Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Horm Res Paediatr ; 88(6): 371-395, 2017.
Article in English | MEDLINE | ID: mdl-29156452

ABSTRACT

This paper represents an international collaboration of paediatric endocrine and other societies (listed in the Appendix) under the International Consortium of Paediatric Endocrinology (ICPE) aiming to improve worldwide care of adolescent girls with polycystic ovary syndrome (PCOS)1. The manuscript examines pathophysiology and guidelines for the diagnosis and management of PCOS during adolescence. The complex pathophysiology of PCOS involves the interaction of genetic and epigenetic changes, primary ovarian abnormalities, neuroendocrine alterations, and endocrine and metabolic modifiers such as anti-Müllerian hormone, hyperinsulinemia, insulin resistance, adiposity, and adiponectin levels. Appropriate diagnosis of adolescent PCOS should include adequate and careful evaluation of symptoms, such as hirsutism, severe acne, and menstrual irregularities 2 years beyond menarche, and elevated androgen levels. Polycystic ovarian morphology on ultrasound without hyperandrogenism or menstrual irregularities should not be used to diagnose adolescent PCOS. Hyperinsulinemia, insulin resistance, and obesity may be present in adolescents with PCOS, but are not considered to be diagnostic criteria. Treatment of adolescent PCOS should include lifestyle intervention, local therapies, and medications. Insulin sensitizers like metformin and oral contraceptive pills provide short-term benefits on PCOS symptoms. There are limited data on anti-androgens and combined therapies showing additive/synergistic actions for adolescents. Reproductive aspects and transition should be taken into account when managing adolescents.


Subject(s)
Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/therapy , Adolescent , Congresses as Topic , Female , Humans
2.
PLoS One ; 11(7): e0157027, 2016.
Article in English | MEDLINE | ID: mdl-27467141

ABSTRACT

The melanocortin system includes five G-protein coupled receptors (family A) defined as MC1R-MC5R, which are stimulated by endogenous agonists derived from proopiomelanocortin (POMC). The melanocortin system has been intensely studied for its central actions in body weight and energy expenditure regulation, which are mainly mediated by MC4R. The pituitary gland is the source of various POMC-derived hormones released to the circulation, which raises the possibility that there may be actions of the melanocortins on peripheral energy homeostasis. In this study, we examined the molecular signaling pathway involved in α-MSH-stimulated glucose uptake in differentiated L6 myotubes and mouse muscle explants. In order to examine the involvement of AMPK, we investigate -MSH stimulation in both wild type and AMPK deficient mice. We found that -MSH significantly induces phosphorylation of TBC1 domain (TBC1D) family member 1 (S237 and T596), which is independent of upstream PKA and AMPK. We find no evidence to support that -MSH-stimulated glucose uptake involves TBC1D4 phosphorylation (T642 and S704) or GLUT4 translocation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , GTPase-Activating Proteins/metabolism , Glucose/metabolism , Muscle, Skeletal/drug effects , alpha-MSH/pharmacology , AMP-Activated Protein Kinases/genetics , Animals , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Phosphorylation , Signal Transduction
3.
Horm Res Paediatr ; 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25833060

ABSTRACT

BACKGROUND/AIMS: The diagnostic criteria for polycystic ovary syndrome (PCOS) in adolescence are controversial, primarily because the diagnostic pathological features used in adult women may be normal pubertal physiological events. Hence, international pediatric and adolescent specialty societies have defined criteria that have sufficient evidence to be used for the diagnosis of PCOS in adolescents. METHODS: The literature has been reviewed and evidence graded to address a series of questions regarding the diagnosis of PCOS during adolescence including the following: clinical and biochemical evidence of hyperandrogenism, criteria for oligo-anovulation and polycystic ovary morphology, diagnostic criteria to exclude other causes of hyperandrogenism and amenorrhea, role of insulin resistance, and intervention. RESULTS AND CONCLUSION: Features of PCOS overlap normal pubertal development. Hence, caution should be taken before diagnosing PCOS without longitudinal evaluation. However, treatment may be indicated even in the absence of a definitive diagnosis. While obesity, insulin resistance, and hyperinsulinemia are common findings in adolescents with hyperandrogenism, these features should not be used to diagnose PCOS among adolescent girls. © 2015 S. Karger AG, Basel.

4.
Endocrinology ; 155(11): 4494-506, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25211588

ABSTRACT

Polycystic ovarian syndrome (PCOS), the most common female endocrine disorder of unknown etiology, is characterized by reproductive abnormalities and associated metabolic conditions comprising insulin resistance, type 2 diabetes mellitus, and dyslipidemia. We previously reported that transgenic overexpression of nerve growth factor (NGF), a marker of sympathetic hyperactivity, directed to the ovary by the mouse 17α-hydroxylase/C17-20 lyase promoter (17NF mice), results in ovarian abnormalities similar to those seen in PCOS women. To investigate whether ovarian overproduction of NGF also induces common metabolic alterations of PCOS, we assessed glucose homeostasis by glucose tolerance test, plasma insulin levels, and body composition by dual-energy x-ray absorptiometry scan in young female 17NF mice and wild-type mice. 17NF mice exhibited increased body weight and alterations in body fat distribution with a greater accumulation of visceral fat compared with sc fat (P < .01). 17NF mice also displayed glucose intolerance (P < .01), decreased insulin-mediated glucose disposal (P < .01), and hyperinsulinemia (P < .05), which, similar to PCOS patients, occurred independently of body weight. Additionally, 17NF mice exhibited increased sympathetic outflow observed as increased interscapular brown adipose tissue temperature. This change was evident during the dark period (7 pm to 7 am) and occurred concomitant with increased interscapular brown adipose tissue uncoupling protein 1 expression. These findings suggest that overexpression of NGF in the ovary may suffice to cause both reproductive and metabolic alterations characteristic of PCOS and support the hypothesis that sympathetic hyperactivity may contribute to the development and/or progression of PCOS.


Subject(s)
Infertility, Female/genetics , Nerve Growth Factor/genetics , Ovary/metabolism , Polycystic Ovary Syndrome/genetics , Animals , Disease Models, Animal , Female , Humans , Infertility, Female/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Growth Factor/metabolism , Ovary/pathology , Phenotype , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Reproduction/genetics , Up-Regulation/genetics
5.
Endocrinology ; 155(11): 4447-60, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25147981

ABSTRACT

In premenopausal and menopausal women in particular, suboptimal estrogens have been linked to the development of the metabolic syndrome as major contributors to fat accumulation. At the same time, estrogens have been described to have a role in regulating body metabolic status. We evaluated how endogenous or administered estrogens impact on the changes associated with high-fat diet (HFD) consumption in 2 different paradigms; ovarian-intact and in ovariectomized mice. When estradiol (E2) was cyclically administered to ovarian-intact HFD-fed mice for 12 weeks, animals gained significantly less weight than ovarian-intact vehicle controls (P < .01). This difference was mainly due to a reduced caloric intake but not to an increase in energy expenditure or locomotor activity. This E2 treatment regime to mice exposed to HFD was overall able to avoid the increase of visceral fat content to levels of those found in mice fed a regular chow diet. In the ovariectomized model, the main body weight and fat content reducing action of E2 was not only through decreasing food intake but also by increasing the whole-body energy expenditure, locomotor activity, and by inducing fat oxidation. Importantly, these animals became responsive to the anorexigenic effects of leptin in contrast to the vehicle-treated and the pair-fed control groups (P < .01). Further, in vitro hypothalamic secretion experiments revealed that treatment of obese mice with E2 is able to modulate the secretion of appetite-regulating neuropeptides; namely, E2 increased the secretion of the anorectic neuropeptide α-melanocyte-stimulating hormone and decreased the secretion of the orexigenic neuropetides neuropeptide Y and Agouti-related peptide. In conclusion, differences in response to E2 treatment of HFD-fed animals depend on their endogenous estrogenic status. Overall, E2 administration overcomes arcuate leptin resistance and partially prevents fat accumulation on these mice.


Subject(s)
Drug Resistance/drug effects , Estradiol/pharmacology , Leptin/pharmacology , Lipid Metabolism/drug effects , Obesity/metabolism , Obesity/prevention & control , Animals , Diet, High-Fat , Female , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Ovariectomy , Sex Factors
6.
Endocrinology ; 155(8): 3098-111, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24877631

ABSTRACT

Neurotrophins (NTs), once believed to be neural-specific trophic factors, are now known to also provide developmental cues to non-neural cells. In the ovary, NTs contribute to both the formation and development of follicles. Here we show that oocyte-specific deletion of the Ntrk2 gene that encodes the NTRK2 receptor (NTRK2) for neurotrophin-4/5 and brain-derived neurotrophic factor (BDNF) results in post-pubertal oocyte death, loss of follicular organization, and early adulthood infertility. Oocytes lacking NTRK2 do not respond to gonadotropins with activation of phosphatidylinositol 3-kinase (PI3K)-AKT-mediated signaling. Before puberty, oocytes only express a truncated NTRK2 form (NTRK2.T1), but at puberty full-length (NTRK2.FL) receptors are rapidly induced by the preovulatory gonadotropin surge. A cell line expressing both NTRK2.T1 and the kisspeptin receptor (KISS1R) responds to BDNF stimulation with activation of Ntrk2 expression only if kisspeptin is present. This suggests that BDNF and kisspeptin that are produced by granulosa cells (GCs) of periovulatory follicles act in concert to mediate the effect of gonadotropins on Ntrk2 expression in oocytes. In keeping with this finding, the oocytes of NTRK2-intact mice fail to respond to gonadotropins with increased Ntrk2 expression in the absence of KISS1R. Our results demonstrate that the preovulatory gonadotropin surge promotes oocyte survival at the onset of reproductive cyclicity by inducing oocyte expression of NTRK2.FL receptors that set in motion an AKT-mediated survival pathway. They also suggest that gonadotropins activate NTRK2.FL expression via a dual communication pathway involving BDNF and kisspeptin produced in GCs and their respective receptors NTRK2.T1 and KISS1R expressed in oocytes.


Subject(s)
Membrane Glycoproteins/metabolism , Oocytes/metabolism , Ovary/metabolism , Primary Ovarian Insufficiency/etiology , Protein-Tyrosine Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Female , Gonadotropins/physiology , Infertility, Female/genetics , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Kisspeptin-1
7.
Endocrinology ; 153(1): 339-49, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22128021

ABSTRACT

Current evidence suggests that the acquisition of female reproductive capacity and the maintenance of mature reproductive function are related processes transcriptionally regulated by gene networks operating within the neuroendocrine brain. One of these genes, termed enhanced at puberty 1 (EAP1), encodes an upstream regulator of these processes. Selective inhibition of EAP1 expression in discrete regions of the rat and nonhuman primate (NHP) hypothalamus, via targeted delivery of RNA interference, either disrupts (rats) or abolishes (monkeys) reproductive cycles. The striking loss of menstrual cyclicity resulting from knocking down hypothalamic EAP1 expression suggests that diminished EAP1 function may contribute to disorders of the menstrual cycle of neuroendocrine origin. Here we show that a single-nucleotide polymorphism in the 5'-flanking region of EAP1 gene is associated with increased incidence of amenorrhea/oligomenorrhea in NHP. In the presence of the risk allele, binding of the transcription factor mothers against decapentaplegic homolog 3 (SMAD3) to its recognition site contained within the polymorphic sequence in the monkey EAP1 promoter is reduced. The risk allele also diminishes the increase in EAP1 promoter activity elicited by TGFß1, a peptide that activates a SMAD3/4-mediated signaling pathway to regulate gene transcription. These findings indicate that common genetic variation in the EAP1 locus increases the susceptibility of NHP to loss/disruption of menstrual cyclicity. They also raise the possibility that polymorphisms in EAP1 may increase the risk of functional hypothalamic amenorrhea in humans.


Subject(s)
Amenorrhea/veterinary , Macaca mulatta/genetics , Monkey Diseases/genetics , Oligomenorrhea/veterinary , Polymorphism, Single Nucleotide , 5' Flanking Region , Amenorrhea/genetics , Amenorrhea/physiopathology , Animals , Base Sequence , Binding Sites/genetics , DNA Primers/genetics , Female , Gene Knockdown Techniques , Linkage Disequilibrium , Macaca mulatta/physiology , Menstrual Cycle/genetics , Menstrual Cycle/physiology , Monkey Diseases/physiopathology , Oligomenorrhea/genetics , Oligomenorrhea/physiopathology , Promoter Regions, Genetic , Smad3 Protein/metabolism , Transcriptional Activation/drug effects , Transforming Growth Factor beta1/pharmacology
8.
Endocrinology ; 152(12): 5005-16, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22028443

ABSTRACT

Tropomyosin-related kinase (TRK) receptor B (TRKB) mediates the supportive actions of neurotrophin 4/5 and brain-derived neurotrophic factor on early ovarian follicle development. Absence of TRKB receptors reduces granulosa cell (GC) proliferation and delays follicle growth. In the present study, we offer mechanistic insights into this phenomenon. DNA array and quantitative PCR analysis of ovaries from TrkB-null mice revealed that by the end of the first week of postnatal life, Jagged1, Hes1, and Hey2 mRNA abundance is reduced in the absence of TRKB receptors. Although Jagged1 encodes a NOTCH receptor ligand, Hes1 and Hey2 are downstream targets of the JAGGED1-NOTCH2 signaling system. Jagged1 is predominantly expressed in oocytes, and the abundance of JAGGED1 is decreased in TrkB(-/-) oocytes. Lack of TRKB receptors also resulted in reduced expression of c-Myc, a NOTCH target gene that promotes entry into the cell cycle, but did not alter the expression of genes encoding core regulators of cell-cycle progression. Selective restoration of JAGGED1 synthesis in oocytes of TrkB(-/-) ovaries via lentiviral-mediated transfer of the Jagged1 gene under the control of the growth differentiation factor 9 (Gdf9) promoter rescued c-Myc expression, GC proliferation, and follicle growth. These results suggest that neurotrophins acting via TRKB receptors facilitate early follicle growth by supporting a JAGGED1-NOTCH2 oocyte-to-GC communication pathway, which promotes GC proliferation via a c-MYC-dependent mechanism.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Communication/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Growth Factors/physiology , Ovary/growth & development , Receptor, Notch2/metabolism , Receptor, trkB/physiology , Animals , Cell Cycle/genetics , Cell Proliferation , Female , Gene Expression Regulation/physiology , Granulosa Cells/cytology , Jagged-1 Protein , Mice , Mice, Knockout , Nerve Growth Factors/metabolism , Ovarian Follicle/cytology , Receptor, Notch2/genetics , Receptor, trkB/metabolism , Serrate-Jagged Proteins
9.
J Neurosci ; 31(34): 12189-97, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21865462

ABSTRACT

Leptin regulates body weight in mice by decreasing appetite and increasing sympathetic nerve activity (SNA), which increases energy expenditure in interscapular brown adipose tissue (iBAT). Diet-induced obese mice (DIO) are resistant to the anorectic actions of leptin. We evaluated whether leptin still stimulated sympathetic outflow in DIO mice. We measured iBAT temperature as a marker of SNA. We found that obese hyperleptinemic mice have higher iBAT temperature than mice on regular diet. Conversely, obese leptin-deficient ob/ob mice have lower iBAT temperature. Additionally, leptin increased SNA in obese (DIO and ob/ob) and control mice, despite DIO mice being resistant to anorectic action of leptin. We demonstrated that neurons in the dorsomedial hypothalamus (DMH) of DIO mice mediate the thermogenic responses to hyperleptinemia in obese mammals because blockade of leptin receptors in the DMH prevented the thermogenic effects of leptin. Peripheral Melotan II (MTII) injection increased iBAT temperature, but it was blunted by blockade of DMH melanocortin receptors (MC4Rs) by injecting agouti-related peptide (AgRP) directly into the DMH, suggesting a physiological role of the DMH on temperature regulation in animals with normal body weight. Nevertheless, obese mice without a functional melanocortin system (MC4R KO mice) have an increased sympathetic outflow to iBAT compared with their littermates, suggesting that higher leptin levels drive sympathoexcitation to iBAT by a melanocortin-independent pathway. Because the sympathetic nervous system contributes in regulating blood pressure, heart rate, and hepatic glucose production, selective leptin resistance may be a crucial mechanism linking adiposity and metabolic syndrome.


Subject(s)
Adipose Tissue, Brown/physiology , Dorsomedial Hypothalamic Nucleus/physiology , Leptin/physiology , Sympathetic Nervous System/physiology , Thermogenesis/physiology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/innervation , Animals , Body Temperature/drug effects , Body Temperature/physiology , Disease Models, Animal , Dorsomedial Hypothalamic Nucleus/drug effects , Drug Resistance/drug effects , Drug Resistance/physiology , Leptin/deficiency , Leptin/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/genetics , Obesity/metabolism , Receptors, Leptin/antagonists & inhibitors , Receptors, Leptin/physiology , Sympathetic Nervous System/drug effects , Thermogenesis/drug effects
10.
Reproduction ; 142(2): 319-31, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21646391

ABSTRACT

Excessive nerve growth factor (NGF) production by the ovary, achieved via a transgenic approach, results in arrested antral follicle growth, reduced ovulatory capacity, and a predisposition to cyst formation in response to mildly elevated LH levels. Two salient features in these mutant mice (termed 17NF) are an elevated production of 17α-hydroxyprogesterone (17-OHP(4)), testosterone, and estradiol (E(2)) in response to gonadotropins, and an increased frequency of granulosa cell (GC) apoptosis. In this study, we show that the increase in steroidal response is associated with enhanced expression of Cyp17a1, Hsd17b, and Cyp19a1, which encode the enzymes catalyzing the synthesis of 17-OHP(4), testosterone, and E(2) respectively. Using a proteomic approach, we identified stathmin (STMN1), as a protein that is overproduced in 17NF ovaries. In its phosphorylated state, STMN1 mediates a cell death signal initiated by tumor necrosis factor α (TNF). STMN1 is expressed in GCs and excessive NGF increases its abundance as well as that of its forms phosphorylated at serine (Ser) 16, 25, and 38. TNF synthesis is also increased in 17NF ovaries, and this change is abolished by blocking neurotrophic tyrosine kinase receptors. Inhibiting TNF actions in vivo by administering a soluble TNF receptor prevented the increase in total and phosphorylated STMN1 production, as well as GC apoptosis in NGF-overproducing ovaries. These results indicate that an excess of NGF in the ovary promotes steroidogenesis by enhancing the expression of enzyme genes involved in 17-OHP(4), testosterone, and E(2) synthesis, and causes GC apoptosis by activating a TNF/ STMN1-mediated cell death pathway.


Subject(s)
Apoptosis , Granulosa Cells/metabolism , Nerve Growth Factor/metabolism , Ovary/metabolism , Signal Transduction , Stathmin/metabolism , Tumor Necrosis Factor-alpha/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Apoptosis/drug effects , Aromatase/genetics , Aromatase/metabolism , Female , Gene Expression Regulation/drug effects , Gonadal Steroid Hormones/metabolism , Gonadotropins/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Transgenic , Nerve Growth Factor/genetics , Ovary/drug effects , Ovary/enzymology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA, Messenger/metabolism , Signal Transduction/drug effects , Stathmin/genetics , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors
11.
Reproduction ; 138(1): 131-40, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19357131

ABSTRACT

Recent studies have demonstrated that neurotrophins (NTs) and their NTRK tyrosine kinase receptors, thought to be exclusively required for the development of the nervous system, are also involved in controlling ovarian development. Here, we show that primordial follicle formation is decreased in the absence of nerve growth factor (NGF) or its receptor NTRK1, and in the absence of NTRK2, the receptor for neurotrophin-4 (NTF4) and brain-derived neurotrophic factor (BDNF). This deficiency is not due to premature oocyte loss, because the ovaries of Ntrk1(-/-) and Ntrk2(-/-) mice do not show an increased rate of oocyte death antedating the initiation of folliculogenesis. Moreover, exposure of NGF-deficient ovaries to NGF rescues the defect in follicular assembly, if NTRK1 receptors are present, suggesting that the absence of NTs causes a delay, and not an irretrievable loss, of follicle formation. Both the number of secondary follicles and FSH receptor (FSHR) expression are diminished in Ntrk1- and Ntrk2-null ovaries, but not in ovaries lacking the common NT receptor NGFR. Transient exposure of wild-type ovaries to NTF4 increases Fshr gene expression and enhances the ability of the ovary to respond to FSH with formation of cyclin D2, a cell cycle protein mediating the proliferative actions of FSH in the ovary. These results indicate that both NTRK1 and NTRK2 receptors are necessary for the timely assembly of primordial follicles and for sustaining early follicular development. They also suggest that a mechanism by which NTRK2 receptors facilitate subsequent follicle development is by inducing the formation of functional FSHR.


Subject(s)
Membrane Glycoproteins/metabolism , Oocytes/metabolism , Ovarian Follicle/metabolism , Ovary/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, trkA/metabolism , Animals , Animals, Newborn , Apoptosis , Cyclin D2/metabolism , Female , Follicle Stimulating Hormone/metabolism , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factor/deficiency , Nerve Growth Factor/genetics , Nerve Growth Factors/metabolism , Oocytes/pathology , Ovarian Follicle/pathology , Ovary/pathology , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Receptor, trkA/deficiency , Receptor, trkA/genetics , Receptors, FSH/metabolism , Signal Transduction , Time Factors , Tissue Culture Techniques
12.
Endocrinology ; 150(6): 2906-14, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19264868

ABSTRACT

Although ovarian nerve growth factor (NGF) facilitates follicular development and ovulation, an excess of the neurotrophin in the rodent ovary reduces ovulatory capacity and causes development of precystic follicles. Here we show that ovarian NGF production is enhanced in patients with polycystic ovarian syndrome (PCOS) and that transgenically driven overproduction of NGF targeted to the ovary results in cystic morphology, when accompanied by elevated LH levels. NGF levels are increased in the follicular fluid from PCOS ovaries and in the culture medium of granulosa cells from PCOS patients, as compared with non-PCOS patients. Ovaries from transgenic mice carrying the NGF gene targeted to thecal-interstitial cells by the 17alpha-hydroxylase gene promoter produce more NGF than wild-type (WT) ovaries and are hyperinnervated by sympathetic nerves. Antral follicle growth is arrested resulting in accumulation of intermediate size follicles, many of which are apoptotic. Peripubertal transgenic mice respond to a gonadotropin challenge with a greater increase in plasma 17-hydroxyprogesterone, estradiol, and testosterone levels than WT controls. Transgenic mice also exhibit a reduced ovulatory response, delayed puberty, and reduced fertility, as assessed by a prolonged interval between litters, and a reduced number of pups per litter. Sustained, but mild, elevation of plasma LH levels results in a heightened incidence of ovarian follicular cysts in transgenic mice as compared with WT controls. These results suggest that overproduction of ovarian NGF is a component of polycystic ovarian morphology in both humans and rodents and that a persistent elevation in plasma LH levels is required for the morphological abnormalities to appear.


Subject(s)
Nerve Growth Factor/metabolism , Ovarian Cysts/metabolism , Ovarian Cysts/pathology , Ovary/metabolism , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , 17-alpha-Hydroxyprogesterone/blood , Animals , Apoptosis/physiology , Cells, Cultured , Disease Models, Animal , Estradiol/blood , Female , Follicular Fluid/metabolism , Gonadotropins, Equine/pharmacology , Granulosa Cells/metabolism , Granulosa Cells/pathology , Humans , Mice , Mice, Transgenic , Nerve Growth Factor/genetics , Ovary/drug effects , Ovary/pathology , Puberty/physiology , RNA, Messenger/metabolism , Reproduction/physiology , Testosterone/blood
13.
Semin Reprod Med ; 27(1): 24-31, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19197802

ABSTRACT

Much is known about the endocrine hormonal mechanisms controlling ovarian development. More recently, attention has focused on identifying regulatory pathways that, operating within the ovarian microenvironment, contribute to the acquisition of ovarian reproductive competence. Within this framework, the concept has developed 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 maturation. The ovary of several species, including rodents, sheep, cows, nonhuman primates, and humans, produce NTs and express both the high-affinity receptors and the common p75 (NTR) receptor required for signaling. Studies in humans and rodents have shown that this expression is initiated during fetal life, before the formation of primordial follicles. Gene targeting approaches have identified TrkB, the high-affinity receptor for neurotrophin-4/5 and brain-derived neurotrophic factor, as a signaling module required for follicular assembly, early follicular growth, and oocyte survival. A similar approach has shown that nerve growth factor contributes independently to the growth of primordial follicles into gonadotropin-responsive structures. Altogether, these observations indicate that NTs are important contributors to the gonadotropin-independent process underlying the formation and initiation of ovarian follicular growth.


Subject(s)
Nerve Growth Factors/physiology , Ovary/embryology , Animals , Female , Humans , Models, Biological , Ovarian Follicle/physiology , Ovary/growth & development , Receptors, Nerve Growth Factor/physiology
14.
Development ; 134(5): 945-57, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17267443

ABSTRACT

In rodents, the formation of ovarian follicles occurs after birth. In recent years, several factors required for follicular assembly and the growth of the newly formed follicles have been identified. We now describe a novel gene, Fxna, identified by differential display in the neonatal rat ovary. Fxna encodes an mRNA of 5.4 kb, and a protein of 898 amino acids. Fxna is a transmembrane metallopeptidase from family M28, localized to the endoplasmic reticulum. In the ovary, Fxna mRNA is expressed in granulosa cells; its abundance is maximal 48 hours after birth, i.e. during the initiation of follicular assembly. Reducing Fxna mRNA levels via lentiviral-mediated delivery of short hairpin RNAs to neonatal ovaries resulted in substantial loss of primordial, primary and secondary follicles, and structural disorganization of the ovary, with many abnormal follicles containing more than one oocyte and clusters of somatic cells not associated with any oocytes. These abnormalities were not attributable to either increased apoptosis or decreased proliferation of granulosa cells. The results indicate that Fxna is required for the organization of somatic cells and oocytes into discrete follicular structures. As an endoplasmic reticulum-bound peptidase, Fxna may facilitate follicular organization by processing precursor proteins required for intraovarian cell-to-cell communication.


Subject(s)
Membrane Proteins/physiology , Metalloproteases/physiology , Ovary/growth & development , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Apoptosis , Base Sequence , Cell Proliferation , Endoplasmic Reticulum/metabolism , Female , Granulosa Cells/cytology , Granulosa Cells/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Metalloproteases/biosynthesis , Metalloproteases/genetics , Molecular Sequence Data , Oocytes/cytology , Oocytes/metabolism , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovary/metabolism , Rats , Rats, Sprague-Dawley , Sequence Homology, Amino Acid
15.
Endocrinology ; 147(1): 155-65, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16195402

ABSTRACT

The insulin receptor-related receptor (IRR) is a member of the insulin receptor family that, on its own, recognizes neither insulin nor any of the identified insulin-related peptides. In both the nervous system and peripheral tissues, IRR mRNA is detected in cells that also express trkA, the nerve growth factor tyrosine kinase receptor. In the ovary, the trkA gene is transiently activated in thecal-interstitial cells of large antral follicles at the time of the preovulatory surge of gonadotropins. The present study shows that the IRR gene is expressed in the same ovarian compartment, that IRR mRNA content increases strikingly in these cells in the afternoon of the first proestrus, and that--as in the case of trkA mRNA--the increase is caused by gonadotropins. The IRR mRNA species primarily affected is that encoding the full-length receptor; its increased abundance was accompanied by a corresponding change in IRR protein content. An extensive molecular search using several approaches, including the screening of cDNA libraries and PCR amplification with degenerate primers, did not yield an IRR ligand. Phylogenetic analysis of 20 insulin-related sequences and 15 relaxin family peptides from selected vertebrates indicated that the mammalian genome is unlikely to contain an additional ligand expressed from a distinct gene that is closely related to the insulin family. Although the functional nature of the relationship between IRR and trkA receptors is unknown, the remarkable temporal and spatial specificities of their coordinated expression in the ovary before ovulation suggests that they target a functionally related set of downstream events associated with the ovulatory process.


Subject(s)
Luteinizing Hormone/metabolism , Ovary/physiology , Proestrus/physiology , Receptor, Insulin/genetics , Theca Cells/physiology , Alternative Splicing , Animals , Base Sequence , Cloning, Molecular , DNA Primers , Female , Gene Expression Regulation , Genetic Variation , Ovary/cytology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptor, trkA/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
16.
Endocrinology ; 146(12): 5267-77, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16150897

ABSTRACT

In the rat ovary, germ and somatic cells become organized into primordial follicles 48-72 h after birth. Although several genes have been implicated in the control of early follicular growth, less is known about the factors involved in the formation of primordial follicles. Using the method of differential display of mRNAs, we found several genes differentially expressed at the time of follicular assembly. One of them encodes synaptonemal complex protein-1 (SCP1), a core component of the protein complex that maintains recombining chromosomes together during prophase I of the first meiotic division in germ cells. This association, evident during the pachytene stage, ends when chromosomal desynapsis begins in the diplotene stage at the end of prophase I. Oocytes become arrested in the diplotene/dictate stage before becoming enclosed into primordial follicles, suggesting that oocytes must complete meiotic prophase I before becoming competent to direct follicle assembly. We now show that attainment of the diplotene stage results in follicular formation. In developing rat ovaries, SCP1 mRNA expression is confined to oocytes and decreases precipitously within 24 h after birth, preceding the organization of primordial follicles. The premature loss of SCP1, achieved via treatment with an antisense oligodeoxynucleotide targeting SCP1 mRNA, resulted in more oocytes reaching the diplotene stage, as evidenced by a decrease in the number of oocytes containing germ cell nuclear antigen-1 (a nuclear protein whose expression ceases in diplotene) and an increase in the number of oocytes expressing MSY2 (a cytoplasmic Y box protein expressed in oocytes that have become arrested in diplotene). SCP1-deficient ovaries exhibited an increased number of newly formed follicles, suggesting that completion of meiotic prophase I endows oocytes with the ability to orchestrate follicular assembly.


Subject(s)
Nuclear Proteins/deficiency , Ovarian Follicle/physiology , Ovary/embryology , Ovary/growth & development , Animals , Animals, Newborn , DNA-Binding Proteins , Female , Fetal Development/drug effects , Fetus/drug effects , Fetus/metabolism , Meiosis , Meiotic Prophase I , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Oligonucleotides, Antisense/pharmacology , Oocytes/cytology , Oocytes/metabolism , Ovarian Follicle/embryology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...