Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Biomedicines ; 10(10)2022 Oct 07.
Article in English | MEDLINE | ID: mdl-36289767

ABSTRACT

This article reviews the main findings on anti-Müllerian hormone (AMH) and its involvement in the pathogenesis of polycystic ovary syndrome (PCOS) and its male equivalent. In women, AMH is produced by granulosa cells from the mid-fetal life to menopause and is a reliable indirect marker of ovarian reserve. AMH protects follicles from atresia, inhibits their differentiation in the ovary, and stimulates gonadotrophin-releasing hormone neurons pulsatility. AMH overexpression in women with PCOS likely contributes to the increase of the follicle cohort and of androgen levels, leading to follicular arrest and anovulation. In the male, AMH is synthesized at high levels by Sertoli cells from fetal life to puberty when serum AMH falls to levels similar to those observed in women. AMH is involved in the differentiation of the genital tract during fetal life and plays a role in Sertoli and Leydig cells differentiation and function. Serum AMH is used to assess Sertoli cell function in children with disorders of sex development and various conditions affecting the hypothalamic-pituitary-testicular axis. Although the reproductive function of male relative of women with PCOS has been poorly investigated, adolescents have elevated levels of AMH which could play a detrimental role on their fertility.

2.
J Biol Chem ; 298(1): 101429, 2022 01.
Article in English | MEDLINE | ID: mdl-34801555

ABSTRACT

Noncovalent complexes of transforming growth factor-ß family growth/differentiation factors with their prodomains are classified as latent or active, depending on whether the complexes can bind their respective receptors. For the anti-Müllerian hormone (AMH), the hormone-prodomain complex is active, and the prodomain is displaced upon binding to its type II receptor, AMH receptor type-2 (AMHR2), on the cell surface. However, the mechanism by which this displacement occurs is unclear. Here, we used ELISA assays to measure the dependence of prodomain displacement on AMH concentration and analyzed results with respect to the behavior expected for reversible binding in combination with ligand-induced receptor dimerization. We found that, in solution, the prodomain has a high affinity for the growth factor (GF) (Kd = 0.4 pM). Binding of the AMH complex to a single AMHR2 molecule does not affect this Kd and does not induce prodomain displacement, indicating that the receptor binding site in the AMH complex is fully accessible to AMHR2. However, recruitment of a second AMHR2 molecule to bind the ligand bivalently leads to a 1000-fold increase in the Kd for the AMH complex, resulting in rapid release of the prodomain. Displacement occurs only if the AMHR2 is presented on a surface, indicating that prodomain displacement is caused by a conformational change in the GF induced by bivalent binding to AMHR2. In addition, we demonstrate that the bone morphogenetic protein 7 prodomain is displaced from the complex with its GF by a similar process, suggesting that this may represent a general mechanism for receptor-mediated prodomain displacement in this ligand family.


Subject(s)
Anti-Mullerian Hormone , Peptide Hormones , Anti-Mullerian Hormone/metabolism , Ligands , Peptide Hormones/metabolism , Protein Domains , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism
3.
Genes (Basel) ; 12(11)2021 11 16.
Article in English | MEDLINE | ID: mdl-34828406

ABSTRACT

Down syndrome (DS) is the most common chromosomal disorder. It is responsible for intellectual disability (ID) and several medical conditions. Although men with DS are thought to be infertile, some spontaneous paternities have been reported. The few studies of the mechanism of infertility in men with DS are now dated. Recent research in zebrafish has indicated that overexpression of DYRK1A (the protein primarily responsible for ID in DS) impairs gonadogenesis at the embryonic stage. To better ascertain DYRK1A's role in infertility in DS, we investigated the effect of DYRK1A overexpression in a transgenic mouse model. We found that overexpression of DYRK1A impairs fertility in transgenic male mice. Interestingly, the mechanism in mice differs slightly from that observed in zebrafish but, with disruption of the early stages of spermatogenesis, is similar to that seen in humans. Unexpectedly, we observed hypogonadotropic hypogonadism in the transgenic mice.


Subject(s)
Hypogonadism/genetics , Infertility, Male/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Spermatogenesis , Animals , Hypogonadism/pathology , Infertility, Male/pathology , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Testis/embryology , Testis/pathology , Up-Regulation , Dyrk Kinases
4.
Endocr Rev ; 42(6): 753-782, 2021 11 16.
Article in English | MEDLINE | ID: mdl-33851994

ABSTRACT

Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance, was shown to be synthesized by the ovary in the 1980s. This article reviews the main findings of the past 20 years on the regulation of the expression of AMH and its specific receptor AMHR2 by granulosa cells, the mechanism of action of AMH, the different roles it plays in the reproductive organs, its clinical utility, and its involvement in the principal pathological conditions affecting women. The findings in respect of regulation tell us that AMH and AMHR2 expression is mainly regulated by bone morphogenetic proteins, gonadotropins, and estrogens. It has now been established that AMH regulates the different steps of folliculogenesis and that it has neuroendocrine effects. On the other hand, the importance of serum AMH as a reliable marker of ovarian reserve and as a useful tool in the prediction of the polycystic ovary syndrome (PCOS) and primary ovarian failure has also been acknowledged. Last but not least, a large body of evidence points to the involvement of AMH in the pathogenesis of PCOS.


Subject(s)
Peptide Hormones , Polycystic Ovary Syndrome , Anti-Mullerian Hormone/metabolism , Female , Granulosa Cells , Humans , Peptide Hormones/metabolism , Polycystic Ovary Syndrome/metabolism , Reproduction
5.
Nat Commun ; 12(1): 1064, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594056

ABSTRACT

Polycystic ovary syndrome (PCOS) is characterized by an oligo-anovulation, hyperandrogenism and polycystic ovarian morphology combined with major metabolic disturbances. However, despite the high prevalence and the human and economic consequences of this syndrome, its etiology remains unknown. In this study, we show that female Goto-Kakizaki (GK) rats, a type 2 diabetes mellitus model, encapsulate naturally all the reproductive and metabolic hallmarks of lean women with PCOS at puberty and in adulthood. The analysis of their gestation and of their fetuses demonstrates that this PCOS-like phenotype is developmentally programmed. GK rats also develop features of ovarian hyperstimulation syndrome. Lastly, a comparison between GK rats and a cohort of women with PCOS reveals a similar reproductive signature. Thus, this spontaneous rodent model of PCOS represents an original tool for the identification of the mechanisms involved in its pathogenesis and for the development of novel strategies for its treatment.


Subject(s)
Polycystic Ovary Syndrome/pathology , Adiposity , Animals , Animals, Newborn , Body Weight , Discriminant Analysis , Disease Models, Animal , Dyslipidemias/pathology , Endocrine System/pathology , Estrous Cycle , Female , Glucose Tolerance Test , Gonadotropins/pharmacology , Hormones/blood , Humans , Insulin Secretion , Least-Squares Analysis , Lipids/chemistry , Male , Maternal-Fetal Exchange , Multivariate Analysis , Ovary/pathology , Ovary/physiopathology , Phenotype , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/physiopathology , Pregnancy , Rats, Wistar , Reproduction , Sexual Maturation
6.
J Clin Endocrinol Metab ; 106(3): e1271-e1289, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33247926

ABSTRACT

PURPOSE: A protective effect of anti-Müllerian hormone (AMH) on follicle atresia was recently demonstrated using long-term treatments, but this effect has never been supported by mechanistic studies. This work aimed to gain an insight into the mechanism of action of AMH on follicle atresia and on how this could account for the increased follicle pool observed in women with polycystic ovary syndrome (PCOS). METHODS: In vivo and in vitro experiments were performed to study the effects of AMH on follicle atresia and on the proliferation and apoptosis of granulosa cells (GCs). RNA-sequencing was carried out to identify new AMH target genes in GCs. The expression of some of these genes in GCs from control and PCOS women was compared using microfluidic real time quantitative RT-PCR. RESULTS: A short-term AMH treatment prevented follicle atresia in prepubertal mice. Consistent with this result, AMH inhibited apoptosis and promoted proliferation of different models of GCs. Moreover, integrative biology analyses of 965 AMH target genes identified in 1 of these GC models, confirmed that AMH had initiated a gene expression program favoring cell survival and proliferation. Finally, on 43 genes selected among the most up- and down-regulated AMH targets, 8 were up-regulated in GCs isolated from PCOS women, of which 5 are involved in cell survival. MAIN CONCLUSIONS: Our results provide for the first time cellular and molecular evidence that AMH protects follicles from atresia by controlling GC survival and suggest that AMH could participate in the increased follicle pool of PCOS patients.


Subject(s)
Anti-Mullerian Hormone/pharmacology , Apoptosis , Granulosa Cells/drug effects , Polycystic Ovary Syndrome/pathology , Adult , Animals , Anti-Mullerian Hormone/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Case-Control Studies , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Granulosa Cells/pathology , Granulosa Cells/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism
7.
Sci Rep ; 10(1): 15062, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934281

ABSTRACT

Anti-Müllerian hormone (AMH) is secreted by Sertoli cells of the testes from early fetal life until puberty, when it is downregulated by androgens. In conditions like complete androgen insensitivity syndrome (CAIS), AMH downregulation does not occur and AMH increases at puberty, due in part to follicle-stimulating hormone (FSH) effect. However, other conditions like Peutz-Jeghers syndrome (PJS), characterised by low FSH, also have increased AMH. Because both CAIS and PJS may present as hyperoestrogenic states, we tested the hypothesis that oestradiol (E2) upregulates AMH expression in peripubertal Sertoli cells and explored the molecular mechanisms potentially involved. The results showed that E2 is capable of inducing an upregulation of endogenous AMH and of the AMH promoter activity in the prepubertal Sertoli cell line SMAT1, signalling through ERα binding to a specific ERE sequence present on the hAMH promoter. A modest action was also mediated through the membrane oestrogen receptor GPER. Additionally, the existence of ERα expression in Sertoli cells in patients with CAIS was confirmed by immunohistochemistry. The evidence presented here provides biological plausibility to the hypothesis that testicular AMH production increases in clinical conditions in response to elevated oestrogen levels.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Anti-Mullerian Hormone/metabolism , Estrogen Receptor alpha/biosynthesis , Neoplasm Proteins/biosynthesis , Peutz-Jeghers Syndrome/metabolism , Response Elements , Sertoli Cells/metabolism , Androgen-Insensitivity Syndrome/pathology , Animals , Cell Line , Child , Child, Preschool , Estradiol/metabolism , Female , Humans , Male , Mice , Peutz-Jeghers Syndrome/pathology , Sertoli Cells/pathology
8.
J Dev Orig Health Dis ; 11(2): 154-158, 2020 04.
Article in English | MEDLINE | ID: mdl-31309911

ABSTRACT

Epidemiological studies have demonstrated an increased risk of developing non-transmittable diseases in adults subjected to adverse early developmental conditions. Metabolic and cardiovascular diseases have been the focus of most studies. Nevertheless, data from animal models also suggest early programming of fertility. In humans, it is difficult to assess the impact of the in utero environment retrospectively. Birthweight is commonly used as an indirect indicator of intrauterine development. This research is part of the ALIFERT study. We investigated a potential link between ponderal index at birth and female fertility in adulthood. Data from 51 infertile and 74 fertile women were analysed. BW was on average higher in infertile women, whereas birth length did not differ between the two groups; thus, resulting in a significantly higher ponderal index at birth in infertile women. Ponderal index at birth has been identified as a risk factor for infertility. These results suggest the importance of the intra-uterine environment, not only for long-term metabolic health but also for fertility.


Subject(s)
Birth Weight/physiology , Body Height/physiology , Fetal Nutrition Disorders/epidemiology , Infertility, Female/epidemiology , Adolescent , Adult , Case-Control Studies , Female , Fertility/physiology , Fetal Nutrition Disorders/diagnosis , Fetal Nutrition Disorders/physiopathology , Humans , Infertility, Female/physiopathology , Pregnancy , Prospective Studies , Retrospective Studies , Risk Factors , Waist Circumference/physiology , Young Adult
9.
Oncogene ; 39(9): 1875-1890, 2020 02.
Article in English | MEDLINE | ID: mdl-31745296

ABSTRACT

Ovarian granulosa cell tumors (GCTs) are indolent tumors of the ovary affecting women at all ages and potentially displaying late recurrence. Even if there is still little information regarding the mechanisms involved in GCT development and progression, FOXL2 would be a major tumor suppressor gene in granulosa cells. We analyzed the mechanisms underlying GCT initiation and progression by using mice with targeted expression of SV40 large T-antigen in granulosa cells (AT mouse), which develop GCTs. Consistent with patients, AT mice with developing GCTs displayed increased levels in circulating anti-Müllerian hormone (AMH), estradiol and androgens, as well as decreased FOXL2 protein abundance. Very few mice developed metastases (1 out of 30). In situ analyses revealed that GCT initiation resulted from both increased granulosa cell survival and proliferation in large antral follicles. Tumorigenesis was associated with the combined inactivation of p53 and Rb pathways, as shown by the impaired expression of respective downstream targets regulating cell apoptosis and proliferation, i.e., Bax, Bak, Gadd45a, Ccna2, Ccne1, E2f1, and Orc1. Importantly, the expression of FOXL2 was still present in newly developed GCTs and its downregulation only started during GCT growth. Collectively, our experiments provide evidence that disrupted p53/Rb signaling can drive tumor initiation and growth. This model challenges the current paradigm that impaired FOXL2 signaling is a major switch of granulosa cell tumorigenesis, albeit possibly contributing to tumor growth.


Subject(s)
Carcinogenesis/pathology , Forkhead Box Protein L2/metabolism , Granulosa Cell Tumor/pathology , Granulosa Cells/pathology , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cells, Cultured , Down-Regulation , Female , Forkhead Box Protein L2/genetics , Granulosa Cell Tumor/genetics , Granulosa Cell Tumor/metabolism , Granulosa Cells/metabolism , Humans , Mice , Mice, Transgenic , Retinoblastoma Protein/genetics , Tumor Suppressor Protein p53/genetics
10.
Cell Mol Life Sci ; 77(6): 1177-1196, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31327046

ABSTRACT

In mammalian ovaries, the theca layers of growing follicles are critical for maintaining their structural integrity and supporting androgen synthesis. Through combining the postnatal monitoring of ovaries by abdominal magnetic resonance imaging, endocrine profiling, hormonal analysis of the follicular fluid of growing follicles, and transcriptomic analysis of follicular theca cells, we provide evidence that the exposure of ovine fetuses to testosterone excess activates postnatal follicular growth and strongly affects the functions of follicular theca in adulthood. Prenatal exposure to testosterone impaired androgen synthesis in the small antral follicles of adults and affected the expression in their theca cells of a wide array of genes encoding extracellular matrix components, their membrane receptors, and signaling pathways. Most expression changes were uncorrelated with the concentrations of gonadotropins, steroids, and anti-Müllerian hormone in the recent hormonal environment of theca cells, suggesting that these changes rather result from the long-term developmental effects of testosterone on theca cell precursors in fetal ovaries. Disruptions of the extracellular matrix structure and signaling in the follicular theca and ovarian cortex can explain the acceleration of follicle growth through altering the stiffness of ovarian tissue. We propose that these mechanisms participate in the etiology of the polycystic ovarian syndrome, a major reproductive pathology in woman.


Subject(s)
Polycystic Ovary Syndrome/metabolism , Prenatal Exposure Delayed Effects/metabolism , Testosterone/metabolism , Theca Cells/metabolism , Animals , Cells, Cultured , Female , Gene Expression Regulation , Gene Regulatory Networks , Humans , Ovarian Follicle/cytology , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Polycystic Ovary Syndrome/genetics , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Sheep , Theca Cells/cytology , Theca Cells/ultrastructure
11.
Hum Mol Genet ; 28(19): 3211-3218, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31238341

ABSTRACT

The persistent Müllerian duct syndrome (PMDS) is a 46,XY disorder of sexual development characterized by the persistence of Müllerian duct derivatives, uterus and tubes, in otherwise normally masculinized males. The condition, transmitted as a recessive autosomal trait, is usually due to mutations in either the anti-Müllerian hormone (AMH) gene or its main receptor. Many variants of these genes have been described, all targeting the coding sequences. We report the first case of PMDS due to a regulatory mutation. The AMH promoter contains two binding sites for steroidogenic factor 1 (SF1), one at -102 and the other at -228. Our patient carries a single base deletion at -225, significantly decreasing its capacity for binding SF1, as measured by the electrophoresis mobility shift assay. Furthermore, by linking the AMH promoter to the luciferase gene, we show that the transactivation capacity of the promoter is significantly decreased by the mutation, in contrast to the disruption of the -102 binding site. To explain the difference in impact we hypothesize that SF1 could partially overcome the lack of binding to the -102 binding site by interacting with a GATA4 molecule linked to a nearby response element. We show that disruption of both the -102 SF1 and the -84 GATA response elements significantly decreases the transactivation capacity of the promoter. In conclusion, we suggest that the distance between mutated SF1 sites and potentially rescuing GATA binding motifs might play a role in the development of PMDS.


Subject(s)
Anti-Mullerian Hormone/chemistry , Anti-Mullerian Hormone/metabolism , Disorder of Sex Development, 46,XY/genetics , Mutation , RNA Splicing Factors/metabolism , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Anti-Mullerian Hormone/genetics , Binding Sites/genetics , Cell Line , Child , Child, Preschool , Female , Humans , Infant, Newborn , Male , Pedigree , Promoter Regions, Genetic , Protein Binding
12.
Biol Reprod ; 99(6): 1303-1312, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29985989

ABSTRACT

Testicular anti-Müllerian hormone (AMH) production is inhibited by androgens around pubertal onset, as observed under normal physiological conditions and in patients with precocious puberty. In agreement, AMH downregulation is absent in patients with androgen insensitivity. The molecular mechanisms underlying the negative regulation of AMH by androgens remain unknown. Our aim was to elucidate the mechanisms through which androgens downregulate AMH expression in the testis. A direct negative effect of androgens on the transcriptional activity of the AMH promoter was found using luciferase reporter assays in the mouse prepubertal Sertoli cell line SMAT1. A strong inhibition of AMH promoter activity was seen in the presence of both testosterone and DHT and of the androgen receptor. By site-directed mutagenesis and chromatin immunoprecipitation assays, we showed that androgen-mediated inhibition involved the binding sites for steroidogenic factor 1 (SF1) present in the proximal promoter of the AMH gene. In this study, we describe for the first time the mechanism behind AMH inhibition by androgens, as seen in physiological and pathological conditions in males. Inhibition of AMH promoter activity by androgens could be due to protein-protein interactions between the ligand-bound androgen receptor and SF1 or by blockage of SF1 binding to its sites on the AMH promoter.


Subject(s)
Androgens/pharmacology , Anti-Mullerian Hormone/metabolism , Sertoli Cells/physiology , Steroidogenic Factor 1/metabolism , Animals , Anti-Mullerian Hormone/genetics , Cell Line , Chromatin Immunoprecipitation , Down-Regulation , Humans , Immunohistochemistry , Male , Mice , Promoter Regions, Genetic , Receptors, Androgen/metabolism , Steroidogenic Factor 1/genetics , Transcriptome
13.
J Clin Endocrinol Metab ; 102(11): 3970-3978, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28938480

ABSTRACT

Context: Anti-Müllerian hormone (AMH) and AMH type II receptor (AMHR2) are overexpressed in granulosa cells (GCs) from women with polycystic ovary syndrome (PCOS), the most common cause of female infertility. Objective: The aim of the study was to compare the regulation of the AMH/AMHR2 system by 5α-dihydrotestosterone (5α-DHT) and estradiol (E2) in GCs from control subjects and women with PCOS. Design, Setting, Patients: Experiments were performed on follicular fluids (FF) and GCs from women undergoing in vitro fertilization. Main Outcome Measures: FF steroid levels were measured by mass spectrometry, and messenger RNA (mRNA) accumulation was quantified by reverse transcription real-time polymerase chain reaction. Results: Total testosterone (T), free T, and 5α-DHT FF levels were significantly higher (P < 0.001) in women with PCOS than in controls. However, E2 and sex hormone-binding globulin concentrations were comparable between the two groups. In GCs from control women, the AMH and AMHR2 expression were not affected by 5α-DHT treatment, whereas AMH mRNA levels were upregulated by 5α-DHT in GCs from patients with PCOS (2.3-fold, P < 0.01) overexpressing the androgen receptor (1.4-fold, P < 0.05). E2 downregulated the AMH and AMHR2 expression in GCs from control women (1.4-fold, P < 0.001 and 1.8-fold, P < 0.01, respectively) but had no effect on these genes in GCs from women with PCOS. This differential effect of E2 was associated with a higher estrogen receptor 1 expression in GCs from women with PCOS (1.9-fold, P < 0.05). Conclusions: In GCs from women with PCOS, the regulation of AMH and AMHR2 expression is altered in a way that promotes the overexpression of the AMH/AMHR2 system, and could contribute to the follicular arrest observed in these patients.


Subject(s)
Anti-Mullerian Hormone/genetics , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Polycystic Ovary Syndrome/genetics , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Adult , Anti-Mullerian Hormone/metabolism , Case-Control Studies , Dihydrotestosterone/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Follicular Phase/drug effects , Follicular Phase/genetics , Follicular Phase/metabolism , Gene Expression Regulation/drug effects , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Humans , Polycystic Ovary Syndrome/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Young Adult
14.
J Clin Endocrinol Metab ; 101(12): 4618-4627, 2016 12.
Article in English | MEDLINE | ID: mdl-27623067

ABSTRACT

CONTEXT: Anti-Müllerian hormone (AMH) is an important clinical marker for diagnosing and assessing the reproductive status and/or disorders in men and women. Most studies have not distinguished between levels of inactive AMH precursor and the cleaved noncovalent complex that binds the AMH type II receptor (AMHRII) and initiates signaling. OBJECTIVE: The objective of the study was to measure the levels of AMH cleavage and bioactivity in human body fluids. DESIGN, SETTING, AND PATIENTS: AMH cleavage levels and bioactivity were measured in the serum of six boys and in the follicular fluid and serum of nine control women and 13 women with the polycystic ovary syndrome (PCOS). MAIN OUTCOME MEASURES: AMH cleavage levels were measured by capturing AMH with an anti-AMH antibody, followed by Western blotting. The bioactivity of cleaved AMH was assessed with an ELISA that measures the levels of AMH capable of binding AMHRII. RESULTS: PCOS women have an elevated level of AMH cleavage in their follicular fluid (24% vs 8% in control women), and most of the cleaved AMH can bind AMHRII. Higher levels of cleavage are observed in female (60%) and male (79%) serum, but very little of the cleaved AMH can bind AMHRII. CONCLUSIONS: These results support an autocrine role for AMH in the pathophysiology of PCOS in the follicle. In addition, they indicate that AMH undergoes interactions or structural changes after cleavage that prevent receptor binding, meaning, unexpectedly, that the level of cleaved AMH in biological fluids does not always reflect the level of bioactive AMH.


Subject(s)
Anti-Mullerian Hormone/metabolism , Follicular Fluid/metabolism , Polycystic Ovary Syndrome/metabolism , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Adult , Anti-Mullerian Hormone/blood , Child , Female , Humans , Male , Polycystic Ovary Syndrome/blood , Protein Binding
15.
Sci Rep ; 6: 23790, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27030385

ABSTRACT

Anti-Müllerian hormone (AMH) contributes to male sexual differentiation and acts on gonads of both sexes. Identification of AMH receptivity in both pituitary and brain has led to the intriguing idea that AMH participates to the hypothalamic-pituitary control of reproduction, however in vivo experimental evidence is still lacking. We show that AMH stimulates secretion and pituitary gene expression of the gonadotropin FSH in vivo in rats. AMH action is sex-dependent, being restricted to females and occurring before puberty. Accordingly, we report higher levels of pituitary AMH receptor transcripts in immature females. We show that AMH is functionally coupled to the Smad pathway in LßT2 gonadotrope cells and dose-dependently increases Fshb transcript levels. Furthermore, AMH was shown to establish complex interrelations with canonical FSH regulators as it cooperates with activin to induce Fshb expression whereas it reduces BMP2 action. We report that GnRH interferes with AMH by decreasing AMH receptivity in vivo in females. Moreover, AMH specifically regulates FSH and not LH, indicating that AMH is a factor contributing to the differential regulation of gonadotropins. Overall, our study uncovers a new role for AMH in regulating gonadotrope function and suggests that AMH participates in the postnatal elevation of FSH secretion in females.


Subject(s)
Anti-Mullerian Hormone/genetics , Follicle Stimulating Hormone/genetics , Gonadotrophs/metabolism , Pituitary Gland, Anterior/metabolism , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Sex Characteristics , Activins/genetics , Activins/metabolism , Animals , Animals, Newborn , Anti-Mullerian Hormone/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cell Line , Female , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation, Developmental , Gonadotrophs/cytology , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Male , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/growth & development , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Sexual Maturation , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism
16.
J Clin Endocrinol Metab ; 101(6): 2602-11, 2016 06.
Article in English | MEDLINE | ID: mdl-27070094

ABSTRACT

CONTEXT: Anti-Müllerian hormone (AMH) is produced by the granulosa cells (GCs) of growing follicles and inhibits follicular development. OBJECTIVE: This study aimed to investigate the regulation of the AMH-specific type 2 receptor (AMHR2) gene expression in GCs by bone morphogenetic protein (BMP)15, BMP4 and growth differentiation factor (GDF)9. DESIGN, SETTING, AND PATIENTS: Their effects on AMHR2 and AMH mRNAs were studied in luteinized human GCs and in ovine GCs (oGCs) from small antral follicles. The effects of BMPs on human AMHR2 and AMH promoter reporter activities were analyzed in transfected oGCs. The in vivo effect of BMP15 on GCs AMHR2 and AMH expression was investigated by using Lacaune and Rasa Aragonesa hyperprolific ewes carrying loss-of-function mutations in BMP15. MAIN OUTCOME MEASURES: mRNAs were quantified by real-time RT-PCR. Promoter reporter constructs activities were quantified by the measurement of their luciferase activity. RESULTS: BMP15 and BMP4 enhanced AMHR2 and AMH expression in human GCs and in oGCs, whereas GDF9 had no effect. In oGCs, GDF9 increased BMP15 effect on AMH expression. Consistent with these results, BMP15 and BMP4, but not GDF9, enhanced AMHR2 promoter activity in oGCs, whereas GDF9 increased BMP15 effect on AMH promoter activity. Moreover, oGCs from both BMP15 mutant ewes had reduced AMHR2 mRNA levels but unchanged AMH expression compared with wild-type ewes. CONCLUSIONS: Altogether, these results suggest that the mechanisms of action of BMP15 on AMHR2 and AMH expression are different, and that by stimulating AMHR2 and AMH expression in GCs BMP15 enhances AMH inhibitory actions in GCs.


Subject(s)
Bone Morphogenetic Protein 15/pharmacology , Gene Expression Regulation/drug effects , Granulosa Cells/drug effects , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Up-Regulation/drug effects , Adult , Animals , Bone Morphogenetic Protein 4/pharmacology , Female , Granulosa Cells/metabolism , Growth Differentiation Factor 9/pharmacology , Humans , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Promoter Regions, Genetic/drug effects , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Sheep , Young Adult
18.
Theriogenology ; 84(8): 1397-404, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26298408

ABSTRACT

Plasma anti-Müllerian hormone (AMH) concentrations have been recently found to be predictive of the number of embryos recovered after FSH superovulatory treatment in the cow. However, the sensitivity of the Active Müllerian-inhibiting substance/AMH ELISA (ref. 10-14400; DSL-Beckman-Coulter) used to make these measurements in bovine plasma samples is low because it was developed to measure human AMH levels. To overcome this limitation, we developed an immunoassay specific for the bovine (B), ovine (O), and caprine (C) species, the bovine-ovine-caprine (BOC) ELISA. For this purpose, we produced recombinant bovine AMH for standardization, and we used monoclonal antibodies raised against bovine AMH, previously prepared by our laboratory. We evaluated the precision, accuracy, specificity, limit of detection, and functional sensitivity of the assay. The intra-assay coefficient of variation ranged between 3.4% and 11.3% for AMH concentrations between 23.68 and 1.74 ng/mL, and the interassay coefficient of variation ranged between 4.8% and 20.5% for concentrations between 25.53 and 1.42 ng/mL, respectively. The assay displayed a good linearity, had a detection limit of 0.4 ng/mL and a functional sensitivity of 1.4 ng/mL. It also cross-reacted with ovine and caprine AMHs. Both the mean and median AMH levels measured in 40 cow plasma samples using the BOC ELISA were approximately 44 fold higher than the mean and median AMH levels measured with the Active Müllerian-inhibiting substance/AMH ELISA. Moreover, a higher correlation was observed between the average number of embryos recovered from each cow after superovulatory treatment and AMH concentrations measured with the BOC ELISA. This BOC ELISA provides a very efficient tool for evaluating the ovarian follicular reserve of cows and predicting their embryo production capacity.


Subject(s)
Anti-Mullerian Hormone/blood , Embryonic Development , Enzyme-Linked Immunosorbent Assay/veterinary , Animals , Cattle , Female , Ovarian Function Tests/methods , Ovarian Function Tests/veterinary , Sensitivity and Specificity
19.
J Cell Sci ; 128(7): 1352-64, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25663701

ABSTRACT

The levels and intracellular localization of wild-type transforming growth factor ß superfamily (TGFß-SF) receptors are tightly regulated by endocytic trafficking, shedding and degradation. In contrast, a main regulatory mechanism of mutation-bearing receptors involves their intracellular retention. Anti-Müllerian hormone receptor II (AMHRII, also known as AMHR2) is the type-II receptor for anti-Müllerian hormone (AMH), a TGFß-SF ligand that mediates Müllerian duct regression in males. Here, we studied AMHRII processing and identified novel mechanisms of its constitutive negative regulation. Immunoblot analysis revealed that a significant portion of AMHRII was missing most of its extracellular domain (ECD) and, although glycosylated, was unfolded and retained in the endoplasmic reticulum. Exogenous expression of AMHRII, but not of type-II TGF-ß receptor (TßRII, also known as TGFR2), resulted in its disulfide-bond-mediated homo-oligomerization and intracellular retention, and in a decrease in its AMH-binding capacity. At the plasma membrane, AMHRII differed from TßRII, forming high levels of non-covalent homomeric complexes, which exhibited a clustered distribution and restricted lateral mobility. This study identifies novel mechanisms of negative regulation of a type-II TGFß-SF receptor through cleavage, intracellular retention and/or promiscuous disulfide-bond mediated homo-oligomerization.


Subject(s)
Protein Processing, Post-Translational , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Animals , Anti-Mullerian Hormone/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Humans , Male , Mice , Protein Binding , Protein Folding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Receptor, Transforming Growth Factor-beta Type II , Receptors, Peptide/chemistry , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
20.
Endocrinology ; 156(1): 301-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25322464

ABSTRACT

In the ovary, anti-Müllerian hormone (AMH) is produced by the granulosa cells of growing follicles and can modulate the recruitment of primordial follicles and the FSH-dependent development of follicles. However, the regulation of its production remains poorly understood. Recently, a stimulating effect of the bone morphogenetic proteins (BMPs) on AMH production by granulosa cells has been shown in vitro, but the molecular mechanisms implicated in this regulation and its physiological importance in ovarian function have not yet been established. In the hyperprolific Booroola ewes carrying the FecB(B) partial loss-of-function mutation in the fecundity gene encoding the FecB/BMP receptor, type 1B, the granulosa cells of antral follicles expressed and secreted low AMH amounts, resulting in low AMH concentrations in blood, despite high numbers of AMH-secreting follicles in ovaries. The presence of the FecB(B) mutation impaired the granulosa cell response to the stimulating action of BMP4 on AMH production, indicating a crucial role of the BMP receptor, type 1B in AMH regulation. In ovine granulosa cells, BMP4 enhanced the transcriptional activity of the human AMH promoter, and this action depended on the presence of SMAD1, acting on a promoter sequence located between -423 and -202 bp upstream of the AMH transcription start site. SMAD1 and SF1 acted in concert to mediate BMP4 action on the AMH promoter. Among the 2 SF1 binding sites present on the AMH promoter, the most proximal site, located at -92 bp upstream of the AMH transcription start site, was found to be critical for ensuring the response of the AMH promoter to BMP4. In conclusion, AMH could mediate the actions of BMPs in regulating follicular development and contributing to the determination of ovulation numbers. A molecular model of regulation of the AMH promoter transactivation by BMP signaling is proposed.


Subject(s)
Anti-Mullerian Hormone/metabolism , Bone Morphogenetic Protein 4/metabolism , Gene Expression Regulation/physiology , Granulosa Cells/metabolism , Sheep/physiology , Animals , Anti-Mullerian Hormone/genetics , Binding Sites , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cells, Cultured , Female , Genotype , Humans , Mutation , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...