Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell Endocrinol ; 546: 111577, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35121076

ABSTRACT

The homeodomain transcription factor SIX3 is a known regulator of eye, nose, and forebrain development, and has recently been implicated in female reproduction. Germline heterozygosity of SIX3 is sufficient to cause subfertility, but the cell populations that mediate this role are unknown. The neuropeptide kisspeptin is a critical component of the reproductive axis and plays roles in sexual maturation, ovulation, and the maintenance of gonadotropin secretion. We used Cre-Lox technology to remove Six3 specifically from kisspeptin neurons in mice to test the hypothesis that SIX3 in kisspeptin neurons is required for reproduction. We found that loss of Six3 in kisspeptin neurons causes subfertility and estrous cycle irregularities in females, but no effect in males. Overall, we find that SIX3 expression in kisspeptin neurons is an important contributor to female fertility.


Subject(s)
Eye Proteins , Homeodomain Proteins , Infertility , Kisspeptins , Nerve Tissue Proteins , Neurons , Animals , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Gonadotropin-Releasing Hormone/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Kisspeptins/genetics , Kisspeptins/metabolism , Male , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Reproduction/physiology , Homeobox Protein SIX3
2.
Endocrinology ; 162(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34529765

ABSTRACT

Polycystic ovary syndrome (PCOS) is a common reproductive disorder characterized by elevated androgens and antimüllerian hormone (AMH). These hormones remain elevated throughout pregnancy, and potential effects of hormone exposure on offspring from women with PCOS remain largely unexplored. Expanding on recent reports of prenatal AMH exposure in mice, we have fully characterized the reproductive consequences of prenatal AMH (pAMH) exposure throughout the lifespan of first- and second-generation offspring of both sexes. We also sought to elucidate mechanisms underlying pAMH-induced reproductive effects. There is a known reciprocal relationship between AMH and androgens, and in PCOS and PCOS-like animal models, androgen feedback is dysregulated at the level of the hypothalamus. Kisspeptin neurons express androgen receptors and play a critical role in sexual development and function. We therefore hypothesized that pAMH-induced reproductive phenotypes would be mediated by androgen signaling at the level of kisspeptin cells. We tested the pAMH model in kisspeptin-specific androgen receptor knockout (KARKO) mice and found that virtually all pAMH-induced phenotypes assayed are eliminated in KARKO offspring compared to littermate controls. By demonstrating the necessity of androgen receptor in kisspeptin cells to induce pAMH phenotypes, we have advanced understanding of the interactions between AMH and androgens in the context of prenatal exposure, which could have significant implications for children of women with PCOS.


Subject(s)
Anti-Mullerian Hormone/pharmacology , Prenatal Exposure Delayed Effects , Receptors, Androgen/physiology , Reproduction/drug effects , Animals , Brain/drug effects , Brain/metabolism , Female , Gonads/drug effects , Gonads/metabolism , Kisspeptins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Receptors, Androgen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...