Much is known about the hormonal mechanisms controlling ovarian development. More recently, a major focus of attention in the field has been the identification of regulatory pathways that, operating within the ovarian microenvironment, contribute to the acquisition of ovarian reproductive competence. Within this framework, our laboratory has developed the concept that neurotrophins (NTs) and their Trk tyrosine kinase receptors, long thought to be exclusively required for the development of the nervous system are also involved in the control of ovarian function. Employing gene targeting approaches we identified trkB, the high-affinity receptor for neurotrophin-4/5 (NT-4/5) and brain-derived neurotrophic factor (BDNF), as a signaling molecule required for early follicular growth and oocyte survival. In addition, we showed that nerve growth factor (NGF) contributes independently to the initiation of follicular growth. Other studies indicated that NGF acting via trkA receptors is also important for ovulation, but that despite this physiological role, an inappropriately sustained increase in intraovarian NGF synthesis results in functional alterations leading to the development of follicular cysts. Based on these findings, the present renewal application proposes the following Specific Aims: 1) To define the TrkB receptor isoform (full-length or truncated) required for early follicle growth and oocyte survival, and identify the cells primarily responsive to TrkB signaling. The objectives of this Aim will be achieved using Cre-loxP technology to specifically disrupt the expression of full-length and truncated TrkB isoforms in either oocytes or granulosa cells. 2) To test the hypothesis that NTs signaling via TrkB receptors promote early follicular growth by supporting an oocyte-to granulosa cell Jagged 1-Notch2 communication pathway.
This aim will be achieved with the combined use of cell-specific trkB KOs and cellular/biochemical approaches to define the relationship that exists between TrkB signaling and the Notch2 pathway. 3) To test the hypotheses that while NGF-dependent trkA signaling is required for the normal development of antral follicles and ovulation, an overproduction of NGF compromises the ability of antral follicles to reach a preovulatory stage, and thus establishes conditions leading to the development of polycystic ovaries. To accomplish this Aim we will use transgenic mice that overexpress NGF in a cell specific manner, and mice in which signaling through p75 (the common NT receptor), or trkA (the high-affinity NGF receptor) are conditionally disrupted in ovarian cells. 4) To test the hypothesis that an excess of ovarian NGF creates conditions in the local microenvironment that favor the development of polycystic ovaries in nonhuman primates. To accomplish this Aim we will use a lentiviral delivery system to enhance the production of NGF in the interstitial compartment of the adult nonhuman 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 #
5R01HD024870-22
Application #
7796869
Study Section
Integrative and Clinical Endocrinology and Reproduction Study Section (ICER)
Program Officer
Grave, Gilman D
Project Start
2007-05-01
Project End
2012-04-30
Budget Start
2010-05-01
Budget End
2011-04-30
Support Year
22
Fiscal Year
2010
Total Cost
$338,116
Indirect Cost
Name
Oregon Health and Science University
Department
Physiology
Type
Schools of Medicine
DUNS #
096997515
City
Portland
State
OR
Country
United States
Zip Code
97239
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
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
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

Showing the most recent 10 out of 61 publications