Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Biol Reprod ; 93(3): 69, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26203175

ABSTRACT

Polycystic ovary syndrome (PCOS) pathophysiology is poorly understood, due partly to lack of PCOS animal models fully recapitulating this complex disorder. Recently, a PCOS rat model using letrozole (LET), a nonsteroidal aromatase inhibitor, mimicked multiple PCOS phenotypes, including metabolic features absent in other models. Given the advantages of using genetic and transgenic mouse models, we investigated whether LET produces a similar PCOS phenotype in mice. Pubertal female C57BL/6N mice were treated for 5 wk with LET, which resulted in increased serum testosterone and normal diestrus levels of estradiol, similar to the hyperandrogenemia and follicular phase estrogen levels of PCOS women. As in PCOS, ovaries from LET mice were larger, polycystic, and lacked corpora lutea versus controls. Most LET females were acyclic, and all were infertile. LET females displayed elevated serum LH levels and higher Lhb mRNA in the pituitary. In contrast, serum FSH and Fshb were significantly reduced in LET females, demonstrating differential effects on gonadotropins, as in PCOS. Within the ovary, LET females had higher Cyp17, Cyp19, and Fsh receptor mRNA expression. In the hypothalamus, LET females had higher kisspeptin receptor mRNA expression but lower progesterone receptor mRNA levels. LET females also gained more weight than controls, had increased abdominal adiposity and adipocyte size, elevated adipose inflammatory mRNA levels, and impaired glucose tolerance, mirroring the metabolic phenotype in PCOS women. This is the first report of a LET paradigm in mice that recapitulates both reproductive and metabolic PCOS phenotypes and will be useful to genetically probe the PCOS condition.


Subject(s)
Enzyme Inhibitors/toxicity , Nitriles/toxicity , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/pathology , Reproduction/drug effects , Triazoles/toxicity , Animals , Corpus Luteum/metabolism , Diestrus/metabolism , Estrous Cycle/drug effects , Female , Hyperandrogenism/blood , Hyperandrogenism/chemically induced , Hypothalamus/drug effects , Hypothalamus/metabolism , Kisspeptins/biosynthesis , Kisspeptins/genetics , Letrozole , Mice , Mice, Inbred C57BL , Phenotype , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Polycystic Ovary Syndrome/metabolism , Pregnancy , Testosterone/blood
2.
Endocrinology ; 155(11): 4433-46, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25147978

ABSTRACT

Hypothalamic GnRH is the master regulator of the neuroendocrine reproductive axis, and its secretion is regulated by many factors. Among these is kisspeptin (Kp), a potent trigger of GnRH secretion. Kp signals via the Kp receptor (KISS1R), a Gαq/11-coupled 7-transmembrane-spanning receptor. Until this study, it was understood that KISS1R mediates GnRH secretion via the Gαq/11-coupled pathway in an ERK1/2-dependent manner. We recently demonstrated that KISS1R also signals independently of Gαq/11 via ß-arrestin and that this pathway also mediates ERK1/2 activation. Because GnRH secretion is ERK1/2-dependent, we hypothesized that KISS1R regulates GnRH secretion via both the Gαq/11- and ß-arrestin-coupled pathways. To test this hypothesis, we measured LH secretion, a surrogate marker of GnRH secretion, in mice lacking either ß-arrestin-1 or ß-arrestin-2. Results revealed that Kp-dependent LH secretion was significantly diminished relative to wild-type mice (P < .001), thus supporting that ß-arrestin mediates Kp-induced GnRH secretion. Based on this, we hypothesized that Gαq/11-uncoupled KISS1R mutants, like L148S, will display Gαq/11-independent signaling. To test this hypothesis, L148S was expressed in HEK 293 cells. and results confirmed that, although strongly uncoupled from Gαq/11, L148S retained the ability to trigger significant Kp-dependent ERK1/2 phosphorylation (P < .05). Furthermore, using mouse embryonic fibroblasts lacking ß-arrestin-1 and -2, we demonstrated that L148S-mediated ERK1/2 phosphorylation is ß-arrestin-dependent. Overall, we conclude that KISS1R signals via Gαq/11 and ß-arrestin to regulate GnRH secretion. This novel and important finding could explain why patients bearing some types of Gαq/11-uncoupled KISS1R mutants display partial gonadotropic deficiency and even a reversal of the condition, idiopathic hypogonadotropic hypogonadism.


Subject(s)
Arrestins/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Luteinizing Hormone/metabolism , Receptors, G-Protein-Coupled/physiology , Animals , Arrestins/genetics , Buserelin/pharmacology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Kisspeptin-1 , Signal Transduction/drug effects , Signal Transduction/genetics , beta-Arrestin 1 , beta-Arrestin 2 , beta-Arrestins
3.
Endocrinology ; 155(10): 3909-19, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25057795

ABSTRACT

Mutations in neurokinin B (NKB) and its receptor, NK3R, were identified in human patients with hypogonadotropic hypogonadism, a disorder characterized by lack of puberty and infertility. Further studies have suggested that NKB acts at the level of the hypothalamus to control GnRH neuron activity, either directly or indirectly. We recently reported that treatment with senktide, a NK3R agonist, induced GnRH secretion and expression of c-fos mRNA in GT1-7 cells. Here, we map the responsive region in the murine c-fos promoter to between -400 and -200 bp, identify the signal transducer and activator of transcription (STAT) (-345) and serum response element (-310) sites as required for induction, a modulatory role for the Ets site (-318), and show that induction is protein kinase C dependent. Using gel shift and Gal4 assays, we further show that phosphorylation of Elk-1 leads to binding to DNA in complex with serum response factor at serum response element and Ets sites within the c-fos promoter. Thus, we determine molecular mechanisms involved in NKB regulation of c-fos induction, which may play a role in modulation of GnRH neuron activation.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neurokinin B/physiology , Neurons/drug effects , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-fos/genetics , Serum Response Factor/metabolism , Transcription, Genetic/physiology , ets-Domain Protein Elk-1/physiology , Animals , Cell Line, Transformed , Mice , Neurokinin B/pharmacology , Neurons/metabolism , Rats , Signal Transduction , Transcription, Genetic/drug effects , Up-Regulation/drug effects
4.
Mol Endocrinol ; 27(3): 437-54, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23393128

ABSTRACT

Genetic studies in human patients with idiopathic hypogonadotropic hypogonadism (IHH) identified mutations in the genes that encode neurokinin B (NKB) and the neurokinin 3 receptor (NK3R). However, determining the mechanism whereby NKB regulates gonadotropin secretion has been difficult because of conflicting results from in vivo studies investigating the luteinizing hormone (LH) response to senktide, a NK3R agonist. NK3R is expressed in a subset of GnRH neurons and in kisspeptin neurons that are known to regulate GnRH secretion. Thus, one potential source of inconsistency is that NKB could produce opposing direct and indirect effects on GnRH secretion. Here, we employ the GT1-7 cell model to elucidate the direct effects of NKB on GnRH neuron function. We find that GT1-7 cells express NK3R and respond to acute senktide treatment with c-Fos induction and increased GnRH secretion. In contrast, long-term senktide treatment decreased GnRH secretion. Next, we focus on the examination of the mechanism underlying the long-term decrease in secretion and determine that senktide treatment represses transcription of GnRH. We further show that this repression of GnRH transcription may involve enhanced c-Fos protein binding at novel activator protein-1 (AP-1) half-sites identified in enhancer 1 and the promoter, as well as chromatin remodeling at the promoter of the GnRH gene. These data indicate that NKB could directly regulate secretion from NK3R-expressing GnRH neurons. Furthermore, whether the response is inhibitory or stimulatory toward GnRH secretion could depend on the history or length of exposure to NKB because of a repressive effect on GnRH transcription.


Subject(s)
Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Transcription, Genetic , Animals , Base Pairing/genetics , Binding Sites , Cell Line , Chromatin Assembly and Disassembly/drug effects , Chromatin Assembly and Disassembly/genetics , Enhancer Elements, Genetic/genetics , Humans , Mice , Neurons/drug effects , Peptide Fragments/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Receptors, Neurokinin-3/metabolism , Sequence Deletion/genetics , Substance P/analogs & derivatives , Substance P/pharmacology , Time Factors , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...