The overall goal of this application is to elucidate the neuroendocrine mechanisms involved in the developmental regulation of ovarian function. The concept is proposed that the developing ovary is subjected to a """"""""neuroendocrinotrophic"""""""" control effected by the interactive participation of the extrinsic innervation, an intragonadal source of neurotransmitters, and a family of neurotrophic genes previously believed to target only the nervous system for their biological actions. This concept stems from a series of observations demonstrating: a) the contribution of the extrinsic innervation to the regulation of ovarian steroidogenesis and follicular development, b) the involvement of nerve growth factor (NGF) in regulating ovarian development indirectly via its neurotrophic effects on the ovarian innervation, c) the ability of neurotransmitters contained in ovarian nerves to initiate the molecular differentiation of early granulosa cells at the onset of follicular growth, and d) the existence in the primate ovary of an intrinsic source of catecholamine biosynthesis. Further studies suggested that an enhanced activity of the ovarian sympathetic innervation may contribute to the etiology of polycystic ovarian syndrome, the most common ovarian pathology affecting women of reproductive age. While studying the role of NGF in supporting the ovarian innervation, it became apparent that the developing ovary not only synthesizes several members of the NGF family of neurotrophins, but unexpectedly, also expresses the tyrosine kinase receptors that mediate the biological actions of neurotrophins in the nervous system. The presence of such receptors in endocrine cells of the ovary implies that neurotrophins can directly affect ovarian function without the intermediacy of the innervation, and raises the intriguing possibility of an hitherto unsuspected role for neurotrophins in the control of ovarian development. The present application proposes a combination of physiological, cellular and molecular approaches to address this issue, and to define the contribution of each of the proposed """"""""neuroendocrinotrophic"""""""" components to the regulation of specific developmental events in ovarian maturation. To this end, the following specific aims are proposed: 1. To examine the hypothesis that two neurotrophins, NGF and neurotrophin-4, play different, but complementary roles in the cytodifferentiation/ organizational process underlying the period of definitive ovarian histogenesis, at the time of follicular formation. 2. To examine the hypothesis that neurotransmitters and neurotrophins interact positively in promoting the molecular differentiation of granulosa cells during early follicular development. 3. To examine the hypothesis that activation of trkA receptors during the hours preceding the first ovulation contributes to the cytodifferentiation process leading to follicular rupture. 4. To examine the hypothesis that activation of ovarian NGF synthesis and that of its low-affinity receptor is a key component in the etiology of polycystic ovarian syndrome. 5. To characterize the molecular forms and cellular sources of tyrosine hydroxylase and dopamine-beta hydroxylase, the two key enzymes in catecholamine synthesis recently found to be expressed in the developing primate ovary.

Agency
National Institute of Health (NIH)
Institute
Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD)
Type
Research Project (R01)
Project #
5R01HD024870-10
Application #
2332252
Study Section
Reproductive Biology Study Section (REB)
Project Start
1988-02-01
Project End
2001-01-31
Budget Start
1997-02-01
Budget End
1998-01-31
Support Year
10
Fiscal Year
1997
Total Cost
Indirect Cost
Name
Oregon Regional Primate Research Center
Department
Type
DUNS #
City
Beaverton
State
OR
Country
United States
Zip Code
97006
Meinel, Sabine; Blohberger, Jan; Berg, Dieter et al. (2015) Pro-nerve growth factor in the ovary and human granulosa cells. Horm Mol Biol Clin Investig 24:91-9
De La Chesnaye, Elsa; Méndez, Juan Pablo; López-Romero, Ricardo et al. (2015) FBXW12, a novel F box protein-encoding gene, is deleted or methylated in some cases of epithelial ovarian cancer. Int J Clin Exp Pathol 8:10192-203
Blohberger, J; Kunz, L; Einwang, D et al. (2015) Readthrough acetylcholinesterase (AChE-R) and regulated necrosis: pharmacological targets for the regulation of ovarian functions? Cell Death Dis 6:e1685
Dorfman, Mauricio D; Garcia-Rudaz, Cecilia; Alderman, Zefora et al. (2014) Loss of Ntrk2/Kiss1r signaling in oocytes causes premature ovarian failure. Endocrinology 155:3098-111
Gaytan, Francisco; Garcia-Galiano, David; Dorfman, Mauricio D et al. (2014) Kisspeptin receptor haplo-insufficiency causes premature ovarian failure despite preserved gonadotropin secretion. Endocrinology 155:3088-97
Wilson, Jenny L; Chen, Weiyi; Dissen, Gregory A et al. (2014) Excess of nerve growth factor in the ovary causes a polycystic ovary-like syndrome in mice, which closely resembles both reproductive and metabolic aspects of the human syndrome. Endocrinology 155:4494-506
Dissen, G A; Lomniczi, A; Boudreau, R L et al. (2012) Targeted gene silencing to induce permanent sterility. Reprod Domest Anim 47 Suppl 4:228-32
Dissen, Gregory A; Lomniczi, Alejandro; Heger, Sabine et al. (2012) Hypothalamic EAP1 (enhanced at puberty 1) is required for menstrual cyclicity in nonhuman primates. Endocrinology 153:350-61
Garcia-Rudaz, Cecilia; Dorfman, Mauricio; Nagalla, Srinivasa et al. (2011) Excessive ovarian production of nerve growth factor elicits granulosa cell apoptosis by setting in motion a tumor necrosis factor ?/stathmin-mediated death signaling pathway. Reproduction 142:319-31
Dorfman, Mauricio D; Kerr, Bredford; Garcia-Rudaz, Cecilia et al. (2011) Neurotrophins acting via TRKB receptors activate the JAGGED1-NOTCH2 cell-cell communication pathway to facilitate early ovarian development. Endocrinology 152:5005-16

Showing the most recent 10 out of 61 publications