Stanford Home
Ovarian Kaleidoscope Database (OKdb)

Home

History

Transgenic Mouse Models

INFORGRAPHICS

Search
Submit
Update
Chroms
Browse
Admin

Hsueh lab

HPMR

Visits
since 01/2001:
176557

anti-Mullerian hormone OKDB#: 850
 Symbols: AMH Species: human
 Synonyms: MIF, MIS  Locus: 19p13.3 in Homo sapiens


For retrieval of Nucleotide and Amino Acid sequences please go to: OMIM Entrez Gene
Mammalian Reproductive Genetics   Endometrium Database Resource   Orthologous Genes   UCSC Genome Browser   GEO Profiles new!   Amazonia (transcriptome data) new!

R-L INTERACTIONS   MGI

DNA Microarrays
SHOW DATA ...
link to BioGPS
General Comment Stage-dependent actions of antimüllerian hormone in regulating granulosa cell proliferation and follicular function in the primate ovary. Xu F et al. (2021) To study the direct action and physiological role of antimüllerian hormone (AMH) in regulating ovarian follicular development and function in vivo in primates. Animals were assigned to six treatment sequences in a crossover design study. Intraovarian infusion was performed during the follicular phase of the menstrual cycle with agents including: control vehicle; recombinant human AMH (rhAMH); and neutralizing anti-human AMH antibody (AMHAb). Before ovariectomy after the final treatment, the animals received intravenous injections of bromodeoxyuridine (BrdU). National primate research center. Adult female rhesus macaques (Macaca mulatta). None. Cycle length, follicle cohorts, and serum steroid levels were assessed. Ovarian histology, as well as granulosa cell (GC) proliferation and oocyte viability, were evaluated. In vehicle-infused ovaries, a dominant follicle was observed at midcycle E2 peak. However, rhAMH-treated ovaries exhibited an increased number of small antral follicles, whereas AMHAb-treated ovaries developed multiple large antral follicles. Serum E2 levels in the follicular phase decreased after rhAMH infusion and increased after AMHAb infusion. The rhAMH infusion increased serum T levels. Whereas early-growing follicles of rhAMH-treated ovaries contained BrdU-positive GCs, antral follicles containing BrdU-positive GCs were identified in AMHAb-treated ovaries. Autophagy was observed in oocytes of early-growing and antral follicles exposed to AMHAb and rhAMH, respectively. AMH enhanced early-stage follicle growth, but prevented antral follicle development and function via its stage-dependent regulation of GC proliferation and oocyte viability. This study provides information relevant to the pathophysiology of ovarian dysfunction and the treatment of infertility.//////////////////Anti-Müllerian hormone is a survival factor and promotes the growth of rhesus macaque preantral follicles during matrix-free culture. Xu J et al. (2018) Anti-Müllerian hormone (AMH) plays a key role during ovarian follicular development, with local actions associated with a dynamic secretion profile by growing follicles. While results for AMH effects on antral follicle growth and function are consistent among studies in various species, any effects on preantral follicle development remain controversial. Therefore, experiments were conducted to investigate the direct actions and role of AMH during follicle development at the preantral stage. Macaque-specific short-hairpin RNAs (shRNAs) targeting AMH mRNA were incorporated into adenoviral vectors to decrease AMH gene expression in rhesus macaque follicles. Secondary follicles were isolated from adult macaque ovaries and cultured individually in the ultra-low-attachment dish containing defined medium supplemented with follicle-stimulating hormone and insulin for 5 weeks. Follicles were randomly assigned to treatment groups: (a) control, (b) non-targeting control shRNA-vector, (c) AMH shRNA-vector, (d) AMH shRNA-vector + recombinant human AMH, and (e) recombinant human AMH. Follicle survival and growth were assessed. Culture media were analyzed for steroid hormone and paracrine factor concentrations. For in vivo study, the non-targeting control shRNA-vector and AMH shRNA-vector were injected into macaque ovaries. Ovaries were collected 9 days post-injection for morphology and immunohistochemistry assessment. Decreased AMH expression reduced preantral follicle survival and growth in nonhuman primates. Supplemental AMH treatment in the culture media promoted preantral follicle growth to the small antral stage in vitro with increased steroid hormone and paracrine factor production, as well as oocyte maturation. These data demonstrate that AMH is a critical follicular paracrine/autocrine factor positively impacting preantral follicle survival and growth in primates.Anti-Müllerian hormone production in the ovary: a comparative study in bovine and porcine granulosa cells†. Estienne A et al. (2020) In this study, we aimed to determine the origin of the difference, in terms of anti-Müllerian hormone production, existing between the bovine and porcine ovaries. We first confirmed by quantitative real-time-Polymerase-Chain Reaction, ELISA assay and immunohistochemistry that anti-Müllerian hormone mRNA and protein production are very low in porcine ovarian growing follicles compared to bovine ones. We then have transfected porcine and bovine granulosa cells with vectors containing the luciferase gene driven by the porcine or the bovine anti-Müllerian hormone promoter. These transfection experiments showed that the porcine anti-Müllerian hormone promoter is less active and less responsive to bone morphogenetic protein stimulations than the bovine promoter in both porcine and bovine cells. Moreover, bovine but not porcine granulosa cells were responsive to bone morphogenetic protein stimulation after transfection of a plasmidic construction including a strong response element to the bone morphogenetic proteins (12 repetitions of the GCCG sequence) upstream of the luciferase reporter gene. We also showed that SMAD6, an inhibitor of the SMAD1-5-8 pathway, is strongly expressed in porcine compared to the bovine granulosa cells. Overall, these results suggest that the low expression of anti-Müllerian hormone in porcine growing follicles is due to both a lack of activity/sensitivity of the porcine anti-Müllerian hormone promoter, and to the lack of responsiveness of porcine granulosa cells to bone morphogenetic protein signaling, potentially due to an overexpression of SMAD6 compared to bovine granulosa cells. We propose that the low levels of anti-Müllerian hormone in the pig would explain the poly-ovulatory phenotype in this species.//////////////////////////////////// AMH/MIS as a contraceptive that protects the ovarian reserve during chemotherapy. Kano M et al. (2017) The ovarian reserve represents the stock of quiescent primordial follicles in the ovary which is gradually depleted during a woman's reproductive lifespan, resulting in menopause. Müllerian inhibiting substance (MIS) (or anti-Müllerian hormone/AMH), which is produced by granulosa cells of growing follicles, has been proposed as a negative regulator of primordial follicle activation. Here we show that long-term parenteral administration of superphysiological doses of MIS, using either an adeno-associated virus serotype 9 (AAV9) gene therapy vector or recombinant protein, resulted in a complete arrest of folliculogenesis in mice. The ovaries of MIS-treated mice were smaller than those in controls and did not contain growing follicles but retained a normal ovarian reserve. When mice treated with AAV9/MIS were paired with male breeders, they exhibited complete and permanent contraception for their entire reproductive lifespan, disrupted vaginal cycling, and hypergonadotropic hypogonadism. However, when ovaries from AAV9-MIS-treated mice were transplanted orthotopically into normal recipient mice, or when treatment with the protein was discontinued, folliculogenesis resumed, suggesting reversibility. One of the important causes of primary ovarian insufficiency is chemotherapy-induced primordial follicle depletion, which has been proposed to be mediated in part by increased activation. To test the hypothesis that MIS could prevent chemotherapy-induced overactivation, mice were given carboplatin, doxorubicin, or cyclophosphamide and were cotreated with AAV9-MIS, recombinant MIS protein, or vehicle controls. We found significantly more primordial follicles in MIS-treated animals than in controls. Thus treatment with MIS may provide a method of contraception with the unique characteristic of blocking primordial follicle activation that could be exploited to prevent the primary ovarian insufficiency often associated with chemotherapy.//////////////////A putative role for anti-Müllerian hormone (AMH) in optimising ovarian reserve expenditure. Pankhurst MW et al. (2017) The mammalian ovary has a finite supply of oocytes which are contained within primordial follicles where they are arrested in a dormant state. The number of primordial follicles in the ovary at puberty is highly variable between females of the same species. Females that enter puberty with a small ovarian reserve are at risk of a shorter reproductive lifespan, as their ovarian reserve is expected to be depleted faster. One of the roles of anti-Müllerian hormone (AMH) is to inhibit primordial follicle activation which slows the rate at which the ovarian reserve is depleted. A simple interpretation is that the function of AMH is to conserve ovarian reserve. However, the females with the lowest ovarian reserve, and greatest risk of early reserve depletion, have the lowest levels of AMH. In contrast, AMH apparently strongly inhibits primordial follicle activation in females with ample ovarian reserve, for reasons that remain unexplained. The rate of primordial follicle activation determines the size of the developing follicle pool which in turn, determines how many oocytes are available to be selected for ovulation. This review discusses the evidence that AMH regulates the size of the developing follicle pool by altering the rate of primordial follicle activation in a context-dependent manner. The expression patterns of AMH across life are also consistent with changing requirements for primordial follicle activation in the aging ovary. A potential role of AMH in the fertility of aging females, is proposed herein.////////////////// OvAge: a new methodology to quantify ovarian reserve combining clinical, biochemical and 3D-ultrasonographic parameters. Venturella R et al. (2015) In the last decade, both endocrine and ultrasound data have been tested to verify their usefulness for assessing ovarian reserve, but the ideal marker does not yet exist. The purpose of this study was to find, if any, a statistical advanced model able to identify a simple, easy to understand and intuitive modality for defining ovarian age by combining clinical, biochemical and 3D-ultrasonographic data. This is a population-based observational study. From January 2012 to March 2014, we enrolled 652 healthy fertile women, 29 patients with clinical suspect of premature ovarian insufficiency (POI) and 29 patients with Polycystic Ovary syndrome (PCOS) at the Unit of Obstetrics & Gynecology of Magna Graecia University of Catanzaro (Italy). In all women we measured Anti Müllerian Hormone (AMH), Follicle Stimulating Hormone (FSH), Estradiol (E2), 3D Antral Follicle Count (AFC), ovarian volume, Vascular Index (VI) and Flow Index (FI) between days 1 and 4 of menstrual cycle. We applied the Generalized Linear Models (GzLM) for producing an equation combining these data to provide a ready to use information about women ovarian reserve, here called OvAge. To introduce this new variable, expression of ovarian reserve, we assumed that in healthy fertile women ovarian age is identical to chronological age. GzLM applied on the healthy fertile controls dataset produced the following equation OvAge = 48.05 - 3.14*AHM + 0.07*FSH - 0.77*AFC - 0.11*FI + 0.25*VI + 0.1*AMH*AFC + 0.02*FSH*AFC. This model showed a high statistical significance for each marker included in the equation. We applied the final equation on POI and PCOS datasets to test its ability of discovering significant deviation from normality and we obtained a mean of predicted ovarian age significantly different from the mean of chronological age in both groups. OvAge is one of the first reliable attempt to create a new method able to identify a simple, easy to understand and intuitive modality for defining ovarian reserve by combining clinical, biochemical and 3D-ultrasonographic data. Although design data prove a statistical high accuracy of the model, we are going to plan a clinical validation of model reliability in predicting reproductive prognosis and distance to menopause.////////////////// Anti-Mullerian hormone (AMH) and its receptor are involved in the regression of Mullerian ducts in male fetuses. Male sex differentiation is mediated by 2 discrete hormones produced by the fetal testis. Testosterone, produced by Leydig cells, virilizes the external genitalia and promotes prostatic growth; anti-Mullerian hormone (AMH), also called Mullerian-inhibiting substance (MIS) or factor (MIF), results in regression of Mullerian ducts which would otherwise differentiate into the uterus and fallopian tubes. Because MIF is also used as the symbol for macrophage migration inhibitory factor, AMH, for anti-Mullerian hormone, will be considered the preferred symbol. Rico C et al. Anti-M?an hormone (AMH) is an endocrine marker which can help predict superovulatory responses to treatments administered to cows for embryo production. However, the optimal time of the estrous cycle at which a blood test should be performed for a highly reliable prognosis has not been established yet. Moreover, little is known about the regulation of AMH production. To answer these questions, a study was designed to investigate the regulation of AMH production in cows selected for their high or low ovulatory responses to superovulation. At the granulosa cell level, AMH production was inhibited by FSH but enhanced by bone morphogenetic proteins. At the follicular level, the expression of AMH within the follicle was dependent on the stage of follicular development. At the ovarian level, the size of the pool of small antral growing follicles determined ovarian AMH production. At the endocrine level, AMH followed a specific dynamic profile during the estrous cycle, which occurred independently of the follicular waves of terminal follicular development. Cows selected for their high or low responses to superovulation did not differ in the regulation of AMH production but cows with higher responses had higher AMH concentrations in plasma throughout the cycle. The optimal period of the estrous cycle to measure AMH concentrations with the aim of selecting the best cows for embryo production was found to be at estrus and after day twelve of the cycle. From this multi-scale study, we propose a model that integrates the different regulatory levels of AMH production. The physiology and clinical utility of anti-Mullerian hormone in women. Dewailly D 2014 et al. BACKGROUNDThe measurement of circulating anti-M?an hormone (AMH) has been applied to a wide array of clinical applications, mainly based on its ability to reflect the number of antral and pre-antral follicles present in the ovaries. AMH has been suggested to predict the ovarian response to hyperstimulation of the ovaries for IVF and the timing of menopause, and to indicate iatrogenic damage to the ovarian follicle reserve. It has also been proposed as a surrogate for antral follicle count (AFC) in the diagnosis of polycystic ovary syndrome (PCOS).METHODSThis paper is a summary of presentations at a European Society of Human Reproduction and Embryology campus workshop on AMH, with literature cited until September 2013. Published peer-reviewed medical literature about AMH was searched through MEDLINE and was subjected to systematic review and critical assessment by the panel of authors.RESULTSPhysiologically, recent data confirm that AMH is a follicular gatekeeper limiting follicle growth initiation, and subsequently estradiol production from small antral follicles prior to selection. AMH assays continue to evolve and technical issues remain; the absence of an international standard is a key issue. The dynamics of circulating AMH levels throughout life can be split into several distinct phases, with a peak in the early 20s before a decline to the menopause, with a strong and positive correlation with non-growing follicle recruitment. There is a more complex rise during childhood and adolescence, which is likely to be more reflective of different stages of follicle development. AMH shows limited short-term variability, but the influence of states such as prolonged oral contraceptive use need to be considered in clinical assessment. There are only very limited data on relationships between AMH and natural fertility at different stages of reproductive life, and while it has a relationship to age at menopause the marked variability in this needs further exploration. AMH may be useful in assessing the need for fertility preservation strategies and detecting post-chemotherapy or surgical damage to the ovarian reserve. Long-term follow-up of patients to ascertain fully the value of post-cancer serum AMH in predicting long-term ovarian function is required. There is a linear relationship between AMH and oocyte yield after ovarian stimulation, which is of value in predicting ovarian hyperstimulation. AMH can also identify 'poor responders', but it seems inappropriate at present to withhold IVF purely on this basis. Women with PCOS show markedly raised AMH levels, due to both the increased number of small antral follicles and intrinsic characteristics of those granulosa cells, and this may contribute to anovulation. The value of AMH in the diagnosis of PCOS remains controversial, but it may replace AFC in the future.CONCLUSIONSFor the first time in female reproductive biology, it is possible to measure the submerged part of the iceberg of follicle growth, i.e. the intrinsic, so-called 'acyclic' ovarian activity. An international standard for AMH and improved assay validity are urgently needed to maximize the clinical utility of this very promising biomarker of ovarian function in a large array of clinical situations, both in childhood and adulthood. /////////////////////////Relationship between anti-Müllerian hormone and antral follicle count across the menstrual cycle using the Beckman Coulter Access assay in comparison with Gen II manual assay. Schiffner J et al. (2016) The study aim was to validate Beckman Coulter's fully automated Access Immunoassay System (BC Access assay) for anti-Müllerian hormone (AMH) and compare it with Beckman Coulter's Modified Manual Generation II assay (BC Mod Gen II), with regard to cycle AMH fluctuations and antral follicle counts. During one complete menstrual cycle, transvaginal ultrasound was performed on regularly menstruating women (n=39; 18-40years) every 2 days until the dominant ovarian follicle reached 16mm, then daily until observed ovulation; blood samples were collected throughout the cycle. Number and size of antral follicles was determined and AMH levels measured using both assays. AMH levels measured by the BC Access assay vary over ovulatory menstrual cycles, with a statistically significant pre-ovulatory decrease from -5 to +2 days around objective ovulation. Mean luteal AMH levels were significantly lower (-7.99%) than mean follicular levels but increased again towards the end of the luteal phase. Antral follicle count can be estimated from AMH (ng/mL, BC Access assay) concentrations on any follicular phase day. BC Access assay-obtained AMH values are considerably lower compared with the BC Mod Gen II assay (-19% on average); conversion equation: AMH BC Access (ng/mL)=0.85 [AMH BC Mod Gen II (ng/mL)]0.95. AMH levels vary throughout the cycle, independently of assay utilised. A formula can be used to convert BC Access assay-obtained AMH levels to BC Mod Gen II values. The number of antral follicles can be consistently estimated from pre-ovulatory AMH levels using either assay.//////////////////

NCBI Summary: This gene encodes a secreted ligand of the TGF-beta (transforming growth factor-beta) superfamily of proteins. Ligands of this family bind various TGF-beta receptors leading to recruitment and activation of SMAD family transcription factors that regulate gene expression. The encoded preproprotein is proteolytically processed to generate N- and C-terminal cleavage products that homodimerize and associate to form a biologically active noncovalent complex. This complex binds to the anti-Mullerian hormone receptor type 2 and causes the regression of Mullerian ducts in the male embryo that would otherwise differentiate into the uterus and fallopian tubes. This protein also plays a role in Leydig cell differentiation and function and follicular development in adult females. Mutations in this gene result in persistent Mullerian duct syndrome. [provided by RefSeq, Jul 2016]
General function Ligand, Hormone
Comment Serum anti-Mullerian hormone levels after ovarian drilling for the second-line treatment of polycystic ovary syndrome: a pilot-randomized study comparing laparoscopy and transvaginal hydrolaparoscopy. Giampaolino P et al. (2016) Aim of the study was to asses and compare serum anti-Mullerian harmone (AMH) levels after laparoscopic ovarian drilling (LOD) and transvaginal hydrolaparoscopy (THL) ovarian drilling in clomifene citrate (CC)-resistant polycystic ovary syndrome (PCOS) patients; secondary outcome was to evaluate postoperative pain to estimate the acceptability of procedures. A total of 246 patients with CC-resistant PCOS were randomized into two groups: 123 underwent LOD and 123 underwent THL ovarian drilling. AMH serum levels were evaluated before and after the procedure; moreover, women were asked to rate pain on a visual analog scale (VAS) from 0 (no pain, perfectly acceptable) to 10 (unbearable pain, completely unacceptable). In both groups, postoperative serum AMH levels were significantly reduced compared to preoperative levels (6.06 ± 1.18 and 5.84 ± 1.16 versus 5.00 ± 1.29 and 4.83 ± 1.10; p < 0.0001). Comparing postoperative serum AMH levels, no statistically significant difference was observed between the two surgical technique. After the procedure, mean pain VAS score was significantly higher for women who underwent LOD ovarian drilling in comparison to THL (3.26 ± 1.1 versus 1.11 ± 0.5; p < 0.0001). In conclusion, THL ovarian drilling is comparable to the LOD in terms of reduction in AMH, but it is preferred by patients in terms of acceptability. These results could support to use of THL ovarian drilling in the treatment of patients with CC- resistant PCOS.////////////////// Levels of antimüllerian hormone in serum during the normal menstrual cycle. Lambert-Messerlian G et al. (2015) To determine whether levels of antimüllerian hormone (AMH) in serum vary during the normal menstrual cycle, using the most recently developed immunoassay method. Prospective cohort study. Not applicable. Women with normal menstrual cycles and between the ages of 18 and 45 years were recruited (n = 45). Blood samples were collected on 5 days within each cycle: two in the follicular phase and three after confirmed ovulation. Exclusion criteria were anovulatory cycles, incomplete sample collection, insufficient blood volume, or non-Caucasian ethnicity. None. Serum samples were tested for levels of AMH using a new immunoassay method (Ansh Labs). The effects of body mass index (BMI) and smoking on serum AMH levels were considered. Serum AMH levels varied significantly during the menstrual cycle, with the highest levels in the follicular phase. When the analysis was stratified by age, AMH variation during the menstrual cycle was significant only for women older than 30 years. Serum AMH levels were not significantly altered by BMI or smoking. The new AMH immunoassay revealed a follicular phase rise in serum levels, particularly in women over the age of 30 years. This is consistent with other reports finding an interaction of menstrual cycle variation in AMH and chronological age. Nonetheless, the extent of variation is small, and sampling on any day of the menstrual cycle is expected to adequately reflect ovarian reserve. ClinicalTrials.gov (www.clinicaltrials.gov) registration no. NCT01337999.////////////////// Age-specific serum anti-mullerian hormone and follicle stimulating hormone concentrations in infertile Iranian women. Raeissi A et al. (2015) Anti-Müllerian hormone (AMH) is secreted by the granulosa cells of growing follicles during the primary to large antral follicle stages. Abnormal levels of AMH and follicle stimulating hormone (FSH) may indicate a woman's diminished ability or inability to conceive. Our aim is to investigate the changes in serum AMH and FSH concentrations at different age groups and its correlation with ovarian reserves in infertile women. This cross-sectional study analyzed serum AMH and FSH levels from 197 infertile women and 176 healthy controls, whose mean ages were 19-47 years. Sample collection was performed by random sampling and analyzed with SPSS version 16 software. There were significantly lower mean serum AMH levels among infertile women compared to the control group. The mean AMH serum levels from different ages of infertile and control group (fertile women) decreased with increasing age. However, this reduction was greater in the infertile group. The mean FSH serum levels of infertile women were significantly higher than the control group. Mean serum FSH levels consistently increased with increasing age in infertile women; however mean luteinizing hormone (LH) levels were not consistent. We have observed increased FSH levels and decreased AMH levels with increasing age in women from 19 to 47 years of age. Assessments of AMH and FSH levels in combination with female age can help in predicting ovarian reserve in infertile women.////////////////// DNA methylome and transcriptome sequencing in human ovarian granulosa cells links age-related changes in gene expression to gene body methylation and 3'-end GC density. Yu B et al. (2015) Diminished ovarian function occurs early and is a primary cause for age-related decline in female fertility; however, its underlying mechanism remains unclear. This study investigated the roles that genome and epigenome structure play in age-related changes in gene expression and ovarian function, using human ovarian granulosa cells as an experimental system. DNA methylomes were compared between two groups of women with distinct age-related differences in ovarian functions, using both Methylated DNA Capture followed by Next Generation Sequencing (MethylCap-seq) and Reduced Representation Bisulfite Sequencing (RRBS); their transcriptomes were investigated using mRNA-seq. Significant, non-random changes in transcriptome and DNA methylome features are observed in human ovarian granulosa cells as women age and their ovarian functions deteriorate. The strongest correlations between methylation and the age-related changes in gene expression are not confined to the promoter region; rather, high densities of hypomethylated CpG-rich regions spanning the gene body are preferentially associated with gene down-regulation. This association is further enhanced where CpG regions are localized near the 3'-end of the gene. Such features characterize several genes crucial in age-related decline in ovarian function, most notably the AMH (Anti-Müllerian Hormone) gene. The genome-wide correlation between the density of hypomethylated intragenic and 3'-end regions and gene expression suggests previously unexplored mechanisms linking epigenome structure to age-related physiology and pathology.////////////////// Biological variability in serum anti-M?an hormone throughout the menstrual cycle in ovulatory and sporadic anovulatory cycles in eumenorrheic women. Kissell KA 2014 et al. STUDY QUESTION Does serum anti-M?an hormone (AMH) vary significantly throughout both ovulatory and sporadic anovulatory menstrual cycles in healthy premenopausal women? SUMMARY ANSWER Serum AMH levels vary statistically significantly across the menstrual cycle in both ovulatory and sporadic anovulatory cycles of healthy eumenorrheic women. WHAT IS KNOWN ALREADY Studies to date evaluating serum AMH levels throughout the menstrual cycle have conflicting results regarding intra-woman cyclicity. No previous studies have evaluated an association between AMH and sporadic anovulation. STUDY DESIGN, SIZE, DURATION We conducted a prospective cohort study of 259 regularly menstruating women recruited between 2005 and 2007. PARTICIPANTS/MATERIALS, SETTING, METHODS Women aged 18-44 years were followed for one (n = 9) or two (n = 250) menstrual cycles. Anovulatory cycles were defined as any cycle with peak progesterone concentration =5 ng/ml and no serum LH peak on the mid or late luteal visits. Serum AMH was measured at up to eight-time points throughout each cycle. MAIN RESULTS AND THE ROLE OF CHANCE Geometric mean AMH levels were observed to vary across the menstrual cycle (P < 0.01) with the highest levels observed during the mid-follicular phase at 2.06 ng/ml, decreasing around the time of ovulation to 1.79 ng/ml and increasing thereafter to 1.93 (mid-follicular versus ovulation, P < 0.01; ovulation versus late luteal, P = 0.01; mid-follicular versus late luteal, P = 0.05). Patterns were similar across all age groups and during ovulatory and anovulatory cycles, with higher levels of AMH observed among women with one or more anovulatory cycles (P = 0.03). LIMITATIONS, REASONS FOR CAUTION Ovulatory status was not verified by direct visualization. AMH was analyzed using the original Generation II enzymatically amplified two-site immunoassay, which has been shown to be susceptible to assay interference. Thus, absolute levels should be interpreted with caution, however, patterns and associations remain consistent and any potential bias would be non-differential. WIDER IMPLICATIONS OF THE FINDINGS This study demonstrates a significant variation in serum AMH levels across the menstrual cycle regardless of ovulatory status. This variability, although statistically significant, is not large enough to warrant a change in current clinical practice to time AMH measurements to cycle day/phase. STUDY FUNDING/COMPETING INTERESTS This research was supported by the Intramural Research Program of the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, MD (Contracts # HHSN275200403394C, HHSN275201100002I Task 1 HHSN27500001). The authors have no conflicts of interest to declare. ///////////////////////// The significance of serum anti-M?an hormone (AMH) levels in patients over age 40 in first IVF treatment. Tokura Y et al. PURPOSE: Although studies of serum anti-M?an hormone (AMH) in predicting ovarian reserve are numerous, many studies utilized patients under age 40. However, the assessment of ovarian reserve is especially critical in older infertile women. This study evaluates the significance of AMH level in patients over age 40 at the time of their first in vitro fertilization (IVF) treatment. METHODS: Forty-nine women over age 40 were studied. Although serum samples were taken prior to their IVF treatments, the data of serum AMH of patients were not taken into consideration to determine the therapy strategy, including follicle induction in which clomiphene citrate and human menopausal gonadotropin were used. RESULT(S): Twelve out of 49 patients achieved a clinical pregnancy (24.4?%). There was a positive correlation between serum AMH levels and the number of oocytes retrieved (P?Bhide P et al. This single-centre retrospective observational study was performed at a university IVF centre. The aim was to examine the predictive power of AMH concentrations for clinical pregnancy rate (CPR) and establish a cut-off concentration of AMH below which no pregnancies were achieved. Data from 820 women with one treatment cycle each were analysed. There was a significant difference in CPR (24.4% and 40.0%; P<0.01) between the lowest and highest quartiles of AMH. This study failed to establish a cut-off concentration of AMH below which there were no clinical pregnancies as several pregnancies were achieved despite an AMH less than 1pmol/l. Log AMH showed a strong positive correlation with number of oocytes retrieved (r=0.522; P<0.001). Log AMH and overall CPR were weakly correlated (r=0.112, P<0.001), but this was not maintained when controlled for the number of oocytes. Age was a stronger independent predictor of CPR than AMH. In conclusion, although an excellent marker of ovarian response, AMH is only a weak predictor of clinical pregnancy. With AMH below the third percentile, CPR was 15%. However AMH is very useful for patient counselling and assessment when used in conjunction with age. Anti-M?an hormone (AMH) is a hormone produced by the ovary and reflects the egg reserve. The aim of this study was to examine the ability of AMH levels to predict pregnancy following IVF treatment and to establish a cut-off concentration of AMH below which no pregnancies were achieved. This retrospective study was carried out at a university IVF centre. It included 820 women who underwent IVF treatment in 1 year. The study showed that there was a strong positive correlation seen between AMH and the number of eggs obtained during IVF treatment. There was a significant difference in pregnancy rates (24.4% and 40%) between the lowest and highest quartiles of AMH. This was not maintained when controlled for the number of oocytes. This shows, that although an excellent marker of the number of eggs retrieved at IVF, AMH is only a weak predictor of clinical pregnancy. Age was a stronger independent predictor of pregnancy than AMH. With AMH below the third percentile, clinical pregnancy rates were still 15%. However, AMH is very useful for patient counselling and assessment when used in conjunction with age. We failed to establish a cut-off concentration of AMH below which there were no clinical pregnancies as several pregnancies were achieved despite having AMH concentrations less than 1pmol/l. Elimination Half-Life of Anti-Mullerian Hormone. Griesinger G et al. Context:Anti-Mullerian hormone (AMH) is a glycoprotein that is secreted by the granulosa cells in the human ovary. In the postpubertal female, circulating AMH reflects the number of follicles within the ovary. It is mandatory to know the serum elimination half-life (t(1/2)) of AMH to study in vivo short-term changes of the hormone.Objective:Our objective was to determine the kinetics of decay of AMH in the human female.Patients, Design, and Setting:Premenopausal women undergoing total abdominal hysterectomy plus bilateral salpingo-oophorectomy participated in this cohort study (n = 21) at an academic tertiary referral center.Interventions:Serum samples were obtained immediately before surgery and in 12-h intervals thereafter for 8 d.Main Outcome Measure:AMH elimination was calculated, applying a one-compartment model with first-order kinetics.Results:Mean preoperative AMH levels were 0.67 ng/ml (range, 0.1-1.78 ng/ml) and dropped to 0.08 ng/ml within 84 h after surgery. The AMH decay followed first-order kinetics. The mean terminal t(1/2) of AMH was calculated as 27.6 ? 0.8 h.Conclusion:AMH elimination reaches approximately 84% after 3 d, approximately 91% after 4 d, approximately 95% after 5 d, and can be considered complete after 8 d. Anti-Mullerian Hormone Predicts Menopause: A Long-Term Follow-Up Study in Normoovulatory Women. Broer SL et al. Context: It has been hypothesized that a fixed interval exists between age at natural sterility and age at menopause. Both events show considerable individual variability, with a range of 20 yr. Correct prediction of age at menopause could open avenues of individualized prevention of age-related infertility and other menopause-related conditions, like cardiovascular disease and breast carcinoma. Objective: The aim of this study was to explore the ability of ovarian reserve tests to predict age at menopause. Design and Setting: We conducted a long-term follow-up study at an academic hospital. Participants: A total of 257 normoovulatory women (age, 21-46 yr) were derived from three cohorts with highly comparable selection criteria. Interventions: Anti-M?an hormone (AMH), antral follicle count, and FSH were assessed at time 1 (T1). At time 2 (T2), approximately 11 yr later, cycle status (strictly regular, menopausal transition, or postmenopause) and age at menopause were inventoried. Main Outcome Measures: Accuracy of the ovarian reserve tests in predicting time to menopause was assessed by Cox regression, and a nomogram was constructed for the relationship between age-specific AMH concentrations at T1 and age at menopause. Results: A total of 48 (19%) women had reached postmenopause at T2. Age, AMH, and antral follicle count at T1 were significantly related with time to menopause (P < 0.001) and showed a good percentage of correct predictions (C-statistic, 0.87, 0.86, and 0.84, respectively). After adjusting for age, only AMH added to this prediction (C-statistic, 0.90). From the constructed nomogram, it appeared that the normal distribution of age at menopause will shift considerably, depending on the individual age-specific AMH level. Conclusions: AMH is highly predictive for timing of menopause. Using age and AMH, the age range in which menopause will subsequently occur can be individually calculated.Pregnancy and neonatal complications in women with polycystic ovary syndrome in relation to second-trimester anti-Müllerian hormone levels. Valdimarsdottir R et al. (2019) An association has been found between high anti-Müllerian hormone (AMH) levels during pregnancy and the development of polycystic ovary syndrome (PCOS)-like phenotypic traits in mouse offspring. The aim of this study was to determine whether AMH levels are associated with maternal testosterone levels, and whether high AMH concentration influences the risk of developing PCOS-related adverse pregnancy outcomes. Maternal serum AMH, testosterone and sex hormone binding globulin levels were measured in blood samples taken in early second-trimester pregnancies from women with PCOS (n = 159) and healthy controls matched for body mass index (n = 320). Possible associations with preeclampsia, gestational hypertension, gestational diabetes, preterm birth and birthweight was explored by logistic and linear regression models. Women with PCOS had higher AMH, higher total testosterone levels and higher free androgen index than controls (P < 0.001 for all three parameters). Among women with PCOS, high testosterone levels (B = 2.7; β = 0.26; P = 0.001) and low first trimester body mass index (B = -0.5; β = -0.17; P = 0.043) remained independently associated with AMH. High AMH levels were associated with decreased risk of gestational hypertension (adjusted OR 0.55; 95% CI 0.34 to 0.87), but no association was found with other adverse pregnancy outcomes or birthweight. Women with PCOS had higher AMH levels during pregnancy compared with controls, but high AMH was not associated with increased risk of adverse pregnancy outcomes or birthweight.////////////////// Circulating antimüllerian hormone and steroid hormone levels remain high in pregnant women with polycystic ovary syndrome at term. Piltonen TT et al. (2019) To investigate plasma antimüllerian hormone (AMH) concentration and its relation to steroid hormone levels in pregnant women with polycystic ovary syndrome (PCOS) and controls at term. Case-control study. University-affiliated hospital. A total of 74 pregnant women at term: 25 women with PCOS (aged 31.6 ± 3.9 years mean ± standard deviation], body mass index 24.0 ± 3.9 kg/m2, mean gestational length 279 ± 9 days) and 49 controls (aged 31.7 ± 3.3 years, body mass index 24.0 ± 3.3 kg/m2, mean gestational length 281 ± 9 days). None. Plasma AMH and steroid hormone levels. Antimüllerian hormone, T, and androstenedione levels were higher in women with PCOS at term compared with controls, whereas estrogen and P levels were similar. The differences were pronounced in women carrying a female fetus. Testosterone and AMH levels correlated positively in both groups, but E2 levels only in women with PCOS. Pregnant women with PCOS present with elevated AMH and androgen levels even at term, suggesting a hormonal imbalance during PCOS pregnancy. Differences were detected especially in pregnancies with a female fetus, raising the question of whether female pregnancies are more susceptible to AMH and steroid hormone actions.////////////////// Elevated prenatal anti-Müllerian hormone reprograms the fetus and induces polycystic ovary syndrome in adulthood. [Tata B et al. (2018) Polycystic ovary syndrome (PCOS) is the main cause of female infertility worldwide and corresponds with a high degree of comorbidities and economic burden. How PCOS is passed on from one generation to the next is not clear, but it may be a developmental condition. Most women with PCOS exhibit higher levels of circulating luteinizing hormone, suggestive of heightened gonadotropin-releasing hormone (GnRH) release, and anti-Müllerian hormone (AMH) as compared to healthy women. Excess AMH in utero may affect the development of the female fetus. However, as AMH levels drop during pregnancy in women with normal fertility, it was unclear whether their levels were also elevated in pregnant women with PCOS. Here we measured AMH in a cohort of pregnant women with PCOS and control pregnant women and found that AMH is significantly more elevated in the former group versus the latter. To determine whether the elevation of AMH during pregnancy in women with PCOS is a bystander effect or a driver of the condition in the offspring, we modeled our clinical findings by treating pregnant mice with AMH and followed the neuroendocrine phenotype of their female progeny postnatally. This treatment resulted in maternal neuroendocrine-driven testosterone excess and diminished placental metabolism of testosterone to estradiol, resulting in a masculinization of the exposed female fetus and a PCOS-like reproductive and neuroendocrine phenotype in adulthood. We found that the affected females had persistently hyperactivated GnRH neurons and that GnRH antagonist treatment in the adult female offspring restored their neuroendocrine phenotype to a normal state. These findings highlight a critical role for excess prenatal AMH exposure and subsequent aberrant GnRH receptor signaling in the neuroendocrine dysfunctions of PCOS, while offering a new potential therapeutic avenue to treat the condition during adulthood.////////////////// Increased antimüllerian hormone levels and other reproductive endocrine changes in adult male relatives of women with polycystic ovary syndrome. Torchen LC et al. (2016) To investigate for differences in reproductive hormone levels in male relatives of women with polycystic ovary syndrome (PCOS). Cross-sectional study. Academic medical center. Sixty-three fathers and 66 brothers of women with PCOS as well as two groups of control men of comparable age to fathers (older control, n = 30) and brothers (younger control, n = 58). A single early morning fasting blood sample was obtained for the measurement of reproductive hormone levels. Testosterone, LH, FSH, antimüllerian hormone (AMH), inhibin B, E2, and estrone (E1) levels were measured. The AMH, LH, and FSH levels were significantly increased in male relatives compared with their respective control groups. The levels of E2, E1, T, and inhibin B did not differ between the groups. The AMH, LH, and FSH levels were increased in adult male relatives of women with PCOS, suggesting that they may have altered testicular function and changes in neuroendocrine regulation of gonadotropin secretion. These changes may reflect effects of PCOS susceptibility genes such as the recently mapped chromosome 11p14.1 locus in the region of the FSH B polypeptide gene.////////////////// Anti-Müllerian hormone in children: a ten-year prospective longitudinal study (EarlyBird 39). Jeffery A et al. (2015) Anti-Müllerian hormone (AMH) is produced by Sertoli cells of the testes and granulosa cells of the ovary. There are limited prospective longitudinal data assessing AMH concentrations throughout childhood in both sexes. This study aimed to examine AMH throughout childhood with particular reference to the relationship of AMH to pubertal development in both sexes. This is a prospective longitudinal non-intervention cohort study with annual sampling for participants aged 5-14 years. Community cohort study. A total of 307 healthy children (170 boys) recruited at 5 years from randomly selected schools in Plymouth, UK, participated in this study. Data sets are complete in 76% of the children at 14 years of age. Annual measures of serum AMH, follicle stimulating hormone (FSH) and luteinising hormone (LH), Tanner stage (TS). Boys: AMH was stable from 5 to 7 years, increased slightly from 8 to 10 years, then declined at TS2. This decline was preceded by rising FSH and the appearance of LH. AMH correlated inversely with gonadotrophic hormones during puberty. Girls: AMH increased slightly between 6 and 10 years, peaking during the final prepubertal year before returning to near baseline levels at TS3. Inverse correlations between AMH and FSH were apparent during the prepubertal years. Our longitudinal data clarified the development of individual AMH levels over a 10-year period. We described modest late prepubertal peaks in both boys and girls, and confirmed the pubertal decline in boys. The inverse association of AMH with gonadotrophins in young females supports its role as a marker of ovarian function, while the precise role for AMH in relation to testicular function in young males remains unclear.////////////////// Lack of serum anti-mullerian hormone responses after recombinant human chorionic gonadotropin stimulation in women with polycystic ovary syndrome. Cook-Andersen H et al. (2015) Polycystic ovary syndrome (PCOS) is an anovulatory disorder characterized by excess androgen production and increased LH secretion. Serum anti-Mullerian hormone (AMH) is also elevated in this disorder. Women with PCOS exhibit a positive correlation between AMH and LH levels and recent in vitro data demonstrate that LH can directly stimulate AMH production by granulosa cells from women with PCOS. The objective of the study was to directly test whether LH increases AMH production in women with PCOS in vivo by assessing responses after recombinant human chorionic gonadotropin (r-hCG) stimulation. This was a prospective study. The study was conducted at a research center at an academic medical center. Women with PCOS (n = 28) and normal controls (n = 29) participated in the study. Blood samples were obtained before and 24 hours after iv administration of 25 μg r-hCG. Basal and stimulated serum AMH, androstenedione, T, and 17-hydroxyprogesterone levels were measured. Baseline AMH levels in women with PCOS were greater than in normal controls and correlated with levels of LH as well as androstenedione, T, and 17-hydroxyprogesterone. A rise of serum AMH levels was not observed after r-hCG administration in women with PCOS or normal ovaries. These findings are in contrast to in vitro evidence demonstrating that AMH secretion by granulosa cells of PCOS women in response to LH stimulation and suggest AMH regulation in vivo is complex and that the elevated serum AMH in women with PCOS is not a direct effect of the excess LH production characteristic of PCOS.////////////////// The role of anti-m?an hormone (AMH) in assessing ovarian reserve. Tran ND 2011 et al. CONTEXT Anti-m?an hormone (AMH) has been suggested as a marker for the quantity of oocytes remaining within the ovaries (ovarian reserve). It was shown to correlate with antral follicle counts (AFC), outcomes from ovarian stimulation, and onset of menopause. Thus, AMH was previously considered to be the ideal marker of ovarian reserve because it is exclusively produced by granulosa cells and is the only marker that was thought to be stable throughout the menstrual cycle. However, recent studies demonstrate fluctuations in AMH levels during the menstrual cycles, questioning the utility of AMH as a marker of oocyte quantity. OBJECTIVE We report the case of a 32-yr-old Gravida 0 woman with idiopathic hypogonadotropic hypogonadism who presented for fertility treatment with unstable AMH levels. PATIENT AND METHODS The patient's initial FSH and LH levels were both below 1.0 mIU/ml. Estradiol was 28 pg/ml. Her initial AMH and AFC were 0.20 ng/ml and 0, respectively. She underwent three cycles of fertility treatment. RESULTS During the 16-wk course of treatment with human menopausal gonadotropins, normal follicular development was observed. Both AMH and AFC gradually increased during treatment and peaked at 1.26 ng/ml and 6, respectively. On the third cycle of treatment, she successfully conceived. CONCLUSION In the case of idiopathic hypogonadotropic hypogonadism, AMH concentration increases because human menopausal gonadotropin stimulates the growth of FSH-dependent follicles. Thus, AMH has limitations because it only reflects the growing follicular pool that is responsive to gonadotropins. Therefore, AMH may not be solely reflective of the underlying primordial pool. ///////////////////////// Can anti-mullerian hormone predict the diagnosis of polycystic ovary syndrome? A systematic review and meta-analysis of extracted data. Iliodromiti S 2013 et al. Context: Existing biochemical tests for polycystic ovary syndrome (PCOS) have poor sensitivity and specificity. Many women with PCOS have high anti-M?an hormone (AMH) concentrations; thus, this may be a useful addition to the diagnostic criteria. Objective: A systematic literature review was performed to assess the true accuracy of AMH in the prediction of PCOS and to determine the optimal diagnostic threshold. Data Sources: Published and gray literature were searched for all years until January 2013. Study Selection: Observational studies defining PCOS according to the Rotterdam criteria and assessing the value of AMH in diagnosing PCOS were selected. Ten studies of the initial 314 hits reporting AMH values in the diagnosis of PCOS were included in the meta-analysis and the construction of the summary receiver-operating characteristic curve. Four studies that plotted individual AMH serum levels of women with PCOS and controls on graphs were selected for individual data extraction. Data Extraction: Two researchers independently assessed the abstracts resulted from the initial search against the inclusion criteria, graded the papers for selection and verification biases, and selected the papers that assessed the value of AMH in diagnosing PCOS. Data were extracted from 4 studies with the plotted individual data on graphs with the help of computer software. Data Synthesis: The meta-analysis of the extracted data demonstrated the specificity and sensitivity in diagnosing PCOS in the symptomatic women of 79.4% and 82.8%, respectively, for a cutoff value of AMH of 4.7 ng/mL. The area under the curve was 0.87 (95% confidence interval 0.83-0.92), identical with the area under the curve of 0.87 for the summary receiver-operating characteristic curve involving 10 separate studies. Conclusions: AMH may be a useful initial diagnostic test for PCOS subject to validation in prospective population cohorts. ///////////////////////// Serum anti-Mullerian hormone levels remain high until late reproductive age and decrease during metformin therapy in women with polycystic ovary syndrome Piltonen T, et al . BACKGROUND: Anti-Mullerian hormone (AMH) is secreted by granulosa cells of ovarian early developing follicles and its serum levels have been shown to correlate with small antral follicle number. Since the pronounced androgen secretion from follicles/stroma in women with polycystic ovary syndrome (PCOS) remains until late reproductive age, and since AMH reflects the number of antral follicles, it was of interest to study the possible age-related relationship between AMH, androgens and follicle number in women with PCOS and in control women. Moreover, the possible effect of metformin on serum AMH levels and the relationship to follicle count and volume were studied. METHODS: Forty-four healthy women (aged 21-44 years) and 65 women with previously diagnosed PCOS (aged 16-44 years) participated in the study. Serum basal AMH levels were correlated with those of serum androstenedione, testosterone, estradiol (E2), LH, FSH and inhibin B, and with follicle number. The effect of metformin on serum AMH concentrations, follicle number and ovarian volume was studied in 26 women (aged 20-41 years) with PCOS after 6 months of treatment. RESULTS: Serum AMH levels were 2- to 3- fold higher in PCOS women than in healthy women. In control women, serum AMH levels correlated positively with those of serum androstenedione (r=0.564, P<0.001) and testosterone (r=0.328, P=0.036) and negatively with serum FSH concentrations (r=-0.374, P=0.012) and age (r=-0.691, P<0.001). In women with PCOS, serum AMH levels correlated positively with those of androstenedione (r=0.311, P=0.011) and testosterone (r=0.310, P=0.011) and with follicle count (r=0.352, P=0.012), and negatively with age (r=-0.300, P=0.014). Serum AMH levels, the number of antral follicles and ovarian volume decreased significantly during metfromin treatment. CONCLUSIONS: Serum AMH levels decreased with age both in healthy women and in women with PCOS, although they were always 2- to 3-fold higher and remained elevated until 40 years of age in PCOS subjects. Thus, since serum AMH levels correlate well with antral follicle count and serum androgen levels, the measurement of AMH could be used as a tool to assess ovarian ageing, to diagnose polycystic ovaries/PCOS and to evaluate treatment efficacy.
Cellular localization Secreted
Comment PMID:34000288////Clin Biochem . 2021 May 14;S0009-9120(21)00148-X. doi: 10.1016/j.clinbiochem.2021.05.007. Online ahead of print. Association of ovarian response with picoAMH in women undergoing controlled ovarian hyperstimulation family123////// //////////////////
Ovarian function Follicle endowment, Follicle development, Initiation of primordial follicle growth, Primary follicle growth, Preantral follicle growth, Antral follicle growth, Follicle atresia, Steroid metabolism, Germ cell development
Comment Control of primordial follicle recruitment by anti-Müllerian hormone in the mouse ovary. Durlinger AL et al. (1999) The dimeric glycoprotein anti-Müllerian hormone (AMH) is a member of the transforming growth factor-beta superfamily of growth and differentiation factors. During male fetal sex differentiation, AMH is produced by Sertoli cells and induces degeneration of the Müllerian ducts, which form the anlagen of part of the internal female genital system. In females, AMH is produced by the ovary, but only postnatally. The function of AMH in the ovary is, however, still unknown. Female AMH null mice were reported to be fertile, with normal litter size, but this does not exclude a more subtle function for ovarian AMH. To investigate the function of AMH in the ovary, the complete follicle population was determined in AMH null mice, in mice heterozygous for the AMH null mutation, and in wild-type mice of different ages: 25 days, 4 months, and 13 months. In the present study we found that ovaries of 25-day- and 4-month-old AMH null females, compared to those of wild-type females, contain more preantral and small antral follicles. In addition, in 4- and 13-month-old AMH null females, smaller numbers of primordial follicles were found. Actually, in 13-month-old AMH null females, almost no primordial follicles could be detected, coinciding with a reduced number of preantral and small antral follicles in these females. In almost all females heterozygous for the AMH null mutation the number of follicles fell in between the numbers found in wild-type and AMH null females. In 4-month-old AMH null females serum inhibin levels were higher and FSH levels were lower compared to those in wild-type females. In contrast, inhibin levels were lower in 13-month-old AMH null females, and FSH levels were unchanged compared to those in wild-type females. Furthermore, the weight of the ovaries was twice as high in the 4-month-old AMH null females as in age-matched wild-type females. We conclude that AMH plays an important role in primordial follicle recruitment, such that more primordial follicles are recruited in AMH null mice than in wild-type mice; the mice heterozygous for the AMH null mutation take an in-between position. Consequently, the ovaries of AMH null females and those of females heterozygous for the AMH null mutation will show a relatively early depletion of their stock of primordial follicles. The female AMH null mouse may thus provide a useful model to study regulation of primordial follicle recruitment and the relation between follicular dynamics and ovarian aging.//////////////////Stage-specific modulation of antimüllerian hormone promotes primate follicular development and oocyte maturation in the matrix-free three-dimensional culture. Xu J et al. (2018) To study whether follicular growth and oocyte maturation can be improved by antimüllerian hormone (AMH) modulation at specific stages of follicular development. Primary and secondary follicles were cultured in a matrix-free system and were assigned to the control group and the group with AMH supplementation during the preantral stage and neutralizing AMH antibody addition during the antral stage. National primate research center. Adult, female rhesus macaques (Macaca mulatta). None. Follicle survival, growth, steroid and paracrine factor production, and oocyte competence were evaluated. Follicles were assessed for expression of genes that are critical for gonadotropin signaling, cumulus cell glycolysis, and oocyte quality. Primary follicles formed "organoids" and developed to the antral stage in group culture. AMH exposure during the preantral stage increased organoid diameters. Oocytes from the AMH-treated organoids had greater diameters and matured to the metaphase II (MII) stage. Secondary follicles developed to the antral stage during individual culture. The AMH exposure during the preantral stage and AMH antibody treatment during the antral stage increased follicle diameters, vascular endothelial growth factor and follistatin production, differentiation factor 9 expression, and oocyte diameters. The MII oocytes from the AMH-modulated group developed to the morula stage after IVF, with one to the blastocyst stage. AMH supplementation at the preantral stage and depletion at the antral stage enhanced primate follicular development and oocyte competence in vitro. The improved embryonic development supports in vitro follicle maturation as a potential approach for fertility preservation.////////////////// Anti-Müllerian hormone inhibits activation and growth of bovine ovarian follicles in vitro and is localized to growing follicles. Yang MY et al. (2017) Does anti-Müllerian hormone (AMH) inhibit activation (initiation of growth) of primordial follicles and attenuate the growth of primary follicles in cattle, an excellent animal model for human ovarian follicular development? AMH inhibited activation of bovine primordial follicles and attenuated the growth of activated follicles in vitro. In mice null mutant for AMH, the pool of primordial follicles is depleted prematurely and AMH inhibits follicle activation in vitro. Results of studies with human ovarian tissue in vitro were inconsistent. Our previous work provided indirect evidence that AMH inhibits follicle activation in bovine ovaries. Pieces of fetal bovine ovarian cortex (2 pieces/culture well), obtained during mid or late pregnancy, were cultured in control medium or with graded doses of AMH for 2, 10 or 12 days. Effects of treatment on follicle activation and growth were determined by histological morphometry; follicles in every 20th histological section were staged (primordial or primary), counted, and measured. In addition, AMH was immunolocalized in bovine ovaries obtained at various times during pregnancy (n = 20 ovaries). Bovine fetal ovaries at mid or late gestation were obtained at a commercial abattoir. Pieces of ovarian cortex were cultured without or with AMH and fixed for histological morphometry on Day 0 and at the end of culture. Treatments were applied to duplicate cultures from each of two or three fetuses. In 12-day cultures, addition of AMH was delayed until the third day. Histological analysis provided information about the types, numbers and sizes of follicles in cortical pieces before and after treatments. Ovaries obtained during the second and third trimesters were assessed for the presence of AMH by immunohistochemistry. AMH (100-500 ng/ml) inhibited follicle activation in response to an activator (insulin) in ovarian cortical pieces from fetal ovaries in late gestation. Dose-dependent inhibitory effects on the diameters of primary follicles and their oocytes were also observed. These results were obtained only when AMH was added to cultures in advance of insulin (presumably because it penetrates tissue more slowly than insulin). Results of experiments with cortical pieces from fetal ovaries at mid-gestation, when follicles are forming, showed that AMH did not inhibit the formation of follicles. Immunohistochemical localization of AMH showed that it is not present in fetal ovaries until the third trimester, when it was localized to the granulosa cells of secondary and small antral follicles. The experiments were performed with fetal ovaries because follicles form and follicle activation begins during fetal life in cattle (as it does in humans), so fetal ovarian cortex of later gestation provides tissue rich in primordial follicles. We assume, but have no experimental evidence, that our findings also apply to post-natal ovaries. Although circulating AMH is used as an indication of the follicular reserve in women, little is known about AMH in human ovaries. Cattle are an excellent non-primate model for human ovarian follicular development and, hence, the findings suggest similar roles for AMH in human follicular development. Not applicable. This research was supported by National Research Initiative Competitive Grants no. 00-35203-9151, 2003-35203-13532, and 2008-35203-05989) from the U.S. Dept. of Agriculture's National Institute of Food and Agriculture to JEF and by an NIH National Research Service Award (F32 HD08264) to RAC. There are no conflicts of interest or competing interests.////////////////// Mullerian-inhibiting substance/anti-Mullerian hormone as a predictor of preterm birth in polycystic ovary syndrome. Hsu JY et al. (2018) There is increasing evidence for MIS/AMH physiologic activity in the human uterus, so it is relevant to study how MIS/AMH levels impact pregnancy. To investigate the association of MIS/AMH levels with risk of adverse obstetric outcomes. Retrospective cohort study. Academic fertility center. Women who became pregnant through in vitro fertilization (IVF) between January 2012-October 2016. Exclusion criteria were: oocyte donation, gestational carrier, multiple gestation, miscarriage before 20 weeks, medically indicated preterm deliveries. None. There were two primary outcomes, (1) preterm birth and (2) Cesarean delivery for arrest of labor. Because MIS/AMH level is highly skewed by certain infertility diagnoses, the preterm birth analysis was stratified by polycystic ovary syndrome (PCOS) diagnosis, and the Cesarean delivery for arrest of labor analysis was stratified by diminished ovarian reserve (DOR) diagnosis. Chi-squared, Mann-Whitney, and t-test were used as appropriate. A p-value of < 0.05 was considered statistically significant.
. Among women with PCOS, those who delivered prematurely had substantially higher MIS/AMH levels (18 vs. 6.4 ng/mL, p = 0.003) than those who delivered at term. At the highest MIS/AMH values, preterm deliveries predominated; above the 90th percentile in PCOS women, all deliveries were premature. No effect of MIS/AMH level was observed in women without PCOS. We found no association between MIS/AMH values and Cesarean delivery for labor arrest. In women with PCOS, substantially elevated MIS/AMH levels are significantly associated with preterm birth, suggesting closer follow-up and further studies to elucidate the underlying mechanisms.
.////////////////// Intra-cellular mechanism of Anti-Müllerian hormone (AMH) in regulation of follicular development. Hayes E et al. (2016) Anti-Müllerian hormone (AMH) is a member of the transforming growth factor-β superfamily and plays a crucial role in testicular and ovarian functions. In clinical practice, AMH is used as a diagnostic and/or prognostic marker in women in association with ovulation induction and in various pathophysiological conditions. Despite widespread clinical use of AMH, our mechanistic understanding of AMH actions in regulating follicular development is limited. Using a mouse model, we in this study report that in vivo AMH treatment while stalls follicular development and inhibits ovulation, also prevents follicular atresia. We further show that these AMH actions are mediated through induction of two miRNAs, miR-181a and miR-181b, which regulate various aspects of FSH signaling and follicular growth, ultimately affecting downstream gene expression and folliculogenesis. We also report that in this mouse model AMH pre-treatment prior to superovulation improves oocyte yield. These studies, therefore, offer new mechanistic insight into AMH actions in folliculogenesis and point toward potential utilization of AMH as a therapeutic agent.////////////////// Anti-Müllerian hormone promotes pre-antral follicle growth, but inhibits antral follicle maturation and dominant follicle selection in primates. Xu J et al. (2016) What are the direct effects and physiological role of anti-Müllerian hormone (AMH) during primate follicular development and function at specific stages of folliculogenesis? AMH actions in the primate ovary may be stage-dependent, directly promoting pre-antral follicle growth while inhibiting antral follicle maturation and dominant follicle selection. AMH is expressed in the adult ovary, particularly in developing follicles. Studies in mice suggest that AMH suppresses pre-antral follicle growth in vitro, and inhibits primordial follicle recruitment and FSH-stimulated antral follicle steroidogenesis. For in vitro study, secondary follicles were isolated from ovaries of 12 rhesus macaques and cultured for 5 weeks. For in vivo study, intraovarian infusion was conducted on five monkeys for the entire follicular phase during two spontaneous menstrual cycles. For in vitro study, individual follicles were cultured in a 5% O2 environment, in alpha minimum essential medium supplemented with recombinant human FSH. Follicles were randomly assigned to treatments of recombinant human AMH protein or neutralizing anti-human AMH antibody (AMH-Ab). Follicle survival, growth, steroid production, steroidogenic enzyme expression, and oocyte maturation were assessed. For in vivo study, ovaries were infused with control vehicle or AMH-Ab during the follicular phase of the menstrual cycle. Cycle length, serum steroid levels, and antral follicle growth were evaluated. AMH exposure during culture weeks 0-3 (pre-antral stage) promoted, while AMH-Ab delayed, antrum formation of growing follicles compared with controls. AMH treatment during culture weeks 3-5 (antral stage) decreased (P < 0.05) estradiol (E2) production, as well as the mRNA expression of cytochrome P450 family 19 subfamily A polypeptide 1, by antral follicles relative to controls, whereas AMH-Ab increased (P < 0.05) follicular mRNA levels of the enzyme. Intraovarian infusion of AMH-Ab during the follicular phase of the menstrual cycle increased (P < 0.05) the average levels of serum E2 compared with those of the control cycles. Three of the five AMH-Ab-treated ovaries displayed multiple (n = 2-9) medium-to-large (2-8 mm) antral follicles at the mid-cycle E2 peak, whereas only one large (4-7 mm) antral follicle was observed in all monkeys during their control cycles. The average levels of serum progesterone were higher (P < 0.05) during the luteal phase of cycles following the AMH-Ab infusion relative to the vehicle infusion. The in vitro study of AMH actions on cultured individual macaque follicles was limited to the interval from the secondary to small antral stage. A sequential study design was used for in vivo experiments, which may limit the power of the study. The current study provides novel information on direct actions and role of AMH during primate follicular development, and selection of a dominant follicle by the late follicular phase of the menstrual cycle. We hypothesize that AMH acts positively on follicular growth during the pre-antral stage in primates, but negatively impacts antral follicle maturation, which is different from what is reported in the mouse model. NIH NICHD R01HD082208, NIH ORWH/NICHD K12HD043488 (BIRCWH), NIH OD P51OD011092 (ONPRC), Collins Medical Trust. There are no conflicts of interest. Not applicable.////////////////// Immunolocalization of the Anti-Müllerian Hormone (AMH) in Caprine Follicles and the Effects of AMH on In Vitro Culture of Caprine Pre-antral Follicles Enclosed in Ovarian Tissue. Rocha R et al. (2016) The aims of this study were to evaluate the localization, by immunohistochemistry, of the anti-Müllerian hormone (AMH) in goat ovaries and to investigate its effects on the in vitro survival and development of caprine pre-antral follicles enclosed in fragments of ovarian tissue. Pre-antral follicles were cultured in vitro for 1 or 7 days in α-MEM(+) in the absence or presence of kit ligand (KL; 50 ng/ml, positive control) or AMH (50 or 150 ng/ml). The results showed that AMH was localized in oocytes and granulosa cells from the primordial follicle to antral follicle stages. Addition of AMH maintained the percentage of developing follicles, similar to that in the uncultured control; however, the percentage of developing follicles was significantly lower than that in the cultured control and KL. Nonetheless, addition of AMH to the culture medium did not affect survival rates and follicular growth. In conclusion, this study demonstrated that the expression of AMH varies according to the compartment and stage of follicular development. Furthermore, AMH inhibits the activation of caprine primordial follicles.////////////////// The anti-Müllerian hormone (AMH) acts as a gatekeeper of ovarian steroidogenesis inhibiting the granulosa cell response to both FSH and LH. Sacchi S et al. (2015) Anti Müllerian Hormone (AMH) has a negative and inhibitory role in many functions of human granulosa-lutein cells (hGCs) including notoriously the reduction of the aromatase CYP19A1 expression induced by follicle-stimulating hormone (FSH). No data have been provided on the possible role of AMH in modulating the response to luteinizing hormone (LH) (alone or combined with FSH) as well as its effect on other enzymes involved in steroidogenesis including aromatase P450scc. The aim of this study was to investigate the role of AMH as regulator of the basal and stimulated steroids production by hGCs. Primary culture of hGCs were incubated with hormones AMH, LH, and FSH, alone or in combination. The CYP19A1 and P450scc messenger RNA (mRNA) expression, normalized by housekeeping ribosomal protein S7 (RpS7) gene, was evaluated by reverse transcriptase quantitative PCR (RT-qPCR). Each reaction was repeated in triplicate. Negative controls using corresponding amount of vehicle control for each hormone treatment were performed. AMH did not modulate the basal mRNA expression of both aromatase genes at any of the concentrations tested. Meanwhile, the strong mRNA induction of CYP19A1 and P450scc generated by a 24-h gonadotropin treatment (alone and combined) was suppressed by 20 ng/ml AMH added to culture medium. These findings contribute in clarifying the relationship between hormones regulating the early phase of steroidogenesis confirming that AMH is playing a suppressive role on CYP19A1 expression stimulated by gonadotropin in hGCs. Furthermore, a similar inhibitory effect for AMH was observed on P450scc gene expression when activated by gonadotropin treatment.////////////////// Is AMH a regulator of follicular atresia? Seifer DB 2014 et al. We discuss the hypothesis that AMH is an intraovarian regulator that inhibits follicular atresia within the human ovary. Several indirect lines of evidence derived from clinical and basic science studies in a variety of different patient populations and model systems collectively support this hypothesis. Evidence presented herein include 1) timing of onset of menopause in women with polycystic ovary syndrome, 2) site of cellular origin and timing of AMH production, 3) AMH's influence on other critical growth factors and enzymes involved in folliculogenesis, and 4) AMH's inhibition of granulosa apoptosis. If this hypothesis is true, it may provide insight for treatment strategies for prevention and treatment of premature ovarian insufficiency, slowing natural ovarian aging, and/or delaying eventual ovarian failure. Such findings may lead to the development of 1) AMH agonists for retarding the onset of menopause and/or as a chemoprotectant prior to cancer therapy and 2) AMH antagonists for the treatment of PCOS. /////////////////////////Follicular fluid anti-M?an hormone: a predictive marker of fertilization capacity of MII oocytes. Trami?ak Milakovic T 2014 et al. PURPOSE The present study aimed to correlate anti-M?an hormone (AMH) levels in follicular fluid (FF) with oocyte maturity stages, morphological quality of metaphase II (MII) oocyte and fertilization capacity of MII oocytes. METHODS A total of 92 infertile women undergoing controlled ovarian stimulation and intracytoplasmic sperm injection were analyzed. Patients were divided into two groups according to age: <35?years (n?=?43) and =35?years (n?=?49). An FF sample was obtained from a single dominant follicle in each patient for a total of 92 follicular fluid samples analyzed. AMH levels in serum and follicular fluid were measured by enzyme-linked immunosorbent assay. Mature MII oocytes, zygotes, and embryos were assessed for morphological quality. RESULTS Serum AMH levels were significantly higher in patients aged <35?years. No correlation was observed between FF AMH level and oocyte maturation stages or morphological quality of MII oocyte. Significantly lower FF AMH levels were observed in fertilized MII oocytes than in non-fertilized MII oocytes in patients aged <35?years (2.56???2.0?ng/ml vs. 4.81???4.14?ng/ml; p?=?0.032). CONCLUSIONS The present study revealed no correlation between FF AMH and oocyte maturity stage or morphological quality of MII oocyte. However, FF AMH might be a predictive marker for fertilization capacity of MII oocytes. ///////////////////////// Anti-M?an hormone inhibits initiation of growth of human primordial ovarian follicles in vitro. Carlsson IB 2006 et al. BACKGROUND Anti-M?an hormone (AMH) inhibits the initiation of the development and early growth of mouse ovarian follicles. Furthermore, the ovarian follicle pool diminishes prematurely in AMH-knockout mice. In this study, we examined whether AMH plays a similar role in humans, controlling ovarian follicle growth. METHODS Human ovarian cortical tissue biopsy specimens were cut into small pieces and cultured for 7 days in medium containing rat recombinant AMH at 0, 10, 30 or 100 ng/ml. The developmental stages and viability of the follicles were evaluated from histological sections. RESULTS Similar to previous studies, significant initiation of follicle growth was observed in almost all culture media, as demonstrated by a significantly smaller proportion of primordial follicles (14-26%) compared with non-cultured control tissue (56%). The exception was tissue in medium supplemented with AMH at 100 ng/ml. Here, the proportion of primordial follicles was not significantly different from that in non-cultured tissue; furthermore, it was significantly greater than that in vehicle control cultures and cultures containing AMH at 10 ng/ml, indicating the inhibition of growth initiation. Viability was unaffected by the presence of AMH when compared with tissues in control media. CONCLUSIONS Recombinant AMH at a concentration of 100 ng/ml has an inhibitory effect on early human ovarian follicular development in vitro, suppressing the initiation of primordial follicle growth. ///////////////////////// Effect of anti-mullerian hormone in culture medium on quality of mouse oocytes matured in vitro. Zhang Y 2014 et al. Anti-mullerian hormone (AMH) is thought to reflect the growth of follicles and the ovarian function. However, the role of AMH in culture medium during in vitro maturation (IVM) on oocyte quality and subsequent development potential is unclear. The objective of this study is to investigate the effect of recombinant human AMH (rh-AMH) supplemented into IVM medium on oocyte quality. Cumulus-oocyte complexes (COCs) were obtained from ICR mice and cultured in vitro with the different concentrations (0-1,000 ng/ml) of rh-AMH. Following 16-18 h of culture, quantitative PCR and ELISA were performed to analyze GDF9 and BMP15 mRNA expression and protein production from the oocytes. Subsequently, in vitro fertilization (IVF) and early embryonic development were employed to further evaluate the quality of in vitro matured oocytes. The results showed that AMH was only expressed in cumulus cells but not in the oocytes. However, AMH most specific receptor, AMHR-II, was expressed in both oocytes and cumulus cells. The levels of GDF9 and BMP15 expression and blastocyst formation rate were significantly increased (p<0.05) when the IVM medium was supplemented with 100 ng/ml of rh-AMH. With AdH1-SiRNA/AMH for knocking down of AMH expression during IVM significantly reduced (p<0.05) the levels of GDF9 and BMP15 expression and blastocysts formation rate. These results suggest that AHM improves oocytes quality by up-regulating GDF9 and BMP15 expressions during IVM. ///////////////////////// Anti-M?an Hormone Recruits BMPR-IA in Immature Granulosa Cells. S?s L 2013 et al. Anti-M?an hormone (AMH) is a member of the TGF-?superfamily secreted by the gonads of both sexes. This hormone is primarily known for its role in the regression of the M?an ducts in male fetuses. In females, AMH is expressed in granulosa cells of developing follicles. Like other members of the TGF-?superfamily, AMH transduces its signal through two transmembrane serine/threonine kinase receptors including a well characterized type II receptor, AMHR-II. The complete signalling pathway of AMH involving Smads proteins and the type I receptor is well known in the M?an duct and in Sertoli and Leydig cells but not in granulosa cells. In addition, few AMH target genes have been identified in these cells. Finally, while several co-receptors have been reported for members of the TGF-?superfamily, none have been described for AMH. Here, we have shown that none of the Bone Morphogenetic Proteins (BMPs) co-receptors, Repulsive guidance molecules (RGMs), were essential for AMH signalling. We also demonstrated that the main Smad proteins used by AMH in granulosa cells were Smad 1 and Smad 5. Like for the other AMH target cells, the most important type I receptor for AMH in these cells was BMPR-IA. Finally, we have identified a new AMH target gene, Id3, which could be involved in the effects of AMH on the differentiation of granulosa cells and its other target cells. ///////////////////////// Long-term treatment with dehydroepiandrosterone may lead to follicular atresia through interaction with anti-Mullerian hormone. Ikeda K 2014 et al. BACKGROUND Hyperandrogenism is the primary manifestation of polycystic ovary syndrome (PCOS), which appears to be caused by excess exposure to androgen. As such, androgenized animal models have been developed and investigated to study the etiology of PCOS. Anti-Mullerian hormone (AMH) is known to be associated with follicle growth, and its levels are two to three times higher in women with PCOS than in those with normal ovaries. We studied how duration of androgen administration affects folliculogenesis and AMH expression. METHODS We divided 30 immature (3-week-old) Sprague Dawley rats into six groups. Three groups were injected each evening with dehydroepiandrosterone (DHEA) (6?mg/100?g body weight/0.2?ml sesame oil) for 7, 15 or 30?days, respectively. The three control groups were injected with 0.2?ml of sesame oil for the corresponding lengths of time. Resected ovaries were sectioned and examined to determine follicle numbers at each developmental stage, and immunostained to assess AMH expression. RESULTS On day 7, follicle numbers and AMH expression levels at each developmental stage of follicle growth were similar in the respective control and DHEA groups. On day 15, the total follicle number (P?=?0.041), the percentage of primordial follicles (P?=?0.039) and AMH expression were significantly greater in the DHEA than the control group. On day 30, the percentages of primordial (P?=?0.005), primary (P?=?0.0002) and atretic (P?=?0.03) follicles were significantly greater in the DHEA group, whereas the percentage of intermediary follicles (early pre-antral, late preantral, and early antral follicles) was significantly lower in the DHEA group (P?=?<0.0001). AMH expression in DHEA-treated rats on day 30 was seen exclusively in the primordial (P?=?0.0413) and late antral follicles (p?=?0.028). CONCLUSIONS Androgen administration increases AMH production in a process that regulates the growth of primordial follicles. That is, androgen-induced AMH expression provides local negative feedback to folliculogenesis augmented by androgen. ///////////////////////// Evidence for a role for anti-Mullerian hormone in the suppression of follicle activation in mouse ovaries and bovine ovarian cortex grafted beneath the chick chorioallantoic membrane. Gigli I 2005 et al. The first critical transition in follicular development, the activation of primordial follicles to leave the pool of resting follicles and begin growth, is poorly understood, but it appears that the balance between inhibitory and stimulatory factors is important in regulating the exodus of follicles from the resting pool. There is evidence that anti-Mullerian hormone (AMH; also known as MIS) inhibits follicle activation in mice, but whether it plays a similar role in non rodent species is not known. When pieces of bovine ovarian cortex, rich in primordial follicles, are cultured in serum-free medium, most follicles initiate growth, but when cortical pieces are grafted beneath the chorioallantoic membrane (CAM) of chick embryos, follicle activation does not occur. Since embryonic chick gonads of both sexes produce and secrete high levels of AMH, the hypothesis that the AMH in the chick circulation inhibits follicle activation was tested. In Experiment 1, whole newborn mouse ovaries were grafted beneath the CAM (placed 'in ovo') or cultured in vitro for 8 days. In vitro (or after 8 days in vivo) follicles activated and proceeded to the primary or secondary stage, but activation was suppressed in ovo. This inhibition was reversed if ovaries were removed from beneath the CAM and cultured in vitro. In contrast, when ovaries from mice null mutant for the AMH type II receptor were CAM-grafted in Experiment 2, follicle activation occurred in a similar fashion to activation in vitro. This finding strongly implicates AMH as the inhibitor of follicle activation in ovo. Since chick embryonic gonads are the source of circulating AMH, chicks were gonadectomized in Experiment 3, prior to grafting of pieces of bovine ovarian cortex beneath their CAMs. Bovine primordial follicles activated in the gonadectomized chicks, similar to the results for mice lacking the AMH type II receptor. Taken together these experiments provide strong evidence that AMH is the inhibitor of mouse follicle activation present in the circulation of embryonic chicks and provide indirect, and hence more tentative, evidence for AMH as an inhibitor of bovine follicle activation. ///////////////////////// Antim?an hormone inhibits follicle-stimulating hormone-induced adenylyl cyclase activation, aromatase expression, and estradiol production in human granulosa-lutein cells. Chang HM et al. OBJECTIVE: To investigate the effects of antim?an hormone (AMH) on basal and FSH-induced cytochrome P450 aromatase (aromatase) expression and E2 production in human granulosa-lutein (hGL) cells, and to elucidate the mechanism by which AMH exerts its effects. DESIGN: Experimental study. SETTING: Academic medical center for reproductive science. PATIENT(S): The hGL cells were obtained from consenting patients undergoing IVF treatment. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Primary cultures of hGL cells were used to examine the effects of AMH (10 ng/mL) on basal and FSH (0.2 IU/mL)-stimulated E2 and intracellular cyclic adenosine 3':5' monophosphate (cAMP) accumulation, as well as aromatase and FSH receptor expression. Small interfering RNA targeting type II AMH receptor (AMHR2) was used to verify the specificity of the effects. RESULT(S): Treatment with AMH significantly reduced FSH-stimulated aromatase expression and E2 accumulation, whereas it had no measurable effects on basal and/or 8-Br-cAMP-stimulated levels. The FSH receptor messenger RNA and protein levels were not altered in AMH-treated cells. Cotreatment with AMH suppressed FSH-induced increases in intracellular cAMP. Knockdown of AMHR2 reversed the effects of AMH on aromatase expression. CONCLUSION(S): The AMH acts through AMHR2 to inhibit FSH-induced adenylyl cyclase activation, aromatase expression, and E2 production. The Role of Anti-Mullerian Hormone (AMH) During Follicle Development in a Monovulatory Species (Sheep). Campbell BK et al. Knockout studies in mice have suggested that anti-M?an hormone (AMH) modulates primordial follicle recruitment and the response of growing follicles to FSH. Little is known of the physiology of AMH in monovular species, despite intense clinical interest in this factor. Using sheep as a model, we sought to investigate the functional role of AMH in modulating follicle development in monovular species. In contrast to the rodent, the results indicate that AMH does not affect the rate of primordial follicle recruitment but appears to regulate the rate at which follicles progress through the gonadotropin-responsive phase, during which it is maximally expressed. Thus, knockdown of AMH bioactivity by active immunization lead to a decline in the population of gonadotropin-responsive preantral and small antral follicles (P < 0.01) and increases in both the number of gonadotropin-dependent antral follicles (P < 0.01) and ovulation rate (P < 0.05). These in vivo findings were consistent with the results of other studies examining the pattern of expression of AMH, which was negatively correlated with aromatase (P < 0.001), and in vitro supplementation experiments, which supported an inhibitory role for AMH in modulating the response of both theca and granulosa cells to LH and FSH, respectively. The elucidation of a functional relationship between AMH and LH-stimulated thecal androgen production may be significant in terms of the etiology of common forms of anovulatory infertility in women. Furthermore, the observed increase in both the number of recruitable antral follicles and ovulatory quota in response to AMH knockdown may have therapeutic value in women who respond poorly to ovarian stimulation. Anti-M?an hormone for the assessment of ovarian response in GnRH-antagonist-treated oocyte donors. Polyzos NP et al. Evidence regarding the role of anti-M?an hormone (AMH) among oocyte donors is limited and only involves gonadotrophin-releasing hormone (GnRH)-agonist-treated donors. This trial assessed the predictive ability of AMH for ovarian response among 108 oocyte donors treated with an antagonist protocol. In multivariate linear regression analysis, both AMH and age were independently associated with ovarian response (unstandardized coefficients 0.904 and -0.378, respectively). In receiver operating characteristic curve analysis, AMH performed better than age, but was a modest predictive marker for low (?6 oocytes) and excessive (>20 oocytes) ovarian response (area under the curve (AUC) 0.643 and 0.695, respectively). Similarly, a multivariate logistic model including AMH and age was also modest (AUC 0.651 and 0.697 for low and excessive responders, respectively). The predictive ability of AMH did not significantly alter when different thresholds were adopted, such as <4 oocytes for low response and >25 for excessive response (AUC 0.759 and 0.724, respectively). Among oocyte donors treated with a GnRH-antagonist protocol, although AMH was correlated with the number of oocytes retrieved, it demonstrates a modest ability in discriminating women with low or excessive ovarian response. Selection of oocyte donors is of paramount importance for the proper and more cost-efficient functionality of the oocyte donation programme. Despite the extensive literature regarding the efficacy of anti-M?an hormone (AMH) for predicting ovarian response among infertile patients, available evidence regarding the role of AMH in oocyte donors is considerably limited and involves only agonist down-regulated cycles. In this trial we assessed whether AMH can be considered a predictive marker for ovarian response among oocyte donors treated with a gonadotrophin-releasing hormone (GnRH)-antagonist protocol. According to our results, among oocyte donors treated with a GnRH-antagonist protocol, although AMH was correlated with the number of oocytes retrieved, the correlation is not strong and it appears that AMH has a modest predictive ability to discriminate women who are likely to demonstrate either impaired or excessive response to ovarian stimulation. Anti-M?an hormone reduces follicle sensitivity to follicle-stimulating hormone in human granulosa cells. Pellatt L et al. OBJECTIVE: In rodents and in luteinized granulosa cells (GC) anti-M?an hormone (AMH) has been shown to inhibit E(2) production. We determined whether this occurs in human cells most highly expressing AMH (i.e., from small antral follicles) and whether this is an effect on aromatase promoter activity. We also investigated the effects of AMH on other factors determining FSH sensitivity. DESIGN: Granulosa cells were exposed to AMH with and without gonadotropins for 48 hours. SETTING: University laboratory. PATIENT(S): Not applicable. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Aromatase and FSH receptor messenger RNA expression measured using real time quantitative polymerase chain reaction (PCR). Aromatase promoter II activity measured using a luciferase assay. Estradiol, inhibin A and B, and vascular endothelial growth factor production were measured in the conditioned medium. RESULT(S): The AMH decreased gonadotropin-stimulated aromatase expression and decreased forskolin-stimulated aromatase in KGN cells and this effect was through a dose-dependent inhibition of promoter II. Surprisingly, AMH also reduced FSH receptor mRNA expression. High AMH doses had no effect on inhibin B, whereas a low dose stimulated production. There was no effect on inhibin A or vascular endothelial growth factor. CONCLUSION(S): The AMH inhibits factors affecting FSH sensitivity. As AMH levels decrease with follicle growth, this inhibition would be removed. The AMH overproduction in anovulatory polycystic ovaries (PCO) may therefore restrict folliculogenesis by an inhibitory effect on FSH sensitivity, thereby contributing to anovulation. Inhibitory Actions of Anti-M?an Hormone (AMH) on Ovarian Primordial Follicle Assembly. Nilsson EE et al. The current study was designed to investigate the actions of Anti-M?an Hormone (AMH) on primordial follicle assembly. Ovarian primordial follicles develop from the breakdown of oocyte nests during fetal development for the human and immediately after birth in rodents. AMH was found to inhibit primordial follicle assembly and decrease the initial primordial follicle pool size in a rat ovarian organ culture. The AMH expression was found to be primarily in the stromal tissue of the ovaries at this period of development, suggesting a stromal-epithelial cell interaction for primordial follicle assembly. AMH was found to promote alterations in the ovarian transcriptome during primordial follicle assembly with over 200 genes with altered expression. A gene network was identified suggesting a potential central role for the Fgf2/Nudt6 antisense transcript in the follicle assembly process. A number of signal transduction pathways are regulated by AMH actions on the ovarian transcriptome, in particular the transforming growth factor - beta (TGF? signaling process. AMH is the first hormone/protein shown to have an inhibitory action on primordial follicle assembly. Due to the critical role of the primordial follicle pool size for female reproduction, elucidation of factors, such as AMH, that regulate the assembly process will provide insights into potential therapeutics to manipulate the pool size and female reproduction. The Rate of In Vitro Maturation of Primary Follicles From Adult Mice and the Quality of Oocytes is Improved in the Absence of Anti-Mullerian Hormone. Park JH et al. Anti-M?an hormone (AMH) inhibits the recruitment of primordial follicles into the growing pool, but its role in primary and secondary follicles is not clear. We isolated primary follicles from the ovaries of 9- to 10-week old mice and examined whether AMH affected follicular development. Follicles were matured in media that was prepared using unsexed fetal bovine serum (FBS) or female FBS (FFBS) with or without added AMH for approximately 2 weeks and maturation rates to secondary follicles and metaphase II (MII) oocytes were measured by standard morphological criteria. Rates of parthenogenetic activation and in vitro fertilization (IVF) were assessed by cleavage and blastocyst development, respectively. Whereas addition of AMH blocked primary to secondary follicle transition, the primary to secondary and secondary to MII follicle maturation rates was significantly improved with FFBS. Folliculogenesis resumed once AMH was removed from the media of the arrested primary follicles. The rates of IVF and parthenogenesis of oocytes after in vitro maturation (IVM) without AMH were also improved compared to controls. The results indicate that removal of AMH from culture conditions during IVM from primary follicular stages should be considered to improve outcome. Anti-Mullerian hormone and polycystic ovary syndrome: A mountain too high? Pellatt L et al. Anti-M?an hormone (AMH) was initially thought to be produced solely by the fetal male during sexual differentiation to promote regression of the M?an ducts. Over the last decade however a new and interesting role has emerged for AMH in the ovary. In human ovaries, AMH is produced by granulosa cells from 36 weeks gestation until menopause, with the highest expression being in small antral follicles. AMH production gradually declines as follicles grow, once follicles reach a size at which they are dominant it has largely disappeared. Its removal from these larger follicles appears to be an important requirement for dominant follicle selection and progression to ovulation as AMH has an inhibitory role in the ovary, reducing both primordial follicle initiation and follicle sensitivity to FSH by inhibition of aromatase. It is for this reason that AMH is a focus of interest in polycystic ovary syndrome. Serum levels are doubled and granulosa cell production is greatly increased. Interestingly, there appears to be 2 groups of women with PCOS who can be distinguished by their AMH level. Those who have high levels which do not reduce with treatment and who respond less well to induction of ovulation and a second group in whom the level is less elevated and reduces on treatment and who seem to respond rather better. Understanding the reason for the raised AMH in PCOS may give clues as to the mechanism of anovulation. To conclude, AMH appears to have a major inhibitory role during folliculogenesis, which may contribute to anovulation in PCOS. Josso N et al reviewed the role of anti-Mullerian hormone in gonadal development. AMH, a member of the transforming growth factor beta produced by immature Sertoli cells and, to a lesser degree, by granulosa cells from birth to the end of reproductive life, does not affect gonadal determination but has a negative effect upon gonadal development in both sexes. It blocks meiosis in fetal ovaries, leading to loss of germ cells and subsequent fibrous degeneration, and inhibits the transcription of aromatase and LH receptor. Vigier B, et al reported that anti-Mullerian hormone produces endocrine sex reversal of fetal ovaries by inhibiting aromatase acitvity. Alexandra L. L. et al (2001) reported that anti-M?an hormone attenuates the effects of FSH on follicle development in the mouse ovary. McGee EA, et al reported that Mullerian inhibitory substance induces growth of rat preantral ovarian follicles. Mullerian inhibitory substance (MIS), also known as anti-Mullerian hormone, is best known as the hormone that regulates the regression of the Mullerian duct in males. In females, MIS is expressed in granulosa cells of preantral and early antral follicles. The specific MIS type II receptor is present in granulosa and theca cells of these small, growing follicles. Because the role of MIS in preantral follicle development is unknown, we have evaluated the effect of MIS on the growth, differentiation, and apoptosis of intact preantral follicles in a serum-free culture system. In this system, treatment with FSH induces an increase in both follicle diameter, cell number, and follicle cell differentiation based on increased inhibin-alpha synthesis. Of interest, treatment with MIS enhances the effect of FSH both on follicle diameter and cell number. Although treatment with activin A also enhances FSH effects on follicle growth, treatment with transforming growth factor (TGF)-ss inhibits the FSH effects on follicle growth. Based on in situ staining of fragmented DNA, MIS was found to have no effect on follicle cell apoptosis, unlike its proapoptotic action on Mullerian ducts. In contrast to MIS and activin, TGF-ss was a potent proapoptotic factor for preantral follicles in culture. Analysis of inhibin-alpha expression of cultured preantral follicles further indicated that in contrast to activin, treatment with MIS did not enhance FSH-stimulated follicle differentiation. Thus, MIS is a unique factor that promotes preantral follicle growth but not preantral follicle cell differentiation and apoptosis. Schmidt KL, et al reported that Anti-Mullerian hormone initiates growth of human primordial follicles in vitro. Survival and growth of follicles in human ovarian tissue is presently only performed with limited success. We evaluated the effect of anti-Mullerian hormone (AMH) and/or testosterone on follicular growth during a 4-week culture period using ovarian cortical tissue from six women in their reproductive years. The cortex of each biopsy was isolated and immediately cryopreserved upon collection and stored in liquid nitrogen. After thawing the tissue was placed in culture. After the culture period all follicles were counted on histological sections and classified for viability and stage of development. Based on evaluation of 6603 follicles it was found that the number of growing follicles significantly increased during the culture period as compared to the uncultured control, irrespective of the composition of the culture medium. Furthermore, significantly more follicles advanced to the primary and secondary stage (p<0.05) in tissue cultured with AMH (54%) as compared to tissue cultured in control medium (41%). The mean diameter of follicles classified as primary follicles was significantly enhanced in tissue cultured in the presence of AMH (p=0.002) and AMH plus testosterone (p<0.001) as compared to that observed in tissue cultured with control medium and medium containing testosterone alone. In contrast the mean diameter of the oocyte and its nucleus remained similar irrespective of culture medium. In conclusion, AMH seems to affect early stages of human follicular development by enhancing recruitment, survival and/or growth during a 4-week culture period. Granulosa cell production of anti-Mullerian hormone is increased in polycystic ovaries. Pellatt L et al. Context: There has been renewed interest in AMH because of its role in the ovary. Data on its actions are sparse, but it appears to inhibit follicle growth. Interestingly, serum AMH is 2-3 times higher in women with polycystic ovary syndrome (PCOS) than in women with normal ovaries. Objective: We examined the production of AMH by cells from a range of follicle sizes from normal ovaries and compared this to production by ovulatory and anovulatory PCO. Design: Granulosa cells (GC), theca and follicular fluid (ff) were isolated from intact follicles. Cells were cultured for 48 h +/- FSH or LH and AMH was measured in ff and in cell-conditioned media (CM). Results: AMH levels in ff and GC-CM ranged from 42-2240ng/ml and 0.025-1.7ng/ml respectively and were low or undetectable in ff and GC-CM from follicles >9 mm, in luteinised cells and in theca and stroma. The mean level of AMH was 4 times higher in GC-CM from ovPCO; mean (range) 1.56 (0.025-7) and 75 times higher from anovPCO; 21.4 (17.2-43ng/ml) than from normal ovaries; 0.37 (0.025-1.7). Neither LH or FSH had an effect on AMH production by GC from normal ovaries, but in cells from PCO FSH significantly decreased AMH and in contrast, LH increased AMH. Conclusions: The reduction of AMH in follicles >9 mm from normal ovaries appears to be an important requirement for the selection of the dominant follicle. AMH production per GC was 75 times higher in anovPCO compared with normal ovaries. This increase in AMH may contribute to failure of follicle growth and ovulation seen in PCOS. M?an inhibiting substance and disrupted folliculogenesis in polycystic ovary syndrome. Wang JG et al. OBJECTIVE: This study determines whether pretreatment levels of m?an inhibiting substance/antim?an hormone (MIS/AMH) would reflect ovarian response to exogenous gonadotropin in women with polycystic ovary syndrome (PCOS) and ovulatory controls matched by age and weight. STUDY DESIGN: Case-control study of 20 women with PCOS and 10 normoovulatory women undergoing controlled ovarian hyperstimulation (COH) for in vitro fertilization (IVF) at an academic medical center. RESULTS: Baseline serum MIS/AMH levels in PCOS were higher than those of normoovulatory women (P < .001). MIS/AMH levels increased after gonadotropin-releasing hormone (GnRH) agonist pituitary suppression; 0.5 ng/mL in PCOS (P = .12) and 0.7 ng/mL in controls (P < .02). In normoovulatory women, MIS/AMH at baseline, after pituitary suppression, and the interval change after pituitary suppression all correlated closely to the number of mature oocytes retrieved (P < .005). In PCOS, however, levels of MIS/AMH at baseline and after pituitary suppression did not show this correlation, whereas only the interval change correlated with the number of mature oocytes retrieved. CONCLUSION: Baseline MIS/AMH is a good predictor of the ovarian response to COH in normoovulatory women but not in PCOS. M?an-inhibiting substance inhibits cytochrome P450 aromatase activity in human granulosa lutein cell culture. Grossman MP et al. OBJECTIVE: To investigate the effects of M?an-inhibiting substance (MIS) on cytochrome P450 aromatase (CYP19) gene expression in cultured human granulosa lutein cells (GLC). DESIGN: In vitro primary cell culture study. SETTING: Academic research laboratory and hospital-based fertility center. PATIENT(S): Eight normo-ovulatory patients undergoing IVF procedures due to male factor or tubal infertility. INTERVENTION(S): Serum and follicular fluid (FF) collected and stored at -80 degrees C until assayed. Granulosa lutein cells were harvested from follicular aspirates obtained during oocyte retrieval and cultured for 7 days with media in the presence or absence of MIS (10 ng/mL) or FSH 0.2 IU/mL. MAIN OUTCOME MEASURE(S): Serum and FF levels of E(2) and MIS, and E(2) production by GLC in culture. Levels of CYP19 mRNA in cultured GLC were determined by quantitative polymerase chain reaction (PCR) and CYP19 protein by Western blot. Statistical comparison used ANOVA and post hoc Tukey tests. RESULT(S): Follicle-stimulating hormone significantly increased E(2) production in cultured GLC compared with control. The increase in E(2) production is associated with higher levels of CYP19 mRNA and protein in GLC. The presence of MIS significantly inhibited FSH-induced E(2) production, with concomitant reduction in CYP19mRNA and protein levels. CONCLUSION(S): M?an-inhibiting substance inhibits FSH augmentation of CYP19 enzyme activity and CYP19 gene expression in GLC. These findings may help to explain the association of high MIS levels and low FF E(2) levels reported in women with polycystic ovary syndrome (PCOS). Anti-M?an hormone reduces follicle sensitivity to follicle-stimulating hormone in human granulosa cells. Pellatt L et al. OBJECTIVE: In rodents and in luteinized granulosa cells (GC) anti-M?an hormone (AMH) has been shown to inhibit E(2) production. We determined whether this occurs in human cells most highly expressing AMH (i.e., from small antral follicles) and whether this is an effect on aromatase promoter activity. We also investigated the effects of AMH on other factors determining FSH sensitivity. DESIGN: Granulosa cells were exposed to AMH with and without gonadotropins for 48 hours. SETTING: University laboratory. PATIENT(S): Not applicable. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Aromatase and FSH receptor messenger RNA expression measured using real time quantitative polymerase chain reaction (PCR). Aromatase promoter II activity measured using a luciferase assay. Estradiol, inhibin A and B, and vascular endothelial growth factor production were measured in the conditioned medium. RESULT(S): The AMH decreased gonadotropin-stimulated aromatase expression and decreased forskolin-stimulated aromatase in KGN cells and this effect was through a dose-dependent inhibition of promoter II. Surprisingly, AMH also reduced FSH receptor mRNA expression. High AMH doses had no effect on inhibin B, whereas a low dose stimulated production. There was no effect on inhibin A or vascular endothelial growth factor. CONCLUSION(S): The AMH inhibits factors affecting FSH sensitivity. As AMH levels decrease with follicle growth, this inhibition would be removed. The AMH overproduction in anovulatory polycystic ovaries (PCO) may therefore restrict folliculogenesis by an inhibitory effect on FSH sensitivity, thereby contributing to anovulation.
Expression regulated by FSH, LH, Steroids, Growth Factors/ cytokines, WT1, BMP4, AMH, FoxL2, GDF9, BMP15, Let7
Comment A novel, noncoding-RNA-mediated, post-transcriptional mechanism of AMH regulation by the H19/let-7 axis. Qin C et al. (2018) In reproductive age women, the pool of primordial follicles is continuously depleted through the process of cyclic recruitment. AMH both inhibits the initial recruitment of primordial follicles into the growing pool and modulates the sensitivity of growing follicles to FSH. Thus, AMH may be an important modulator of female infertility and ovarian reserve; however, the mechanisms regulating AMH remain unclear.To evaluate AMH levels in the absence of H19 lncRNA, H19 knockout (H19KO) mice were evaluated for analysis of ovarian AMH gene expression, protein production, and reproductive function, including assessment of follicle numbers and litter size analysis. To further investigate regulation of AMH by the H19/let-7 axis, let-7 binding sites on AMH were predicted, and in vitro studies of the effect of H19 knockdown/overexpression with let-7 rescue were performed. Lastly, response to superovulation was assessed via oocyte counts and estradiol measurements.The H19KO mouse demonstrates subfertility and accelerated follicular recruitment with increased spontaneous development of secondary, preantral and antral follicles. Ovaries of H19KO mice have decreased AMH mRNA and protein, and AMH mRNA has a functional let-7 binding site, suggesting a plausible ncRNA-mediated mechanism for AMH regulation by H19/let-7. Lastly, in the absence of H19, superovulation results in higher estradiol and more oocytes, suggesting that H19 functions to limit the number of follicles that mature, produce estradiol, and ovulate. Thus, AMH's inhibitory actions are regulated at least in part by H19, likely via let-7, marking this ncRNA pair as important regulators of the establishment and maintenance of the follicular pool.////////////////// Dysregulation of the anti-Müllerian hormone system by steroids in women with polycystic ovary syndrome. Pierre A et al. (2017) 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. 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 and PCOS women. Experiments were performed on follicular fluids (FF) and GCs from women undergoing in vitro fertilization. FF steroids levels were measured by mass spectrometry and mRNA accumulation was quantified by real-time RT-PCR. Total testosterone (T), free T and 5α-DHT FF levels were significantly higher (P<0.001) in PCOS than in controls. However, E2 and SHBG 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 up-regulated by 5α-DHT in GCs from PCOS patients (2.3-fold, p<0.01) overexpressing the androgen receptor (1.4-fold, p<0.05). Estradiol down-regulated 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 PCOS women. This differential effect of E2 was associated with a higher estrogen receptor one expression in GCs from PCOS women (1.9-fold, p<0.05). In GCs from PCOS women, the regulation of AMH and AMHR2 expression is altered in a way which promotes the overexpression of the AMH/AMHR2 system, and could contribute to the follicular arrest observed in these patients.////////////////// REGULATION OF AMH BY OOCYTE SPECIFIC GROWTH FACTORS IN HUMAN PRIMARY CUMULUS CELLS. Convissar S et al. (2017) The regulation of AMH production by follicular cells is poorly understood. The purpose of this study was to determine the role of the oocyte-secreted factors, growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15), on AMH production in primary human cumulus cells. Cumulus cells from IVF patients were cultured with a combination of GDF9, BMP15, recombinant FSH, and specific signaling inhibitors. Stimulation with GDF9 or BMP15 separately had no significant effect on AMH mRNA levels. In contrast, simultaneous stimulation with GDF9 and BMP15 (G+B) resulted in a significant increase in AMH mRNA expression. Increasing concentration of G+B (0.6, 2.5, 5 and 10 ng/ml) stimulated AMH in a dose-dependent manner, showing a maximal effect at 5 ng/ml. Western blot analyses revealed an average 16-fold increase in AMH protein levels in cells treated with G+B when compared to controls. FSH co-treatment decreased the stimulation of AMH expression by G+B. The stimulatory effect of G+B on the expression of AMH was significantly decreased by inhibitors of the SMAD2/3 signaling pathway. These findings show for the first time that AMH production is regulated by oocyte-secreted factors in primary human cumulus cells. Moreover, our novel findings establish that the combination of GDF9 + BMP15 potently stimulates AMH expression.////////////////// Positive cross talk between FOXL2 and antim?an hormone regulates ovarian reserve. Park M 2014 et al. OBJECTIVE To demonstrate interregulation between FOXL2 and antim?an hormone (AMH) in ovarian folliculogenesis. DESIGN Cell culture and animal study. SETTING University research laboratory. ANIMAL(S) Five-week-old B6C3F1 mice. INTERVENTIONS(S) Molecular analysis and in?vivo mouse experiment were performed to demonstrate that AMH is a target gene of FOXL2 in the ovary. MAIN OUTCOME MEASURE(S) To determine whether FOXL2 transactivates AMH, luciferase reporter assay, electrophoretic mobility shift assay, and chromatin immuniprecipitation were conducted. Using an in?vivo nucleic acid delivery system, the expression of AMH and/or FOXL2 was modulated in the mouse, and the ovaries were histologically analyzed. RESULT(S) AMH is an endogenous target gene of FOXL2. In contrast, mutated FOXL2s found in premature ovarian failure patients were defective in their ability to activate AMH transcription in human granulosa cells. In?vivo mouse gene delivery experiments revealed that Amh-knockdown accelerated follicle growth; however, the acceleration was prevented by ectopic expression of FOXL2. CONCLUSION(S) AMH and FOXL2 collaboratively work to reserve ovarian follicles. ///////////////////////// Anti-müllerian hormone regulation by the bone morphogenetic proteins in the sheep ovary: deciphering a direct regulatory pathway. Estienne A et al. (2014) 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.////////////////// Regulation of anti-M?an hormone production in the cow: a multiscale study at endocrine, ovarian, follicular, and granulosa cell levels. Rico C 2011 et al. Anti-M?an hormone (AMH) is an endocrine marker that can help predict superovulatory responses to treatments administered to cows for embryo production. However, the optimal time of the estrous cycle at which a blood test should be performed for a highly reliable prognosis has not yet been established. Moreover, little is known about the regulation of AMH production. To answer these questions, a study was designed to investigate the regulation of AMH production in cows selected for their high or low ovulatory responses to superovulation. At the granulosa cell level, AMH production was inhibited by follicle-stimulating hormone but enhanced by bone morphogenetic proteins. At the follicular level, the expression of AMH within the follicle was dependent on the stage of follicular development. At the ovarian level, the size of the pool of small antral growing follicles determined ovarian AMH production. At the endocrine level, AMH followed a specific dynamic profile during the estrous cycle, which occurred independently of the follicular waves of terminal follicular development. Cows selected for their high or low responses to superovulation did not differ in the regulation of AMH production, but cows with higher responses had higher plasma AMH concentrations throughout the cycle. The optimal period of the estrous cycle at which to measure AMH concentrations with the aim of selecting the best cows for embryo production was found to be at estrus and after Day 12 of the cycle. Based on this multiscale study, we propose a model that integrates the different regulatory levels of AMH production. ///////////////////////// Loss of LH-induced down-regulation of anti-Mullerian hormone receptor expression may contribute to anovulation in women with polycystic ovary syndrome. Pierre A et al. STUDY QUESTION: Are anti-M?an hormone (AMH) and AMH type II receptor (AMHR-II) mRNAs similarly regulated by gonadotrophins in lutein granulosa cells (GCs) from control, normo-ovulatory and oligo/anovulatory women with polycystic ovary syndrome (PCOS)? SUMMARY ANSWER: AMH mRNA expression was induced by LH only in lutein GC of oligo/anovulatory PCOS women; down-regulation of AMHR-II, induced by LH in control and normo-ovulatory PCOS women, was absent in oligo/anovulatory women. WHAT IS KNOWN ALREADY: It was suggested that AMH could be responsible for the blockade of follicles at the small antral stage in PCOS women. In keeping with this hypothesis, both AMH and AMHR-II are overexpressed in lutein GCs from oligo/anovulatory PCOS women. STUDY DESIGN, SIZE, DURATION: Women undergoing IVF were included in this prospective study, either in the control group (30 women) or in the PCOS group (21 normo-ovulatory and 19 oligo/anovulatory patients) between January 2010 and July 2012. PARTICIPANTS/MATERIALS, SETTING, METHODS: Human lutein GCs were isolated from follicular fluid during IVF protocols. Twenty-four hours after seeding, lutein GCs from each woman were serum starved and cultured for 48 h ? FSH, LH or cAMP. Then AMH and AMHR-II mRNAs were quantified by quantitative RT-PCR and AMH protein concentration was measured in the culture medium by ELISA. Experimental results were analyzed, within each group of women, by the non-parametric Wilcoxon test for paired comparisons between cells cultured in control medium and FSH, LH or cAMP treated cells. Clinical comparisons between the three groups of women were performed on log values using the ANOVA test with Bonferroni correction. MAIN RESULTS AND THE ROLE OF CHANCE: FSH up-regulated both AMH expression and secretion by lutein GCs from the three groups of women (P < 0.05). LH had no effect on AMH mRNAs levels in lutein GCs from controls and normo-ovulatory PCOS women, but increased AMH expression in oligo/anovulatory PCOS women (P < 0.05). Interestingly, LH and cAMP treatments reduced AMHR-II expression by lutein GCs from controls and normo-ovulatory PCOS women (P < 0.05), but had no effect on AMHR-II mRNA levels in oligo/anovulatory PCOS women. LIMITATIONS, REASONS FOR CAUTION: The lutein GCs are not the best model to study AMH and AMHR-II regulation by gonadotrophins. Indeed, AMH and AMHR-II are down-regulated in luteinized cells. Furthermore, these cells have been exposed to non-physiological levels of gonadotrophins and hCG. However, AMH and AMHR-II mRNAs are quantifiable by real-time RT-PCR, and the cells are still responsive to FSH and LH. The age of patients is significantly different between control and oligo/anovulatory PCOS women: this may be a bias in the interpretation of results but older women in the control group had a good ovarian reserve. WIDER IMPLICATIONS OF THE FINDINGS: The overexpression of AMH and AMHR-II in oligo/anovulatory PCOS women could be due to increased LH levels and/or inhibition of its repressive action. The fact that this dysregulation is observed in oligo/anovulatory, but not in normo-ovulatory, PCOS women emphasizes the role of LH in the follicular arrest of PCOS women and suggests that this involves the AMH/AMHR-II system. STUDY FUNDING/COMPETING INTEREST(S): The Assistance-Publique H?aux de Paris provided a Contrat d'Interface and the Agence de Biom?cine provided a grant to Nathalie di Clemente. Schering-Plough provided an FARO grant to Alice Pierre. The authors have nothing to disclose. Anti-Mullerian hormone (AMH) is induced by bone morphogenetic protein (BMP) cytokines in human granulosa cells. Ogura-Nose S et al. OBJECTIVES: Serum concentration of anti-Mullerian hormone (AMH) is used as a biomarker in clinical practice. Therefore, it is important to elucidate the mechanism by which AMH is regulated in granulosa cells (GC). An important first step in understanding AMH regulation is to determine which factors up-regulate AMH expression. STUDY DESIGN: Human GC, obtained from 28 women undergoing oocyte retrieval for in vitro fertilization, were stimulated with various intraovarian cytokines including bone morphogenetic protein (BMP)-2, -6, -7 -15, activin-A and growth differentiation factor (GDF)-9 (100ng/ml). The expression of AMH mRNA was evaluated with reverse transcription and quantitative real-time polymerase chain reaction (PCR), and AMH protein in cultured supernatant was measured with EIA kit. RESULTS: BMP-2, -6, -7 and -15, but not activin-A and GDF-9, significantly induced AMH expression in GC at mRNA and protein level, while all stimuli increased FSH receptor mRNA and decreased steroidogenic acute regulatory protein (StAR) mRNA level. CONCLUSIONS: Among the transforming growth factor (TGF)-?superfamily, BMP-2, -6, -7 and -15 significantly induced AMH expression in human GC. FSH and Its Second Messenger cAMP Stimulate the Transcription of Human Anti-Mullerian Hormone in Cultured Granulosa Cells. Taieb J et al. Anti-M?an hormone (AMH), also called M?an-inhibiting substance, a member of the TGF-?family, is responsible for the regression of M?an ducts in the male fetus. In females, AMH is synthesized by granulosa cells of preantral and small antral follicles, and production wanes at later stages of follicle maturation. Using RT-PCR in luteal granulosa cells in primary culture and reporter gene techniques in the KK1 granulosa cell line, we show that FSH and cAMP enhance AMH transcription, and LH has an additive effect. Gonadotropins and cAMP act through protein kinase A and p38 MAPK signaling pathways and involve the GATA binding factor-4 and steroidogenic factor-1 transcription factors, among others. The expression profile of AMH and the dynamics of serum AMH after gonadotropin stimulation have been interpreted as a down-regulating effect of FSH upon AMH production by granulosa cells. The specific effect of gonadotropins upon granulosa cells may be obscured in vivo by the effect of FSH upon follicular maturation and by the presence of other hormones and growth factors, acting individually or in concert. Anti-Mullerian hormone plasma levels in spontaneous menstrual cycle and during treatment with FSH to induce ovulation La Marca A, et al . BACKGROUND: Anti-M?an hormone (AMH) is member of the transforming growth factor-beta superfamily of growth factors. AMH is detected in serum from women of reproductive age and its levels vary slightly with the menstrual cycle, reaching the peak value in the late follicular phase. The present study was undertaken to assess the effect of controlled ovarian stimulation on AMH secretion by the ovary in healthy women in order to obtain more insight into the relationship between this peptide and gonadal steroids. METHODS: Twenty-four normally cycling women attending the infertility clinic volunteered for this study and AMH was measured in blood samples obtained during both spontaneous and FSH-treated cycles. RESULTS: AMH plasma levels did not change significantly from day 2 to day 6 in spontaneous cycles. On the contrary, AMH levels decreased progressively from day 2 to day 6 in FSH-treated cycles. A significant positive correlation was found between the decrease in AMH and the increase in estradiol plasma levels in FSH-treated cycles and between basal AMH and the peak estradiol (E2) during exogenous FSH administration. CONCLUSIONS: The present study demonstrated that AMH plasma levels did not change during the follicular phase of the menstrual cycle and that exogenous FSH administration is followed by a significant reduction in AMH levels which is probably secondary to the gonadotrophin effect on the process of follicular development. Serum Anti-Mullerian Hormone Concentrations Are Not Altered by Acute Administration of Follicle Stimulating Hormone in Polycystic Ovary Syndrome and Normal Women. Wachs DS et al. Context: In the human ovary, expression of anti-mullerian hormone (AMH) is detected primarily in granulosa cells of preantral and small antral follicles. This finding is consistent with the tight correlation between circulating AMH levels and the number of small antral follicles (2-5 mm) in normal and polycystic ovary syndrome (PCOS) women. In addition, the greater follicle count in PCOS is mirrored by significantly higher serum AMH levels compared to those of normal women. Despite the utility of AMH measurements in evaluating ovarian physiology and function, the regulation of AMH remains poorly understood. Objective: To determine whether gonadotropins acutely regulate serum AMH in women with PCOS and normal women. Design: Prospective study to compare ovarian responses to follicle stimulating hormone (FSH) in two groups of women. Setting: GCRC in a tertiary academic medical center. Patients: Women with PCOS, 18-35 years (n = 16), and normal ovulatory controls, 18-35 years (n = 11), were recruited for study. Interventions: Serum samples were measured over a 24 hour period following an intravenous injection of r-hFSH, 150IU. Main Outcome Measure(s): Serum AMH responses following FSH administration. Results: Basal serum AMH levels were markedly increased in women with PCOS compared to that observed in normal women. Following FSH injection, PCOS women failed to demonstrate changes in circulating AMH over 24 hours. A similar lack of alteration in serum AMH was observed in normal women. Conclusions: These findings suggest that in PCOS and normal women, acute exposure to FSH does not appear to exert an effect on AMH production. The Wilms tumor protein Wt1 is an activator of the anti-Mullerian hormone receptor gene Amhr2. Klattig J et al. The Wilms tumor protein Wt1 plays an essential role in mammalian urogenital development. WT1 mutations in humans lead to a variety of disorders including Wilms tumor, a pediatric kidney cancer, as well as Frasier and Denys-Drash syndrome. Phenotypic anomalies in the latter include pseudohermaphroditism and sex reversal in extreme cases. We have used cDNA microarray analyses on Wt1 knockout mice to identify Wt1-dependent genes involved in sexual development. The gene most dramatically affected by Wt1 inactivation was Amhr2 encoding the anti-M?an hormone (Amh) receptor 2. Amhr2 is an essential factor for the regression of the M?an duct in males and mutations in AMHR2 lead to the persistent M?an duct syndrome (PMDS), a rare form of male pseudohermaphroditism. Here we show that Wt1 and Amhr2 are co-expressed during urogenital development and that the Wt1 protein binds to the promoter region of the Amhr2 gene. Inactivation and over-expression of Wt1 in cell lines was followed by immediate changes of Amhr2 expression. The identification of Amhr2 as a Wt1-target provides new insights into the role of Wt1 in sexual differentiation and indicates - in addition to its function in early gonad development and sex determination - a novel function for Wt1, namely in M?an duct regression.
Ovarian localization Oocyte, Cumulus, Granulosa, Follicular Fluid
Comment Trends in anti-Müllerian hormone concentrations across different stages of pregnancy in women with polycystic ovary syndrome. Köninger A et al. (2018) What are the trends in anti-Müllerian hormone (AMH) concentrations from pre-conception to the third trimester of pregnancy in women with polycystic ovary syndrome (PCOS)? Observational study including cross-sectional and longitudinal data analysis. The Beckman Coulter AMH Gen II Assay was used to determine AMH levels longitudinally before pregnancy from 52 women with PCOS and 51 controls during all trimesters. Differences in AMH levels across successive stages of pregnancy were examined with the Wilcoxon signed-rank test for paired values. Linear regression models, adjusted for body-mass index (BMI), gestational and maternal age were used to compare AMH levels of PCOS and controls. AMH levels decreased significantly (all P < 0.05) from pre-pregnancy level throughout each trimester in women with PCOS and healthy controls. After adjusting for maternal age, gestational age and maternal BMI, AMH levels before pregnancy were 1.89 (95% CI 1.46 to 2.44; P < 0.0001) times higher among women with PCOS compared with controls (median 7.66 versus 2.67 ng/ml). During the first trimester, AMH levels were 1.61 (95% CI 1.22 to 2.13; P = 0.001) times higher among women with PCOS compared with controls (median 5.33 versus 2.48 ng/ml). Differences in AMH levels between women with PCOS and controls in the second trimester (1.68 times higher; 95% CI 0.94 to 3.01; median: 5.50 versus 2.20 ng/ml) and the third trimester (1.45 times higher; 95% CI 1.01 to 2.07; median: 1.36 versus 1.06 ng/ml) were not statistically significant. These findings indicate a pregnancy-associated AMH-decline independent of pre-pregnancy elevated AMH levels.////////////////// Most Cleaved Anti-Müllerian Hormone Binds Its Receptor in Human Follicular Fluid but Little Is Competent in Serum. Pierre A et al. (2017) 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. The objective of the study was to measure the levels of AMH cleavage and bioactivity in human body fluids. 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). 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. 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. 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.////////////////// Most cleaved anti-Müllerian hormone binds its receptor in human follicular fluid but little is competent in serum. J Clin Endocrinol Metab. 2016 Baseline AMH level associated with ovulation following ovulation induction in women with Polycystic Ovary Syndrome. Mumford SL et al. (2016) AMH reduces aromatase activity and sensitivity of follicles to FSH stimulation. Therefore, elevated serum AMH may indicate a higher threshold for response to ovulation induction in women with polycystic ovary syndrome (PCOS). To determine the association between AMH levels and ovulatory response to treatment among the women enrolled in the Pregnancy in PCOS II (PPCOS II) trial. Secondary analysis of data from a randomized clinical trial. Academic health centers throughout the United States. 748 women aged 18 to 40 years, with PCOS and measured AMH levels at baseline were included in this study. Couples were followed for up to 5 treatment cycles to determine ovulation (mid-luteal serum progesterone >5 ng/mL) and the dose required to achieve ovulation. A lower mean AMH and AMH per follicle was observed among women who ovulated compared to women who never achieved ovulation during the study (geometric mean AMH 5.54 vs. 7.35 ng/mL, p=0.0001; geometric mean AMH per follicle 0.14 vs 0.18, p=0.01) after adjustment for age, body mass index, testosterone, and insulin level. As AMH levels increased the dose of ovulation induction medication needed to achieve ovulation also increased. No associations were observed between antral follicle count and ovulation. These results suggest that high serum AMH is associated with a reduced response to ovulation induction among women with PCOS; women with higher AMH levels may require higher doses of medication to achieve ovulation.////////////////// Anti-mllerian hormone levels in the follicular fluid of the preovulatory follicle: a predictor for oocyte fertilization and quality of embryo. Kim JH 2014 et al. This prospective study investigated the relationship between anti-Mllerian hormone (AMH) level in the follicular fluid (FF) and the quality of the oocyte and embryo. A total of 65 FF samples from 54 women were included in this study. FF was collected from the largest preovulatory follicle sized=20 mm of mean diameter from each ovary. Samples were divided into 3 groups according to the FF AMH levels: below the 33th percentile (low group, FF AMH<2.1 ng/mL, n=21), between the 33th and the 67th percentile (intermediate group, FF AMH=2.1-3.6 ng/mL, n=22), and above the 67th percentile (high group, FF AMH>3.6 ng/mL, n=22). The quality of the ensuing oocytes and embryos was evaluated by fertilization rate and embryo score. FF AMH levels correlated positively with the matched embryo score on day 3 after fertilization (r=0.331, P=0.015). The normal fertilization rate was significantly lower in the low group than in the intermediate group (61.9% vs. 95.5% vs. 77.3%, respectively, P=0.028). Our results suggest that the FF AMH level could be a predictor of the ensuing oocyte and embryo quality. ///////////////////////// Follicular-fluid anti-Mullerian hormone (FF AMH) is a plausible biochemical indicator of functional viability of oocyte in conventional in vitro fertilization (IVF) cycles. Mehta BN 2013 et al. CONTEXT Oocyte quality may be a governing factor in influencing in vitro fertilization (IVF) outcomes. However, morphological evaluation of oocyte quality is difficult in conventional IVF cycles. Follicular-fluid (FF), the site for oocyte growth and development, has not yet been sufficiently explored to obtain a marker indicative of oocyte quality. Anti-Mullerian hormone (AMH) is produced by granulosa cells of preantral and early-antral follicles and is released in FF. AIM To investigate AMH as a biochemical indicator of functional viability/quality of oocyte produced in the FF micro-environmental milieu. SETTINGS AND DESIGN Prospective study involving 132 cycles of conventional IVF-embryo transfer (ET) in infertile women. SUBJECTS AND METHODS AMH concentration was estimated in pooled FF on day of oocyte pickup. Cycles were sorted into low and high groups according to median (50 (th) centile) values of measurement. Main outcome measure was oocyte viability, which included morphological assessment of oocyte quality, fertilization rate, clinical pregnancy, and implantation rates. STATISTICAL ANALYSIS Graph-pad Prism 5 statistical package. RESULTS Low FF AMH group shows significantly higher percentage of top-quality oocytes (65.08 ? 24.88 vs. 50.18 ? 25.01%, P =0.0126), fertilization (83.65 ? 18.38 vs. 75.78 ? 21.02%, P =0.0171), clinical pregnancy (57.57 vs. 16.67%, P >0.0001), and embryo implantation rates (29.79 vs. 7.69%, P >0.0001) compared to high FF AMH group. FF AMH shares an inverse correlation with FF E2 (Pearson r = -0.43, r(2) = 0.18) and clinical pregnancy (Pearson r = -0.46, r(2) = 0.21). Threshold value of FF AMH for pregnancy is >1.750 ng/mg protein. CONCLUSION FF AMH is a plausible biochemical indicator of functional viability of oocyte in conventional IVF cycles. ///////////////////////// Elevated serum level of anti-mullerian hormone in patients with polycystic ovary syndrome: relationship to the ovarian follicle excess and to the follicular arrest. Pigny P, et al . The serum level of anti-Mullerian hormone (AMH), a product from granulosa cells involved in follicle growth, has been shown to correlate tightly with the small antral follicle number (FN) at ultrasonography (U/S) in women who do not have polycystic ovary syndrome (PCOS). Because PCOS is associated with a 2- to 3-fold increase in growing FN, we investigated whether an increased AMH serum level correlates to other hormonal and/or U/S features of PCOS. Serum AMH has been assayed in 104 women (59 symptomatic PCOS, 45 controls) between d 2 and 7 after the last either spontaneous or progestin-induced (in PCOS) menstrual period. Mean serum AMH level was markedly increased in the PCOS group (47.1 +/- 22.9 vs. 20.8 +/- 11.6 pmol/liter in controls; P < 0.0001), an increase in the same order of magnitude as the one of the FN in the 2- to 5-mm range at U/S (12.8 +/- 8.3 vs. 4.8 +/- 1.9; P < 0.0001, respectively). The ratio AMH/FN was similar between the two groups (4.8 +/- 3.4 vs. 4.8 +/- 2.9; P = 0.55). By simple regression, both in PCOS and controls, the AMH level was positively related to the 2- to 5-mm FN at U/S (P < 0.0001 and P < 0.03, respectively), but not to the 6- to 9-mm FN, and was negatively correlated to the serum FSH level (P < 0.02 and P < 0.04, respectively). AMH was also positively related to the serum testosterone and androstenedione levels, in PCOS exclusively (P < 0.0005 and <0.002, respectively). No relationship was found between AMH and age, serum estradiol, inhibin B, and LH levels in both groups. After multiple regression only the 2- to 5-mm FN remained significantly related to AMH in PCOS whereas testosterone, androstenedione, and FSH were no longer. In conclusion, the assay of the serum AMH may represent an important breakthrough in the diagnosis and in the understanding of PCOS. Our data suggest that the increase of AMH serum level in PCOS is the consequence of the androgen-induced excess in small antral FN and that each follicle produces a normal amount of AMH. We hypothesize that an increased AMH tone within the cohort could be involved in the follicular arrest of PCOS, by interacting negatively with FSH at the time of selection. The circadian variation in anti-m?an hormone in patients with polycystic ovary syndrome differs significantly from normally ovulating women. Bungum L 2013 et al. OBECTIVE To improve the biologic understanding of the Polycystic Ovarian Syndrome (PCOS) condition by examining the circadian variation and relationship between Anti M?an Hormone (AMH), gonadotropins and ovarian steroids in PCOS patients compared to normally ovulating and menstruating women. By comparing the pattern of co-variation between AMH and Luteinizing Hormone, two compounds closely linked to hyperandrogenism and anovulation in PCOS, the involvement of the Hypothalamic-Pituitary-Ovarian axis in PCOS pathology could be elucidated. PATIENTS Eight normal-weighted young, anovulatory PCOS-women as study group and ten normal menstruating and ovulating women as controls. INTERVENTIONS Observational prospective study of the circadian variation in AMH, gonadotropins, sex steroids and androgens in a study and a control group. A circadian profile was performed in each study and control subject during a 24-h period by blood sampling every second hour, starting at 8:00 a.m. and continuing until 8:00 a.m. the following day. RESULTS Significant differences in hormonal levels were found between the groups, with higher concentrations of AMH, LH and androgens in the PCOS group and lower amounts of FSH and progesterone. A distinct difference in the circadian variation pattern of AMH and LH between PCOS patients and normal controls was seen, with PCOS patients presenting a uniform pattern in serum levels of AMH and LH throughout the study period, without significant nadir late-night values as was seen in the control group. In PCOS women, a significant positive association between LH/ FSH and testosterone was found opposite to controls. MAIN OUTCOME MEASURES Circadian variation in Anti-M?an Hormone, gonadotropins and ovarian steroids and the covariation between them. CONCLUSION A significant difference in the circadian secretion of LH and AMH in PCOS women compared to normally ovulating women indicate an increased GnRH pulse, creating high and constant LH serum concentrations. A significant co-variation between LH and AMH may suggest LH as a factor involved in the control of AMH secretion. ///////////////////////// Over-Expression of M?an Inhibiting Substance mRNA in the Turner Syndrome Ovary. Modi D et al. Turner Syndrome (TS) is a disorder of human females associated with complete or partial loss of one of the X chromosomes, varying degrees of multiple congenital malformations and gonadal dysgenesis. However, the reason for the premature loss of germ cells in the TS ovaries is currently unknown. To understand the molecular basis of the gonadal dysgenesis the mRNA expression of M?an Inhibiting Substance (MIS) was examined in human fetal and adult TS ovaries and compared with normal ovaries by in situ hybridization. The expression of MIS was found to be increased in the granulosa cells of the TS ovaries as compared to that in normal ovaries, and these granulosa cells were organized to form testicular tubule like structures. MIS was also found to be ectopically expressed in the oocytes of the developing TS gonads. The stromal cells of the streak gonads of adult TS women abundantly expressed MIS. We speculate that the absence of a second X chromosome leads to over-expression of MIS that may be co-responsible for failure of ovarian differentiation in TS. MIS may be a potential negative regulator of ovarian development in humans. Anti-M?an hormone remains highly expressed in human cumulus cells during the final stages of folliculogenesis. Gr?l ML et al. This study evaluated whether anti-M?an hormone (AMH) was differentially expressed in cumulus (CC) and granulosa (GC) cells from large antral and pre-ovulatory follicles collected from individual follicles in women undergoing in-vitro maturation (IVM) or IVF treatment. Expression studies of AMH, AMH receptor 2, FSH receptor, aromatase and androgen receptor were performed in CC in IVM patients where cumulus-oocyte-complex had expanded, CC in IVM patients where cumulus-oocyte-complex remained compacted, GC from immature follicles and CC and GC from IVF patients. Microarray data on corresponding GC and CC from follicles from IVF patients was included. AMH expression was significantly higher in CC than in GC from both mature and immature follicles and in CC from immature follicles than in CC from pre-ovulatory follicles from IVF patients (P<0.05). AMH expression was significantly higher in CC that remained compacted compared with those that had expanded (P<0.008). AMH was correlated to the expression of FSH receptor, androgen receptor and AMH receptor 2 but not to aromatase expression. The expression pattern of AMH receptor 2 reflected that of AMH. AMH may exert intrafollicular functions even in human large antral and pre-ovulatory follicles and may be related to follicular health. During folliculogenesis, the cells surrounding the oocyte differentiate from one to two cell types: granulosa cells that mainly ensure hormone and growth factor production and cumulus cells that nurse the oocyte. This study focused on differences in gene activity between the two cell types and between late stages of follicle development. The genes investigated included anti-M?an hormone (AMH), which was found to be differentially expressed in the two cell types and between the developmental stages. Strong correlation was found between AMH gene activity and that of several other central genes that has been suggested to play a role in the health of the follicle.
Follicle stages Primary, Secondary, Antral
Comment Anti-Müllerian hormone is produced heterogeneously in primate preantral follicles and is a potential biomarker for follicle growth and oocyte maturation in vitro. Xu J et al. (2016) The main goals of this study were to investigate the expression of anti-Müllerian hormone (AMH) and its receptor (AMHR2) during follicular development in primates, and to evaluate the potential of AMH as a biomarker for follicle growth and oocyte maturation in vitro. The mRNA and protein expression of AMH and AMHR2 were determined using isolated follicles and ovarian sections from rhesus macaques (n = 4) by real-time PCR and immunohistochemistry, respectively. Isolated secondary follicles were cultured individually. Follicle growth and media AMH concentrations were assessed by ELISA. The mRNA expression profiles, obtained from RNA sequencing, of in vitro- and in vivo-developed antral follicles were compared. Secondary follicles from additional animals (n = 35) were cultured. Follicle growth, oocyte maturation, and media AMH concentrations were evaluated for forecasting follicular development in vitro by AMH levels. AMH immunostaining was heterogeneous in the population of preantral follicles that were also stained for AMHR2. The mRNA expression profiles were comparable between in vivo- and in vitro-developed follicles. AMH levels produced by growing follicles were higher than those of nongrowing follicles in culture. With a cutoff value of 1.40 ng/ml, 85 % of nongrowing follicles could be identified while eliminating only 5 % of growing follicles. Growing follicles that generated metaphase II-stage oocytes secreted greater amounts of AMH than did those yielding immature germinal vesicle-stage oocytes. AMH, co-expressed with AMHR2, was produced heterogeneously by preantral follicles in macaques with levels correlated positively with follicle growth and oocyte maturation. AMH may serve as a biomarker for primate follicular development in vitro.////////////////// Anti Müllerian Hormone (AMH) level and expression in mural and cumulus cells in relation to age. Kedem A et al. (2014) Serum AMH is declining with age and is highly associated with ovarian follicular reserve and disordered folliculogenesis. However, the precise role of AMH in the process of human follicular aging has still to be determined. This study investigates AMH level in the follicular fluid (FF) and mRNA expression pattern in cumulus and mural granulosa cells of human ovarian follicles in relation to age. We conducted a prospective study. Sixty-eight women undergoing In vitro fertilization (IVF) treatment were enrolled in the study. We obtained FF, mural and cumulus granulosa cells from large preovulatory follicles (17-20 mm) of 21-35 years old women (n = 40) and 40-45 years old women (n = 28) during oocyte pickup. Higher level of AMH mRNA expression in cumulus cells was observed in the older age group compared to the younger (P <0.01). In accordance with AMH mRNA expression results, FF AMH protein levels were significantly higher in the older group than in the younger group (4.7 ± 1.1 ngml and 2.3 ± 0.2 ngml respectively, p < 0.002). AMH is highly expressed and secreted from cumulus GCs of advanced age patients. This remarkable correlation between AMH mRNA levels in cumulus cells in respect to age suggests that AMH may be involved in follicular aging process.////////////////// Dynamics of anti-M?an hormone during controlled ovarian stimulation. B?her B 2013 et al. Abstract Aim: To evaluate the dynamics of anti-M?an hormone (AMH) during controlled ovarian stimulation (COH) and to correlate changes in AMH to age, estradiol (E2) levels, and the presence of polycystic ovary syndrome (PCOS). Methods: Data were retrospectively collected from women presenting for COH in the outpatient clinic of a university hospital between January and July 2011. Concentrations of AMH and E2 during COH with gonadotropins for in vitro fertilization (IVF) (n?=?68) and clomiphene or low-dose gonadotropin stimulation cycles (n?=?27) for intrauterine insemination were evaluated. Percentage change in AMH and E2 from pre-stimulation values was calculated. Dynamics of hormonal changes were analyzed using non-parametric tests. Correlations between changes in AMH and E2 were analyzed with Spearman correlation. Results: During IVF stimulation, AMH declined steadily from pre-stimulation values. No significant change in AMH dynamics was observed during clomiphene or low-dose stimulation cycles. Percentage decline in AMH during IVF stimulation correlated with rise in E2 at all time points. Conclusions: The observed phenomena contribute to an improved understanding of AMH expression and its role in the follicular development. Our data support the concept that AMH is produced by secondary, preantral and small antral follicles in the early part of stimulation and declines as these follicles are recruited into dominant growing follicles. ///////////////////////// Developmental expression and cellular distribution of Mullerian inhibiting substance in the primate ovary. Modi D 2006 et al. Ovarian follicle formation during development and follicle maturation in adulthood are crucial determinants of female fertility and disruptions in these processes may result in subfertility or infertility. Among the several factors that are involved in ovarian physiology, M?an inhibiting substance (MIS) also known as anti-M?an hormone has emerged as an important marker to predict the follicle reserve. However, the roles of MIS in human ovarian physiology are unknown. To gain an insight into the potential roles of MIS in human ovarian differentiation during development and its regulation in adulthood, the expression profiles of MIS mRNA in the developing and adult human and monkey ovaries was examined by in situ hybridization. The results revealed that in the fetal human ovaries, MIS is specifically expressed at low levels in the granulosa cells of the developing primordial follicles; a small subset (approximately 2-3%) of oocytes express high amounts of MIS. In the adult human and monkey ovary, MIS mRNA is expressed at low levels in the primordial follicles, maximally in the primary and secondary follicles, and the expression is downregulated in the antral and atetric follicles. MIS expression is extinguished in the granulosa cells only after ovulation. These observations strongly favor the regulatory roles of MIS in folliculogenesis. MIS in the primate ovary may exert its effect during the primordial follicle formation to the terminal granulosa cell differentiation. The presence of MIS in a small subset of oocytes in the fetal ovary further points towards its additional role during fetal oocyte development. ///////////////////////// Mullerian inhibiting substance in the adult rat ovary during various stages of the estrous cycle. Ueno S et al. Mullerian inhibiting substance (MIS) in the adult rat ovary can be detected by immunohistochemistry in the granulosa cells of growing preantral follicles and in the cumulus oophorus and periluminal granulosa cells of antral follicles in estrus, metestrus, diestrus, and proestrus. Neither the corpus luteum nor atreitic follicles stained for MIS. During proestrus, dramatic changes occurred in the large preovulatory antral follicles, which early in the day manifest intense MIS-specific staining in the granulosa cells located close to the oocyte, however, at 2300 h, just before ovulation when the germinal vesicle was extruded to indicate resumed meiotic division, MIS staining waned. When the morphology of the late preovulatory stage was created experimentally in 26-day-old immature ovaries by stimulating 48 h earlier with hCG, the intense staining of the granulosa cells surrounding the oocytes from untreated ovaries was lost in the cumulus cells of such hCG-treated animals. This temporal pattern of MIS staining and the prior demonstration that MIS could inhibit in vitro meiosis of oocytes from untreated immature rats suggest that this regressor, well characterized in the fetal testis, might function in the ovary as a regulator of oocyte maturation and follicular development during the adult reproductive cycle.Immunocytochemical study of anti-M?an hormone in sheep ovarian follicles during fetal and post-natal development. B?rd J 1987 et al. Anti-M?an hormone (AMH) was detected in perinatal and postnatal sheep ovaries, using avidin-biotin immunohistochemistry with a monoclonal antibody specific for ruminant AMH. Immunoreactivity was limited to granulosa cells, and was influenced both by the degree of follicular development, and by the age of the animal. In the fetus, only the most advanced follicles exhibited a faint immunoreactivity at 120 days gestation, and no reaction was observed in younger animals. Immediately before and after birth, primordial follicles were still negative, but a faint reaction was elicited in young growing follicles, increasing with follicle size. Strong immunoreactivity was visible in antral follicles, especially in the innermost granulosa cell layers, close to the oocyte and lining the antral cavity. ///////////////////////// Rajpert-De Meyts E et al reported the expression of anti-Mullerian hormone during normal and pathological gonadal development: association with differentiation of Sertoli and granulosa cells. The ontogeny of expression of anti-Mullerian hormone (AMH) was examined by immunohistochemistry in 135 human gonadal tissue specimens of various developmental age, ranging from 6 weeks of fetal development to 38 yr of postnatal age. AMH expression was found only in Sertoli and granulosa cells. In fetal ovaries, AMH was first detected at 36 weeks gestation in granulosa cells of preantral follicles. Serum Anti-Mullerian Hormone as a Surrogate for Antral Follicle Count for Definition of the Polycystic Ovary Syndrome Pigny P, et al . Context: Despite its frequency, the Polycystic ovary syndrome (PCOS) is still a difficult diagnosis in endocrinology, gynaecology and reproductive medicine. To help solving this issue, the Rotterdam consensus conference proposed to include the ultrasonographic (U/S) follicle count as a new diagnostic criterion, in addition to hyperandrogenism and oligo-anovulation. Unfortunately its assessment does not offer sufficient reliability worldwide. Objective: The aim of our study was to check whether AMH measurement in the serum could be a surrogate for antral follicle count in the diagnostic criteria of PCOS. Design, setting and patients: Measurement of serum AMH with a 2(nd) generation immunoassay in a cohort of seventy-three PCOS patients and 96 controls, and evalutation of its diagnostic power by Receiver Operating Characteristic (ROC) curves. PCOS was diagnosed according to the Rotterdam definition. Results: Serum AMH levels were 3-fold higher in PCOS patients than in controls (81.6 pmol/L vs. 33.5 pmol/L, P < 0.001) and were significantly related to the follicle number (FN) in the 2 groups. The area under the ROC curve for the AMH assay was 0.851, indicating a good diagnostic potency. Setting the threshold at 60 pmol/L offered the best compromise between specificity (92%) and sensitivity (67%). Conclusions: The serum AMH level is an accurate marker of the ovarian early antral FN and offers a good diagnostic potency. In situations where accurate U/S data are not available, AMH could thus be used instead of the follicle count as a diagnostic criterion and incorporated as such in the Rotterdam definition of PCOS. Anti-M?an hormone and inhibin B variability during normal menstrual cycles. Sowers M et al. OBJECTIVE: To describe anti-M?an hormone (AMH) variation across normal menstrual cycles. DESIGN: Cohort study. SETTING: Academic environment. PATIENT(S): Twenty regularly menstruating women. INTERVENTION(S): Serum AMH and inhibin B assayed daily during one normal menstrual cycle. MAIN OUTCOME MEASURE(S): Intracycle variability of AMH and inhibin B. RESULT(S): Data were classified into quartiles of AMH area-under-the-curve (AUCs). Mean AMH AUC was 15.7 ng/mL for quartile 1 versus 43.5, 80.9 and 144.9 ng/mL for quartiles 2, 3, and 4. Mean AMH levels (ng/mL) were 0.67, 1.71, 3.02, and 5.33, respectively. There was no variation in quartile 1 AMH rate of change from stochastic modeling, but in quartiles 2 to 4, there were increased rates of change in days 2 to 7. Women in quartile 1 had the lowest mean inhibin B (24.2 pg/mL vs. 44.3, 43.2, and 42.2 pg/mL), and had shorter menstrual cycles (24.6 days) than women in quartiles 3 and 4 (28.2 and 28.4 days). CONCLUSION(S): There were two menstrual cycle patterns of AMH. The 'aging ovary' pattern included low AMH levels with little variation, lower inhibin B, and shorter cycle lengths. The 'younger ovary' pattern included higher AMH levels with significant variation days 2 to 7, suggesting that for women with AMH >1 ng/mL, the interpretation of AMH levels is contingent upon the day of the menstrual cycle on which the specimen is obtained.
Phenotypes PCO (polycystic ovarian syndrome)
POF (premature ovarian failure)
Mutations 13 mutations

Species: mouse
Mutation name: None
type: null mutation
fertility: fertile
Comment: Increased oocyte degeneration and follicular atresia during the estrous cycle in AMH null mice. Visser JA et al. Anti-M?an hormone (AMH) plays an important role in folliculogenesis. AMH null mice display an increased recruitment of primordial follicles. Nevertheless, these mice do not have proportionally more preovulatory follicles. Therefore, AMH null mice provide an interesting genetic model to study the regulation of species-specific number of preovulatory follicles. We studied the follicle pool throughout the estrous cycle at four months of age. Analysis of the follicle pool revealed that AMH null mice have an increased and earlier cyclic recruitment of growing follicles despite a blunted FSH surge at estrus. However, FSH levels at estrus were apparently too low to support growth to the preovulatory stage since an increased level of atresia was observed, which neutralized the increased cyclic recruitment. When AMH null mice were subjected to a superovulation scheme, the rise in FSH levels resulted in the rescue of the recruited cohort of growing follicles. Analysis of the follicle pool also revealed that the increased recruitment of primordial follicles in AMH null mice was neutralized by an increased loss of follicles during the transition from small preantral to large preantral follicle. This major loss of follicles was not completely reflected by a corresponding augmentation of atresia, but did correspond with an increased number of oocyte remnants observed in AMH null mice. We conclude that a combination of increased oocyte degeneration and increased follicular atresia neutralizes the increased initial and cyclic recruitment in AMH null mice to a normal number of preovulatory follicles.

Species: human
Mutation name: None
type: naturally occurring
fertility: subfertile
Comment: A functional Anti-Mullerian Hormone Gene polymorphism is associated with follicle number and androgen levels in Polycystic Ovary Syndrome patients. Kevenaar ME et al. Context: The common characteristic of Polycystic Ovary syndrome (PCOS) is a disturbance in the selection of the dominant follicle resulting in anovulation. In PCOS women serum Anti-M?an Hormone (AMH) levels are elevated. As AMH decreases FSH sensitivity in mice, the elevated AMH levels may contribute to the disturbed follicle selection in PCOS women. Objective: To investigate the role of the AMH signaling pathway in the pathophysiology of PCOS using a genetic approach. Design: The association of the AMH Ile(49)Ser (rs10407022) and the AMH type II receptor -482 A>G (rs2002555) polymorphism with PCOS susceptibility and phenotype was studied in large a cohort of PCOS women. Setting/ Subjects: 331 women with PCOS, 32 normo-ovulatory controls and 3635 population based controls were included. Main Outcome Measures: Ovarian parameters, serum AMH, FSH, androgen and estradiol levels were measured. Results: Genotype and allele frequencies for the AMH Ile(49)Ser and AMHR2 -482 A>G polymorphism were similar in PCOS women and controls. However, within the group of PCOS women, carriers of the AMH (49)Ser allele had less often polycystic ovaries (92.7% versus 99.5%, P=0.0004), lower follicle numbers (P=0.03) and lower androgen levels compared to non-carriers (P=0.04). In addition, in vitro studies demonstrated that the bioactivity of the AMH (49)Ser protein is diminished compared to the AMH (49)Ile protein (P<0.0001). Conclusions: Genetic variants in the AMH and AMHR2 gene do not influence PCOS susceptibility. However, our results suggest that the AMH Ile(49)Ser polymorphism contributes to the severity of the PCOS phenotype. [

Species: human
Mutation name: None
type: naturally occurring
fertility: subfertile
Comment: First evidence of genetic association between the MIF-173G/C single-nucleotide polymorphisms and polycystic ovary syndrome. Li C 2011 et al. PROBLEM The purpose of this study was to investigate whether polymorphism of MIF gene is associated with PCOS. METHOD OF STUDY The MIF-173G/C single-nucleotide polymorphism (SNP) was detected in 529 PCOS patients and 585 healthy female controls of Chinese Han ancestry. The association of the gene variants with clinical and metabolic parameters and hormone levels was investigated. RESULTS The frequencies of genotypes and allelotypes of the MIF-173G/C SNP did significantly differ between women with PCOS and healthy controls (P = 0.017 and P = 0.003, respectively). They did significantly differ between obese PCOS patients and obese controls (P = 0.029 and P = 0.039, respectively). The MIF-173 CC and CG genotypes were associated with higher body mass index (BMI) and waist-to-hip ratio (WHR) in both PCOS patients (P < 0.001, P = 0.001) and normal controls (P < 0.001, P = 0.002). The PCOS patients with CC and CG genotypes had higher fasting plasma glucose levels (P < 0.001), higher fasting insulin levels (P < 0.001), and higher HOMA-IR (P < 0.001) compared with patients with the GG genotype. CONCLUSION The MIF-173G/C polymorphism is associated with PCOS in Chinese Han women and may contribute to the phenotypic expression of PCOS. /////////////////////////

Species: mouse
Mutation name: None
type: targeted overexpression
fertility: infertile - ovarian defect
Comment: The m?an inhibitor and mammalian sexual development. Behringer RR 1996 et al. The elegant embryological experiments of Jost demonstrated the existence of a foetal testicular factor that is required to cause the regression of the m?an duct system, the anlagen of the uterus, oviducts and upper portion of the vagina, during male sexual development. The m?an inhibitor currently known as m?an-inhibiting substance (MIS) or anti-m?an hormone (AMH), is a member of the transforming growth factor-beta (TGF-beta) family of growth and differentiation factors. The genetic manipulation of the mouse germline has lead to the generation of animal models for MIS function. Female transgenic mice that chronically express MIS during embryogenesis are born without a uterus or oviducts and their ovaries lose germ cells and degenerate, recapitulating the phenotype of the bovine freemartin. Some male transgenic mice from very high MIS-expressing lines are feminized, suggesting alterations in androgen biosynthesis. Male mice homozygous for a targeted mutation of the MIS gene develop as male pseudohermaphrodites with both male (testes and Wolffian duct-derived) and female (m?an duct-derived) reproductive organs. Most are infertile because the development of two reproductive systems physically blocks the exit of sperm from these males. In addition, Leydig cell hyperplasia is detected in a proportion of these males and in one case a Leydig cell tumour was found. Recently, a gene encoding a TGF-beta family type II Ser/Thr kinase membrane-bound receptor has been isolated that is expressed in both male and female gonads and in the mesenchyme surrounding the m?an ducts during embryogenesis. These findings suggest that MIS-mediated m?an duct regression occurs indirectly through mesenchymal tissue. A targeted mutation of this receptor has been established in the mouse germline. Mice homozygous for this receptor mutation should be useful in understanding the MIS signalling pathway for m?an duct regression and gonadal function. /////////////////////////

Species: mouse
Mutation name: None
type: targeted overexpression
fertility: infertile - ovarian defect
Comment: A novel AMH-Treck transgenic mouse line allows toxin-dependent loss of supporting cells in gonads. Shinomura M 2014 et al. Cell ablation technology is useful for studying specific cell lineages in a developing organ in vivo. Herein, we established a novel anti-M?an hormone (AMH)-toxin receptor-mediated cell knockout (Treck) mouse line, in which the diphtheria toxin (DT) receptor was specifically activated in Sertoli and granulosa cells in postnatal testes and ovaries, respectively. In postnatal testes of AMH-Treck transgenic (Tg) male mice, DT injection induced a specific loss of the Sertoli cells in a dose-dependent manner, as well as the specific degeneration of granulosa cells in the primary and secondary follicles caused by DT injection in Tg females (high-dose treatment). In the testes with Sertoli cell depletion, germ cells appeared to survive for only several days after DT treatment and rapidly underwent cell degeneration, which led to the accumulation of a large amount of cell debris within the seminiferous tubules by day 10 after DT treatment. Transplantation of exogenous healthy Sertoli cells following DT treatment rescued the germ cell loss in the transplantation sites of the seminiferous epithelia, leading to a partial recovery of the spermatogenesis. These results provide not only in vivo evidence of the crucial role of Sertoli cells in maintaining the germ cells, but also show that the AMH-Treck Tg line is a useful in vivo model of the function of the supporting cell lineage in developing mammalian gonads. /////////////////////////

Species: mouse
Mutation name:
type: null mutation
fertility: fertile
Comment: Durlinger AL et al 1999 reported the control of primordial follicle recruitment by anti-Mullerian hormone in the mouse ovary. Female AMH null mice were reported to be fertile, with normal litter size, but this does not exclude a more subtle function for ovarian AMH. To investigate the function of AMH in the ovary, the complete follicle population was determined in AMH null mice, in mice heterozygous for the AMH null mutation, and in wild-type mice of different ages: 25 days, 4 months, and 13 months. It was found that ovaries of 25-day- and 4-month-old AMH null females, compared to those of wild-type females, contain more preantral and small antral follicles. In addition, in 4- and 13-month-old AMH null females, smaller numbers of primordial follicles were found. Actually, in 13-month-old AMH null females, almost no primordial follicles could be detected, coinciding with a reduced number of preantral and small antral follicles in these females. In almost all females heterozygous for the AMH null mutation the number of follicles fell in between the numbers found in wild-type and AMH null females. Furthermore, the weight of the ovaries was twice as high in the 4-month-old AMH null females as in age-matched wild-type females. It was concluded that AMH plays an important role in primordial follicle recruitment, such that more primordial follicles are recruited in AMH null mice than in wild-type mice; the mice heterozygous for the AMH null mutation take an in-between position. Consequently, the ovaries of AMH null females and those of females heterozygous for the AMH null mutation will show a relatively early depletion of their stock of primordial follicles. The female AMH null mouse may thus provide a useful model to study regulation of primordial follicle recruitment and the relation between follicular dynamics and ovarian aging. Mishina et al. (1996) produced and examined AMH receptor 2 knockout mice. They observed that mutant males were internal pseudohermaphrodites, having both male and female reproductive organs. The phenotype of AMH/AMHR2 double-knockout mutant males was indistinguishable from that of either single mutant. Furthermore, the phenotypes of AMH/alpha-inhibin and AMHR2/alpha-inhibin double-knockout mutant males were also identical, suggesting to the authors that AMH is the only ligand of the AMHR2 receptor. Behringer RR reviewed genetic animal models for MIS function. Female transgenic mice that chronically express MIS during embryogenesis are born without a uterus or oviducts and their ovaries lose germ cells and degenerate, recapitulating the phenotype of the bovine freemartin. Some male transgenic mice from very high MIS-expressing lines are feminized, suggesting alterations in androgen biosynthesis. Male mice homozygous for a targeted mutation of the MIS gene develop as male pseudohermaphrodites with both male (testes and Wolffian duct-derived) and female (mullerian duct-derived) reproductive organs. Most are infertile because the development of two reproductive systems physically blocks the exit of sperm from these males.

Species: human
Mutation name:
type: naturally occurring
fertility: subfertile
Comment: Association study of anti-Mullerian hormone and anti-Mullerian hormone type II receptor polymorphisms with idiopathic primary ovarian insufficiency. Yoon SH 2013 et al. STUDY QUESTION Are the genetic polymorphisms of the anti-M?an hormone (AMH) and anti-M?an hormone type II receptor (AMHR2) genes associated with idiopathic primary ovarian insufficiency (POI) in a Korean population? SUMMARY ANSWER The distribution of the AMH and the AMHR2 polymorphisms in a Korean POI population was not significantly different from controls. WHAT IS KNOWN ALREADY AMH plays an important role in regulating both the primordial follicle recruitment and the cyclic selection of the antral follicles. The AMHR2 -482A>G polymorphism was associated with an earlier menopause and nulliparous women with the GG genotype had a 2.6 years earlier onset of menopause compared with the AA genotype women. Therefore, genetic variants in the AMH signal transduction pathway might affect the ovarian function of women. STUDY DESIGN, SIZE, DURATION Case-control study. The subjects consisted of 211 idiopathic POI patients and 233 post-menopausal controls. PARTICIPANTS/MATERIALS, SETTING, METHODS The frequency of the AMH Ile(49)Ser and AMHR2 -482A>G polymorphisms was analyzed in 211 patients with idiopathic POI and in 233 post-menopausal controls, and we also analyzed clinical characteristics, such as age at the time of POI and LH, FSH?as well as estradiol levels according to the specific genotype. Genotyping for the AMH Ile(49)Ser and the AMHR2 -482A>G polymorphisms was performed by a minor groove binder primer/probe Taqman assay. MAIN RESULTS AND THE ROLE OF CHANCE The genotype distributions and allele frequencies for the AMH Ile(49)Ser and the AMHR2 -482A>G polymorphisms were similar between the POI patients and the controls. Within POI population, the AMH Ile(49)Ser and the AMHR2 -482A>G polymorphisms were not associated with age at the time of POI and LH, FSH as well as estradiol levels. Haplotype analysis also showed no significant difference between groups. LIMITATIONS, REASONS FOR CAUTION Study is limited to a Korean population. WIDER IMPLICATIONS OF THE FINDINGS Our findings suggest that genetic variants in the AMH signal transduction pathway may not influence the susceptibility of idiopathic POI. This is the first report on the association between the AMH and AMHR2 polymorphisms and idiopathic POI. STUDY FUNDING/COMPETING INTEREST(S) No conflict of interest exists. This study was supported by a grant of Seoul National University Hospital Research Fund (04-2011-0870). TRIAL REGISTRATION NUMBER: N/A. /////////////////////////

Species: human
Mutation name:
type: naturally occurring
fertility: subfertile
Comment: AMH mutations with reduced in vitro bioactivity are related to premature ovarian insufficiency. Alvaro Mercadal B et al. (2015) Could anti-Müllerian hormone (AMH) mutations be implicated in the development of idiopathic premature ovarian insufficiency (POI)? Three rare or unknown missense variants of the AMH gene resulting in were identified in a cohort of 55 POI patients; all three variants showed a drastically reduced in vitro bioactivity. Genetic factors are implicated in 5-15% of cases of POI. However, only a few genes have been shown to be involved in its development. AMH inhibits the recruitment of primordial follicles in the ovary and defective or absent AMH leads to premature depletion of the primordial follicle pool in AMH null mice. The whole coding sequence and the exon-intron junction of the AMH gene was sequenced in a cohort of 55 POI patients recruited over a period of 8 years. The studied variants were also sequenced in 197 ethnically matched controls. POI was defined as amenorrhea of more than 4 months with increased FSH before the age of 40. Patients with POI resulting from radio- or chemotherapy, surgery, chromosomal anomalies or FMR1 gene pre-mutation were excluded from the study. Recombinant human wild-type (wt) and mutated AMH proteins were produced in HEK293 T cells. KK-1 cells transfected with the AMH receptor type 2 (AMHR2) and a BMP responsive element coupled to a luciferase reporter vector were stimulated with different concentrations of wt AMH and the three tested variants. The whole coding sequence of the AMH gene could be performed and analyzed for 50 POI patients: 16 variants were found, including 6 missense variants from which 1 was unknown (R444H) and 2 were very rare (G264R and D288E). The variant D288E was also found in one of the patient's mother who also underwent POI at 32 years old. The stimulation of the AMHR2 assessed by the luciferase activity was drastically reduced for the three variants when compared with the wt AMH. The study is limited by a relatively small number of patients in the POI cohort. This is the first time that the bioactivity of AMH variants related to POI patients is tested in vitro. The functional study showed a drastic reduction of the protein activity for the three variants, supporting their contribution to the development of the ovarian insufficiency. The familial segregation further supports the implication of AMH in the development of POI. The study was performed thanks to funding from the 'Fondation Erasme'. No conflicts of interest are declared.//////////////////

Species: human
Mutation name:
type: naturally occurring
fertility: subfertile
Comment: Associations of Polymorphisms in Anti-Müllerian Hormone (AMH Ile49Ser) and its Type II Receptor (AMHRII -482 A>G) on Reproductive Outcomes and Polycystic Ovary Syndrome: a Systematic Review and Meta-Analysis. Pabalan N et al. (2016) Reported associations of reproductive outcomes (RO) and polycystic ovary syndrome (PCOS) with genotypes of the Ile49Ser and -482A>G polymorphisms in the Anti-Müllerian hormone (AMH) gene and its type II receptor (AMHRII), respectively, have conflicting results. PubMed, Google Scholar and Science Direct databases were searched for studies that investigated Ile49Ser and -482A>G in RO and PCOS. Using the metaanalytic approach, we estimated risk (odds ratio [OR] with 95% confidence intervals) using standard genetic models. All calculated summary effects were non-significant. Overall associations of Ile49Ser and -482A>G with RO were absent (OR 0.95-0.99, P = 0.76-0.96) but implied increased risk in PCOS (OR 1.07-1.17, P = 0.49-0.55). Where heterogeneity of the pooled ORs were present, its sources were explored using the Galbraith plot. Detection and omission of the outlying studies in both polymorphisms not only erased heterogeneity of the recalculated pooled outcomes but also changed direction of association, where null effects turned to increased risk (Ile49Ser in RO) and increased risk became reduced risk (-482A>G in PCOS). Implications of the Ile49Ser and -482A>G, effects pointed to protection for Caucasians (OR 0.64-0.89, P = 0.36-0.73) in RO and increased risk in PCOS (OR 1.19-1.45, P = 0.28-0.65). Asian effects in RO and PCOS were variable (OR 0.97-1.24, P = 0.58-0.91). In summary, we found no evidence of significant associations of Ile49Ser and -482A>G with RO and PCOS, although contrasting Ile49Ser effects were implied among Caucasians between RO (up to 0.36% reduced risk) and PCOS (up to 1.5-fold increased risk).//////////////////

Species: human
Mutation name:
type: naturally occurring
fertility: subfertile
Comment: Pathogenic Anti-Müllerian Hormone Variants in Polycystic Ovary Syndrome. Gorsic LK et al. (2017) Polycystic ovary syndrome (PCOS), a common endocrine condition, is the leading cause of anovulatory infertility. Given that common disease-susceptibility variants account for only a small percentage of the estimated PCOS heritability, we tested the hypothesis that rare variants contribute to this deficit in heritability. Unbiased whole genome sequencing (WGS) of 80 PCOS cases and 24 reproductively normal control women identified potentially deleterious variants in AMH, the gene encoding anti-Müllerian hormone (AMH). Targeted sequencing of AMH of 643 PCOS cases and 153 controls was used to replicate WGS findings. Dual luciferase reporter assays measured the impact of the variants on downstream AMH signaling. We found 24 rare (MAF<0.01) AMH variants in PCOS cases and controls; 18 variants were specific to PCOS cases. Seventeen of 18 (94%) PCOS-specific variants had significantly reduced AMH signaling, while 0 out 6 variants observed in controls showed significant defects in signaling. Thus, we identified rare AMH coding variants that reduced AMH mediated signaling in a subset of PCOS cases. This study is the first to identify rare genetic variants that are associated with a common PCOS phenotype. Our findings suggest a novel mechanism for in the pathogenesis of PCOS, decreased AMH signaling. AMH decreases androgen biosynthesis by inhibiting CYP17 activity; a potential mechanism of action for AMH variants in PCOS, therefore, is to increase androgen biosynthesis due to decreased AMH mediated inhibition of CYP17 activity.//////////////////

Species: mouse
Mutation name:
type: targeted overexpression
fertility: infertile - ovarian defect
Comment: Anti-Müllerian hormone overexpression restricts preantral ovarian follicle survival. Pankhurst MW et al. (2018) Anti-Müllerian hormone (AMH) is an ovarian regulator that affects folliculogenesis. AMH inhibits the developmental activation of the dormant primordial follicles and the oocyte within. In more mature follicles, AMH reduces granulosa cell sensitivity to follicle-stimulating hormone (FSH). We examined the effects of AMH overexpression on the stages of ovarian folliculogenesis, and the development of embryos, with a transgenic mouse that overexpresses human AMH in central nervous system neurons under the control of the mouse Thy1.2 promoter (Thy1.2-AMHTg mice). These mice are severely sub-fertile, despite relatively normal ovulation rates. The embryos of Thy1.2-AMHTg females exhibited delayed preimplantation development and extensive mid-gestation fetal resorption. Young Thy1.2-AMHTg mouse ovaries exhibited only a slight reduction in the rate of primordial follicle activation but large declines in the number of developing follicles surviving past the primary stage. It was expected that Thy1.2-AMHTg mice would retain more primordial follicles as they aged, but at 5 months-old their number was significantly reduced relative to wild type females. These data indicate that moderate elevations in AMH levels can severely restrict reproductive output and the number of developing follicles in the ovary. This evidence suggests that early antral follicles are a target for AMH signaling, which may regulate early follicle survival.//////////////////

Species: human
Mutation name:
type: naturally occurring
fertility: subfertile
Comment: Functional Genetic Variation in the Anti-Müllerian Hormone Pathway in Women with Polycystic Ovary Syndrome. Gorsic LK et al. (2019) PCOS is a highly heritable, common endocrine disorder characterized by hyperandrogenism, irregular menses and polycystic ovaries. PCOS is often accompanied by elevated levels of anti-Müllerian hormone (AMH). AMH inhibits follicle maturation. AMH also inhibits steroidogenesis through transcriptional repression of CYP17A1. We recently identified 16 rare PCOS-specific pathogenic variants in AMH. To test whether additional members of the AMH signaling pathway also contribute to the etiology of PCOS. Targeted re-sequencing of coding and regulatory regions of AMH and its specific type 2 receptor, AMHR2 was performed on 608 PCOS-affected women and 142 reproductively normal control women. Prediction tools of deleteriousness and in silico evidence of epigenetic modification were used to prioritize variants for functional evaluation. Dual luciferase reporter assays and splicing assays were used to measure the impact of genetic variants on function. We identified 20 additional variants in/near AMH and AMHR2 with significantly reduced signaling activity in in vitro assays. Collectively, from our previous study and reported herein, we have identified a total of 37 variants with impaired activity in/near AMH and AMHR2 in 41 women affected with PCOS, or 6.7% of our PCOS cohort. Furthermore, no functional variants were observed in the 142 phenotyped controls. The functional variants were significantly associated with PCOS in our cohort of 608 women with PCOS and 142 controls (p=2.3 x 10-5) and very strongly associated with PCOS relative to a larger non-Finnish European (gnomAD) population-based control cohort (p<1x10-9). AMH signaling cascade plays an important role in PCOS etiology.//////////////////

Species: mouse
Mutation name:
type: null mutation
fertility: fertile
Comment: Accelerated ovarian reserve depletion in female anti-Müllerian hormone knockout mice has no effect on lifetime fertility. Guo R et al. (2019) Anti-Müllerian hormone (AMH) inhibits the activation of primordial follicles in the ovary. This causes an increased rate of ovarian reserve depletion in Amh-/- mice. The depletion of the ovarian reserve is responsible for the onset of menopause but age-related infertility occurs in advance of ovarian reserve depletion. To determine whether accelerated loss of primordial follicles leads to earlier onset infertility, Amh-/- and Amh+/+ females were paired with Amh+/+ stud males and birth rates were recorded across the females' reproductive lifespan. The number of primordial follicles remaining in the ovaries of Amh-/- and Amh+/+ females were quantified in 2 cohorts at 11-12 and 12-13 months of age. As expected, the ovarian reserve in the Amh-/- females became depleted approximately 1 month earlier than Amh+/+ females. However, no difference was observed in the cumulative number of births over the lifespan, nor were there any differences in mean littersize at any age. It is possible that the reproductive lifespan of mice is too short for sufficient divergence of primordial follicles numbers to cause differences in Amh-/- and Amh+/+ female fertility. An alternative explanation contradicts current thinking; the function of AMH may be unrelated to the longevity of the reproductive lifespan in female mice.//////////////////

Genomic Region show genomic region
Phenotypes and GWAS show phenotypes and GWAS
Links
OMIM (Online Mendelian Inheritance in Man: an excellent source of general gene description and genetic information.)
OMIM \ Animal Model
KEGG Pathways
Recent Publications
None
Search for Antibody


created: Feb. 16, 2000, midnight by: genetron   email:
home page:
last update: Aug. 10, 2021, 8:51 a.m. by: hsueh    email:



Use the back button of your browser to return to the Gene List.

Click here to return to gene search form