates sympathetic neurite outgrowth by modulating brain derived neurotrophic factor synthesis and release by the rodent uterus.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD3srns1Olsw%3D%3D&md5=e40580eb4b5f2b43f9a663f5ec92664cCAS | 14656325PubMed |

Lee, E., Jeong, Y. I., Park, S. M., Lee, J. Y., Kim, J. H., Park, S. W., Hossein, M. S., Jeong, Y. W., Kim, S., Hyun, S. H., and Hwang, W. S. (2007). Beneficial effects of brain-derived neurotropic factor on in vitro maturation of porcine oocytes. Reproduction 134, 405–414.
Beneficial effects of brain-derived neurotropic factor on in vitro maturation of porcine oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhtFKnur3N&md5=c9522ae3f115f02020e0bf193b0f6bfdCAS | 17709559PubMed |

Li, H., Chen, Y., Yan, L. Y., and Qiao, J. (2013). Increased expression of P450scc and CYP17 in development of endogenous hyperandrogenism in aratmodel of PCOS. Endocrine 43, 184–190.
Increased expression of P450scc and CYP17 in development of endogenous hyperandrogenism in aratmodel of PCOS.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhtVegurvN&md5=2ae048c691b090996e2a8dbffb326958CAS | 22798247PubMed |

Linher, K., Wu, D., and Li, J. (2007). Glial cell line-derived neurotrophic factor: an intraovarian factor that enhances oocyte developmental competence in vitro. Endocrinology 148, 4292–4301.
Glial cell line-derived neurotrophic factor: an intraovarian factor that enhances oocyte developmental competence in vitro.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXpslCntb0%3D&md5=aa3fc61d44b8012de1ed2791637825c9CAS | 17540724PubMed |

Linher-Melville, K., and Li, J. (2013). The roles of glial cell line-derived neurotrophic factor, brain-derived neurotrophic factor and nerve growth factor during the final stage of folliculogenesis: a focus on oocyte maturation. Reproduction 145, R43–R54.
The roles of glial cell line-derived neurotrophic factor, brain-derived neurotrophic factor and nerve growth factor during the final stage of folliculogenesis: a focus on oocyte maturation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXjtlOntr8%3D&md5=cf721d50985def315ccba5bf9d8e22aaCAS | 23166367PubMed |

Martins da Silva, S. J., Gardner, J. O., Taylor, J. E., Springbett, A., De Sousa, P. A., and Anderson, R. A. (2005). Brain-derived neurotrophic factor promotes bovine oocyte cytoplasmic competence for embryo development. Reproduction 129, 423–434.
Brain-derived neurotrophic factor promotes bovine oocyte cytoplasmic competence for embryo development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXjs1ertLk%3D&md5=a506bb998546b37fb047276508521d17CAS | 15798017PubMed |

Ni, X. R., Sun, Z. J., Hu, G. H., and Wang, R. H. (2015). High concentration of insulin promotes apoptosis of primary cultured rat ovarian granulose cells via its increase in extracellular HMGB1. Reprod. Sci. 22, 271–277.
High concentration of insulin promotes apoptosis of primary cultured rat ovarian granulose cells via its increase in extracellular HMGB1.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXlt1Glur4%3D&md5=cc6944876576b1e2d15a621b68e5920dCAS | 25228632PubMed |

Norman, R. J., Dewailly, D., Legro, R. S., and Hickey, T. E. (2007). Polycystic ovary syndrome. Lancet 370, 685–697.
Polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXpsFWrtrk%3D&md5=a35d7ef075ea1679b1c6c244f1d11ac6CAS | 17720020PubMed |

Numakawa, T., Suzuki, S., Kumamaru, E., Adachi, N., Richards, M., and Kunugi, H. (2010). BDNF function and intracellular signaling in neurons. Histol. Histopathol. 25, 237–258.
| 1:CAS:528:DC%2BC3cXislamt70%3D&md5=ab014feddcece496188470432484e105CAS | 20017110PubMed |

Oron, G., Ao, A., Friedman, O., Fisch, B., Zhang, X. Y., Ben-Haroush, A., Peled, Y., and Abir, R. (2011). Expression of neurotrophin 3 and its tropomyosin-related kinase receptor C in human preantral follicles. Fertil. Steril. 95, 2056–2062.
Expression of neurotrophin 3 and its tropomyosin-related kinase receptor C in human preantral follicles.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXltFymtLg%3D&md5=ba7df9cb1508d90e3a481d1332c56a7bCAS | 21392742PubMed |

Pan, W., Banks, W. A., Fasold, M. B., Bluth, J., and Kastin, A. J. (1998). Transport of brain-derived neurotrophic factor across the blood–brain barrier. Neuropharmacology 37, 1553–1561.
Transport of brain-derived neurotrophic factor across the blood–brain barrier.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1MXisFensQ%3D%3D&md5=21ad3f57421356dd773f318451bf068fCAS | 9886678PubMed |

Paredes, A., Romero, C., Dissen, G. A., DeChiara, T. M., Reichardt, L., Cornea, A., Ojeda, S. R., and Xu, B. (2004). TrkB receptors are required for follicular growth and oocyte survival in the mammalian ovary. Dev. Biol. 267, 430–449.
TrkB receptors are required for follicular growth and oocyte survival in the mammalian ovary.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXhvV2htr8%3D&md5=bd7497e56ba1db861bcb69320e23deedCAS | 15013804PubMed |

Perreault, S. D., Barbee, R. R., and Slott, V. L. (1988). Importance of glutathione in the acquisition and maintenance of sperm nuclear decondensing activity in maturing hamster oocytes. Dev. Biol. 125, 181–186.
Importance of glutathione in the acquisition and maintenance of sperm nuclear decondensing activity in maturing hamster oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL1cXhvFOltw%3D%3D&md5=6a735eb7e54b7112e04eae7935698012CAS | 3334716PubMed |

Qiao, J., and Feng, H. L. (2011). Extra- and intra-ovarian factors in polycystic ovary syndrome: impact on oocyte maturation and embryo developmental competence. Hum. Reprod. Update 17, 17–33.
Extra- and intra-ovarian factors in polycystic ovary syndrome: impact on oocyte maturation and embryo developmental competence.Crossref | GoogleScholarGoogle Scholar | 20639519PubMed |

Reddy, P., Shen, L., Ren, C., Boman, K., Lundin, E., Ottander, U., Lindgren, P., Liu, Y. X., Sun, Q. Y., and Liu, K. (2005). Activation of Akt (PKB) and suppression of FKHRL1 in mouse and rat oocytes by stem cell factor during follicular activation and development. Dev. Biol. 281, 160–170.
Activation of Akt (PKB) and suppression of FKHRL1 in mouse and rat oocytes by stem cell factor during follicular activation and development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXkt1Gjtr4%3D&md5=ed87535a69a641261c9613f5eaea4b95CAS | 15893970PubMed |

Rieger, D., Luciano, A. M., Modina, S., Pocar, P., Lauria, A., and Gandolfi, F. (1998). The effects of epidermal growth factor and insulin-like growth factor I on the metabolic activity, nuclear maturation and subsequent development of cattle oocytes in vitro. J. Reprod. Fertil. 112, 123–130.
The effects of epidermal growth factor and insulin-like growth factor I on the metabolic activity, nuclear maturation and subsequent development of cattle oocytes in vitro.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXitVKqtLw%3D&md5=54d1dfdf37bc8112511a6b04f603a278CAS | 9538337PubMed |

Rose, C. R., Blum, R., Pichler, B., Lepier, A., Kafitz, K. W., and Konnerth, A. (2003). Truncated TrkB-T1 mediates neurotrophin-evoked calcium signaling in glia cells. Nature 426, 74–78.
Truncated TrkB-T1 mediates neurotrophin-evoked calcium signaling in glia cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXoslSjtL0%3D&md5=4020727aa6141c90b2a0b185b5a00360CAS | 14603320PubMed |

Russo, N., Russo, M., Daino, D., Bucci, F., Pluchino, N., Casarosa, E., Artini, P. G., Cela, V., Luisi, M., and Genazzani, A. R. (2012). Polycystic ovary syndrome: brain-derived neurotrophic factor (BDNF) plasma and follicular fluid levels. Gynecol. Endocrinol. 28, 241–244.
Polycystic ovary syndrome: brain-derived neurotrophic factor (BDNF) plasma and follicular fluid levels.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XktV2jsb8%3D&md5=8091b2e76797b3f8538edc60556e88f5CAS | 22420627PubMed |

Seifer, D. B., Feng, B., Shelden, R. M., Chen, S., and Dreyfus, C. F. (2002a). Brain-derived neurotrophic factor: a novel human ovarian follicular protein. J. Clin. Endocrinol. Metab. 87, 655–659.
Brain-derived neurotrophic factor: a novel human ovarian follicular protein.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38XhsVSju7c%3D&md5=555f2c817bd41ca0131b6c2d3dd9632bCAS | 11836300PubMed |

Seifer, D. B., Feng, B., Shelden, R. M., Chen, S., and Dreyfus, C. F. (2002b). Neurotrophin-4/5 and neurotrophin-3 are present within the human ovarian follicle but appear to have different paracrine/autocrine functions. J. Clin. Endocrinol. Metab. 87, 4569–4571.
Neurotrophin-4/5 and neurotrophin-3 are present within the human ovarian follicle but appear to have different paracrine/autocrine functions.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38XnvFCrsrk%3D&md5=58e8edef9f913c833ec83835688ccd54CAS | 12364436PubMed |

Seifer, D. B., Lambert-Messerlian, G., and Schneyer, A. L. (2003). Ovarian brain-derived neurotrophic factor is present in follicular fluid from normally cycling women. Fertil. Steril. 79, 451–452.
Ovarian brain-derived neurotrophic factor is present in follicular fluid from normally cycling women.Crossref | GoogleScholarGoogle Scholar | 12568867PubMed |

Seifer, D. B., Feng, B., and Shelden, R. M. (2006). Immunocytochemical evidence for the presence and location of the neurotrophin-Trk receptor family in adult human preovulatory ovarian follicles. Am. J. Obstet. Gynecol. 194, 1129–1134.
Immunocytochemical evidence for the presence and location of the neurotrophin-Trk receptor family in adult human preovulatory ovarian follicles.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XjtFajtLc%3D&md5=012d9dfefa3d4f57080895ea39ad2ac0CAS | 16580310PubMed |

Skup, M., Dwornik, A., Macias, M., Sulejczak, D., Wiater, M., and Czarkowska-Bauch, J. (2002). Long-term locomotor training up regulates TrkB (FL) receptor-like proteins, brain derived neurotrophic factor, and neurotrophin 4 with different topographies of expression in oligodendroglia and neurons in the spinal cord. Exp. Neurol. 176, 289–307.
Long-term locomotor training up regulates TrkB (FL) receptor-like proteins, brain derived neurotrophic factor, and neurotrophin 4 with different topographies of expression in oligodendroglia and neurons in the spinal cord.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38XntF2jt7g%3D&md5=4e522b52bff31459c2241ad67468f6ccCAS | 12359171PubMed |

Spears, N., Molinek, M. D., Robinson, L. L., Fulton, N., Cameron, H., Shimoda, K., Telfer, E. E., Anderson, R. A., and Price, D. J. (2003). The role of neurotrophin receptors in female germ-cell survival in mouse and human. Development 130, 5481–5491.
The role of neurotrophin receptors in female germ-cell survival in mouse and human.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXps12qtL4%3D&md5=8719029d46339649c9d548d9744ed9e6CAS | 14507777PubMed |

Wang, X., Sun, Z., Zhen, J., and Yu, Q. (2011). Brain-derived neurotrophic factor from follicular fluid is positively associated with rate of mature ooocytes collected and cleavage rate in intracytoplasmic sperm injection patients. J. Assist. Reprod. Genet. 28, 1053–1058.
Brain-derived neurotrophic factor from follicular fluid is positively associated with rate of mature ooocytes collected and cleavage rate in intracytoplasmic sperm injection patients.Crossref | GoogleScholarGoogle Scholar | 21901363PubMed |

Wessels, J. M., Wu, L., Leyland, N. A., Wang, H., and Foster, W. G. (2014). The brain–uterus connection: brain derived neurotrophic factor (BDNF) and its receptor (Ntrk2) are conserved in the mammalian uterus. PLoS One 9, e94036.
The brain–uterus connection: brain derived neurotrophic factor (BDNF) and its receptor (Ntrk2) are conserved in the mammalian uterus.Crossref | GoogleScholarGoogle Scholar | 24714156PubMed |

Yu, Y., Yan, J., Li, M., Yan, L., Zhao, Y., Lian, Y., Li, R., Liu, P., and Qiao, J. (2012). Effects of combined epidermal growth factor, brain-derived neurotrophic factor and insulin-like growth factor-1 on human oocyte maturation and early fertilized and cloned embryo development. Hum. Reprod. 27, 2146–2159.
Effects of combined epidermal growth factor, brain-derived neurotrophic factor and insulin-like growth factor-1 on human oocyte maturation and early fertilized and cloned embryo development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XptVGju7g%3D&md5=351d8153e9b897e73d23b2d589cdd056CAS | 22532606PubMed |

Zhang, L., Liang, Y., Liu, Y., and Xiong, C. L. (2010). The role of brain-derived neurotrophic factor in mouse oocyte maturation in vitro involves activation of protein kinase B. Theriogenology 73, 1096–1103.
The role of brain-derived neurotrophic factor in mouse oocyte maturation in vitro involves activation of protein kinase B.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXktlClu7c%3D&md5=60fdfb1313f15da1d26e42a87689ea65CAS | 20171721PubMed |

Zhao, P., Qiao, J., Huang, S., Zhang, Y., Liu, S., Yan, L. Y., Hsueh, A. J., and Duan, E. K. (2011). Gonadotrophin-induced paracrine regulation of human oocyte maturation by BDNF and GDNF secreted by granulosa cells. Hum. Reprod. 26, 695–702.
Gonadotrophin-induced paracrine regulation of human oocyte maturation by BDNF and GDNF secreted by granulosa cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXitVSntrY%3D&md5=f0ca5645a8d1b40162e730030847199cCAS | 21227937PubMed |

Zhao, H., Xu, X., Xing, X., Wang, J., He, L., Shi, Y., Shi, Y., Zhao, Y., and Chen, Z. J. (2012). Family-based analysis of susceptibility loci for polycystic ovary syndrome on chromosome 2p16.3, 2p21 and 9q33.3. Hum. Reprod. 27, 294–298.
Family-based analysis of susceptibility loci for polycystic ovary syndrome on chromosome 2p16.3, 2p21 and 9q33.3.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXhs1Cqtr3P&md5=6612678fe8ea49d1b9119c9b8c63bae1CAS | 22081247PubMed |