1
|
Yildiz S, Moolhuijsen LME, Visser JA. The Role of Anti-Müllerian Hormone in Ovarian Function. Semin Reprod Med 2024; 42:15-24. [PMID: 38781987 DOI: 10.1055/s-0044-1786732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Anti-Müllerian hormone (AMH) is a member of the transforming growth factor β (TGFβ) superfamily, whose actions are restricted to the endocrine-reproductive system. Initially known for its role in male sex differentiation, AMH plays a role in the ovary, acting as a gatekeeper in folliculogenesis by regulating the rate of recruitment and growth of follicles. In the ovary, AMH is predominantly expressed by granulosa cells of preantral and antral follicles (i.e., post primordial follicle recruitment and prior to follicle-stimulating hormone (FSH) selection). AMH signals through a BMP-like signaling pathway in a manner distinct from other TGFβ family members. In this review, the latest insights in AMH processing, signaling, its regulation of spatial and temporal expression pattern, and functioning in folliculogenesis are summarized. In addition, effects of AMH variants on ovarian function are reviewed.
Collapse
Affiliation(s)
- Sena Yildiz
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Loes M E Moolhuijsen
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jenny A Visser
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
2
|
Hagen CP, Fischer MB, Mola G, Mikkelsen TB, Cleemann LH, Gravholt CH, Viuff MH, Juul A, Pedersen AT, Main KM. AMH and other markers of ovarian function in patients with Turner syndrome - a single center experience of transition from pediatric to gynecological follow up. Front Endocrinol (Lausanne) 2023; 14:1173600. [PMID: 37455919 PMCID: PMC10339808 DOI: 10.3389/fendo.2023.1173600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Turner syndrome (TS) is a chromosomal disorder that affects about 1 in 2500 female births and is characterized by the partial or complete absence of the second X chromosome. Depending on karyotype, TS is associated with primary ovarian insufficiency (POI). Approximately 50% of girls with a mosaic 45, X/46, XX karyotype may enter puberty spontaneously, but only 5-10% of women with TS achieve pregnancy without egg donation. In this review, we will evaluate the clinical use of markers of ovarian function in TS patients. Based on longitudinal studies of serum concentrations of reproductive hormones as well as ovarian morphology in healthy females and patients with TS, we will evaluate how they can be applied in a clinical setting. This is important when counseling patients and their families about future ovarian function essential for pubertal development and fertility. Furthermore, we will report on 20 years of experience of transition from pediatric to gynecological and adult endocrinological care in our center at Rigshospitalet, Copenhagen, Denmark.
Collapse
Affiliation(s)
- Casper P. Hagen
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), University of Copenhagen, Rigshospitalet, Denmark
| | - Margit Bistrup Fischer
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), University of Copenhagen, Rigshospitalet, Denmark
| | - Gylli Mola
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), University of Copenhagen, Rigshospitalet, Denmark
| | - Theis Bech Mikkelsen
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), University of Copenhagen, Rigshospitalet, Denmark
| | - Line Hartvig Cleemann
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), University of Copenhagen, Rigshospitalet, Denmark
| | - Claus Højbjerg Gravholt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Diabetes and Endocrine Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Mette H. Viuff
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), University of Copenhagen, Rigshospitalet, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anette Tønnes Pedersen
- Department of Gynecology, The Fertility Clinic, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| | - Katharina Maria Main
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), University of Copenhagen, Rigshospitalet, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Hagen CP, Fischer MB, Wohlfahrt-Veje C, Assens M, Busch AS, Pedersen AT, Juul A, Main KM. AMH concentrations in infancy and mid-childhood predict ovarian activity in adolescence: A long-term longitudinal study of healthy girls. EClinicalMedicine 2023; 55:101742. [PMID: 36386030 PMCID: PMC9661496 DOI: 10.1016/j.eclinm.2022.101742] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Anti-Müllerian hormone (AMH) is produced by granulosa cells in small growing ovarian follicles. In adult women, serum concentrations of AMH reflect the ovarian reserve of resting primordial follicles, and low AMH is associated with risk of early menopause. In contrast, patients with polycystic ovary syndrome (PCOS) have elevated AMH. The primary aim of this study was to evaluate the individual tracking of serum AMH concentrations, as well as whether AMH in early childhood reflects ovarian activity in adolescence. METHODS In this large longitudinal study of healthy girls were examined from infancy to adolescence (1997-2019) including physical examination, assessment of serum concentrations of reproductive hormones (in infancy, median age 0.3 yrs; mid-childhood, 7.2 yrs; puberty, 11.3 yrs; and adolescence, 15.9 yrs), transabdominal ultrasound (TAUS, puberty and adolescence) and magnetic resonance imaging (MRI, puberty) of the ovaries. FINDINGS Each girl maintained her relative AMH concentration (expressed as standard deviation (SD) scores) over time; mean variation of individual age adjusted AMH concentrations was 0.56 ± 0.31 SD.Serum concentrations of AMH in adolescence correlated with AMH in infancy and childhood; infancy: r = 0.347; mid-childhood: r = 0.637; puberty: r = 0.675, all p < 0.001.AMH correlated negatively with FSH concentrations in all age groups (infancy: r = -0.645, p < 0.001; mid-childhood: r = -0.222, p < 0.001; puberty: r = -0.354, p < 0.001; adolescence: n = 275, r = -0.175, p = 0.004).Serum AMH concentrations in mid-childhood correlated with the number of follicles in puberty (TAUS and MRI) as well as in adolescence (TAUS); e.g. total number of follicles: TAUS puberty (r = 0.607), MRI puberty (r = 0.379), TAUS adolescence (r = 0.414), all p < 0.001.AMH concentration in infancy as well as in mid-childhood predicted low AMH (<10 pmol/L) in adolescence; AMH infancy <7.5 pmol/L as predictor of low AMH in adolescence: sensitivity 0.71, specificity 0.70, AUC 0.759; AMH mid-childhood < 8.4 pmol/L as predictor of low AMH in adolescence: sensitivity 0.88, specificity 0.87, AUC 0.949.Girls with high serum AMH concentration in mid-childhood (AMH >30.0 pmol/L vs. other girls) had higher adolescent LH (median 4.53 vs. 3.29 U/L p = 0.041), LH/FSH ratio (1.00 vs 0.67, p = 0.019), testosterone (1.05 vs 0.81 nmol/L, p = 0.005), total number of follicles (23 vs. 19, p = 0.004), and higher prevalence of irregular cycles (10/15 = 67% vs. 28/113 = 25%, p = 0.002). INTERPRETATION The present findings suggest remarkably stable ovarian activity from small growing follicles in healthy girls, supporting AMH in early life as a useful clinical tool to predict future ovarian activity. FUNDING The work was supported by The Center on Endocrine Disruptors (CeHoS) under The Danish Environmental Protection Agency and The Ministry of Environment and Food (grant number: MST-621-00 065), the EU (QLK4-CT1999-01422; QLK4-2001-00269), the Novo Nordisk Foundation and The Danish Ministry of Science Technology and Innovation (2107-05-0006). A.S.B. is funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) - 464240267. KM receives honoraria from Novo Nordisk A/S for teaching at the Danish annual postgraduate course of pituitary diseases.
Collapse
Affiliation(s)
- Casper P. Hagen
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark
- Corresponding author. Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark.
| | - Margit Bistrup Fischer
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark
| | - Christine Wohlfahrt-Veje
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark
| | - Maria Assens
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark
| | - Alexander S. Busch
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark
| | - Anette Tønnes Pedersen
- Department of Gynaecology, The Fertility Clinic. Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Katharina M. Main
- Department of Growth and Reproduction, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Buratini J, Dellaqua TT, Dal Canto M, La Marca A, Carone D, Mignini Renzini M, Webb R. The putative roles of FSH and AMH in the regulation of oocyte developmental competence: from fertility prognosis to mechanisms underlying age-related subfertility. Hum Reprod Update 2021; 28:232-254. [PMID: 34969065 DOI: 10.1093/humupd/dmab044] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 11/18/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Fertility loss during female ageing is associated with increasing basal FSH and decreasing anti-Müllerian hormone (AMH) concentrations, together with compromised oocyte quality, presumably due to increased oxidative stress (OS) and DNA damage, as well as reduced metabolic and meiotic competences. Basal FSH and AMH circulatory concentrations have been broadly utilized as IVF success predictors, regardless of fluctuations in prognostic accuracy; basal FSH and AMH perform better in pre-advanced maternal age (AMA: >35 years) and AMA patients, respectively. The relationships between FSH and AMH intrafollicular levels and IVF outcomes suggest, nevertheless, that both hormones regulate oocyte competence, supporting the hypothesis that changes in FSH/AMH levels cause, at least in part, oocyte quality degradation during ageing. To understand the reasons behind the fluctuations in FSH and AMH prognostic accuracies and to clarify their participation in mechanisms determining oocyte competence and age-related subfertility, a deeper knowledge of the regulation of FSH and AMH intrafollicular signalling during the female reproductive lifespan, and of their effects on the cumulus-oocyte complex, is required. OBJECTIVE AND RATIONALE An extensive body of information on the regulation of FSH and AMH intrafollicular availability and signalling, as well as on the control of folliculogenesis and oocyte metabolism, has been accumulated. However, these datasets have been explored within the relatively narrow boundaries of their specific subjects. Given the aforementioned gaps in knowledge and their clinical relevance, herein we integrate clinical and basic data, within a wide biological perspective, aiming to shed light on (i) the reasons for the variability in the accuracy of serum FSH and AMH as fertility markers, and on (ii) the potential roles of these hormones in mechanisms regulating oocyte quality, particularly those associated with ageing. SEARCH METHODS The PubMed database encompassing the period between 1960 and 2021 was searched. Principal search terms were FSH, FSH receptor, AMH, oocyte, maternal age, cumulus, transzonal projections (TZPs), actin, OS, redox, reactive oxygen species, mitochondria, DNA damage, DNA repair, aneuploidy, spindle, meiosis, gene expression, transcription, translation, oocyte secreted factors (OSFs), cAMP, cyclic guanosine monophosphate, natriuretic peptide C, growth differentiation factor 9, bone morphogenetic protein 15 and fibroblast growth factor. OUTCOMES Our analysis suggests that variations in the accuracy of fertility prognosis reflect a modest association between circulatory AMH levels and oocyte quality as well as increasing basal FSH inter-cycle variability with age. In addition, the basic and clinical data articulated herein support the hypothesis that increased intrafollicular FSH levels, as maternal age advances, may override the physiological protective influences of AMH and OSFs against excessive FSH signalling in cumulus cells. This would result in the disruption of oocyte homeostasis via reduced TZP-mediated transfer of cumulus-derived molecules essential for meiotic competence, gene expression, redox activity and DNA repair. WIDER IMPLICATIONS In-depth data analysis, encompassing a wide biological perspective has revealed potential causative mechanisms of age-related subfertility triggered by alterations in FSH/AMH signalling during the female reproductive life. Insights from new mechanistic models arising from this analysis should contribute to advancing our comprehension of oocyte biology in humans and serve as a valuable reference for novel AMA subfertility treatments aimed at improving oocyte quality through the modulation of AMH/FSH action.
Collapse
Affiliation(s)
- Jose Buratini
- Biogenesi Reproductive Medicine Centre-Eugin Group, Istituti Clinici Zucchi, Monza, Italy.,Clinica Eugin Modena, Modena, Italy.,Department of Structural and Functional Biology, Sao Paulo State University, Botucatu, Brazil
| | - Thaisy Tino Dellaqua
- Department of Structural and Functional Biology, Sao Paulo State University, Botucatu, Brazil
| | - Mariabeatrice Dal Canto
- Biogenesi Reproductive Medicine Centre-Eugin Group, Istituti Clinici Zucchi, Monza, Italy.,Clinica Eugin Modena, Modena, Italy
| | - Antonio La Marca
- Clinica Eugin Modena, Modena, Italy.,Department of Medical and Surgical Sciences of the Mother, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Mario Mignini Renzini
- Biogenesi Reproductive Medicine Centre-Eugin Group, Istituti Clinici Zucchi, Monza, Italy.,Clinica Eugin Modena, Modena, Italy
| | - Robert Webb
- Division of Animal Sciences, School of Biosciences, University of Nottingham, Nottinghamshire, UK
| |
Collapse
|
5
|
di Clemente N, Racine C, Pierre A, Taieb J. Anti-Müllerian Hormone in Female Reproduction. Endocr Rev 2021; 42:753-782. [PMID: 33851994 DOI: 10.1210/endrev/bnab012] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/26/2022]
Abstract
Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance, was shown to be synthesized by the ovary in the 1980s. This article reviews the main findings of the past 20 years on the regulation of the expression of AMH and its specific receptor AMHR2 by granulosa cells, the mechanism of action of AMH, the different roles it plays in the reproductive organs, its clinical utility, and its involvement in the principal pathological conditions affecting women. The findings in respect of regulation tell us that AMH and AMHR2 expression is mainly regulated by bone morphogenetic proteins, gonadotropins, and estrogens. It has now been established that AMH regulates the different steps of folliculogenesis and that it has neuroendocrine effects. On the other hand, the importance of serum AMH as a reliable marker of ovarian reserve and as a useful tool in the prediction of the polycystic ovary syndrome (PCOS) and primary ovarian failure has also been acknowledged. Last but not least, a large body of evidence points to the involvement of AMH in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Nathalie di Clemente
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Chrystèle Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France.,Sorbonne Paris Cité, Paris-Diderot Université, Paris, France
| | - Alice Pierre
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| | - Joëlle Taieb
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| |
Collapse
|
6
|
Mamsen LS, Bøtkjær JA, Kristensen SG, Pors SE, Jeppesen JV, Kumar A, Kalra B, Ernst E, Andersen CY. High Variability of Molecular Isoforms of AMH in Follicular Fluid and Granulosa Cells From Human Small Antral Follicles. Front Endocrinol (Lausanne) 2021; 12:617523. [PMID: 33737910 PMCID: PMC7961079 DOI: 10.3389/fendo.2021.617523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/25/2021] [Indexed: 11/29/2022] Open
Abstract
Anti-Müllerian hormone (AMH) is a member of the TGF-β superfamily produced by follicular granulosa cells (GCs) in women from late gestation to the end of reproductive life. AMH is thought to inhibit aromatase (i.e., CYP19) expression and decrease the conversion of androgens to oestrogens, especially in small antral follicles before dominance is achieved. Thus, AMH acts as a gatekeeper of ovarian steroidogenesis. However, the exact function and processing of AMH has not been fully elucidated. The present study measured and determined AMH isoforms in human follicular fluid (FF) from small antral follicles and in human GCs using four ELISAs, western blot, and immunofluorescence analysis. We evaluated the presence of the following isoforms: full-length AMH precursor (proAMH), cleaved associated AMH (AMHN,C), N-terminal pro-region (AMHN), and active C-terminal (AMHC) AMH. A negative correlation between follicle diameter and the AMH forms was detected. Moreover, western blot analysis detected various AMH forms in both FFs and GCs, which did not match our consensus forms, suggesting an unknown proteolytic processing of AMH. The presence of these new molecular weight isoforms of AMH differs between individual follicles of identical size in the same woman. This study detected several AMH forms in FF and GCs obtained from human small antral follicles, which suggests that intrafollicular processing of AMH is complex and variable. Thus, it may be difficult to develop an antibody-based AMH assay that detects all AMH isoforms. Furthermore, the variability between follicles suggests that designing a recombinant AMH standard will be difficult.
Collapse
Affiliation(s)
- Linn Salto Mamsen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- *Correspondence: Linn Salto Mamsen,
| | - Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Janni Vikkelsø Jeppesen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ajay Kumar
- Ansh Labs, LLC, Webster, TX, United States
| | | | - Erik Ernst
- Department of Obstetrics and Gynaecology, Regional Hospital of Randers, Randers, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
7
|
von Wolff M, Andersen CY, Woodruff TK, Nawroth F. FertiPROTEKT, Oncofertility Consortium and the Danish Fertility-Preservation Networks - What Can We Learn From Their Experiences? CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2019; 13:1179558119845865. [PMID: 31068758 PMCID: PMC6495450 DOI: 10.1177/1179558119845865] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 02/27/2019] [Indexed: 01/18/2023]
Abstract
Fertility preservation is an increasingly important discipline. It requires close coordination between reproductive medicine specialists, reproductive biologists, and oncologists in various disciplines. In addition, it represents a particular health policy challenge, since fertility-protection measures are to be understood as a treatment for side effects of gonadotoxic treatments and would therefore normally have to be reimbursed by health insurance companies. Therefore, it is inevitable that fertility-preservation activities should organise themselves into a network structure both as a medical-logistic network and as a professional medical society. The necessary network structures can differ significantly at regional, national, and international level, as the size of the regions to be integrated and the local cultural and geographical conditions, as well as the political conditions are very different. To address these issues, the current review aims to point out the basic importance and the chances but also the difficulties of fertility-protection networks and give practical guidance for the development of such network structures. We will not only discuss network structures theoretically but also present them based on three established, different sized networks, such as the Danish Network (www.rigshospitalet.dk), representing a centralised network in a small country; the German-Austrian-Swiss network FertiPROTEKT® (www.fertiprotekt.com), representing a centralised as well as decentralised network in a large country; and the Oncofertility® Consortium (www.oncofertility.northwestern.edu), representing a decentralised, internationally oriented network, primarily serving the transfer of knowledge among its members.
Collapse
Affiliation(s)
- Michael von Wolff
- University Women’s Hospital, Division of Gynaecological Endocrinology and Reproductive Medicine, Inselspital, University Hospital, Bern, Switzerland
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Faculty of Health Science, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Teresa K Woodruff
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA
| | - Frank Nawroth
- Centre for Infertility, Prenatal Medicine, Endocrinology and Osteology, Amedes group, Hamburg, Germany
| |
Collapse
|
8
|
Wang JL, Liu B, Zhang C, Wang XM, Zhen D, Huang XM, Chen W, Gao JM. Effects of icariin on ovarian function in d-galactose-induced aging mice. Theriogenology 2019; 125:157-167. [PMID: 30447495 DOI: 10.1016/j.theriogenology.2018.10.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 10/13/2018] [Accepted: 10/29/2018] [Indexed: 01/15/2023]
Abstract
In this study, effects of icariin (Ica) on were examined in a mouse model of d-galactose (D-gal)-induced ovarian aging. Kunming white mice were divided into three groups: aging group induced with D-gal, experiment group treated with Ica at low (50 mg/kg), middle (100 mg/kg) and high (200 mg/kg) concentrations, and control group with no treatment. Ovarian histomorphology, serum FSH, LH and E2 levels, and reproductive function were compared among the groups. Ovarian expression of Amh, Bax and Bcl-2 was examined by qPCR and western blotting. Our results showed that diameters of secondary and tertiary follicles were significantly reduced in the aging group when compared with control group (P < 0.01), and were restored to normal in Ica 100 and Ica 200 treatment groups. The diameter of atretic follicles was significantly smaller in the aging group compared with control group and Ica 200 treatment group (P < 0.05). The proportion of secondary and atretic follicles was higher in the aging group compared with control group, Ica 100 and 200 treatment groups, whereas the proportion of tertiary and mature follicles was reduced in the aging group versus control, Ica 100 and 200 groups. The aging group lacked mature follicles, whereas Ica treatment induced mature follicle development. Primary and secondary follicles exhibited similar theca cell numbers and theca interna and externa cell layers in all groups examined, whereas theca interna and externa cell layers were decreased and increased, respectively, in tertiary follicles of aging group compared with control and I 200 groups. In the aging group, FSH and LH levels were significantly higher than those in control and Ica 200 groups (P < 0.05), and the E2 level was significantly reduced compared with control (P < 0.01), Ica 200 (P < 0.01), and Ica 100 (P < 0.05) groups. Serum hormone levels were equivalent in the control, Ica 100 and Ica 200 groups. The pregnancy rate was reduced in the aging group compared with other groups. The average litter size per birth, birth litter weight, and weaning weight of litters were all significantly lower in the aging group compared with control, Ica 100 and 200 groups (P < 0.05). The ovarian expression of AMH and Bcl-2 mRNA was significantly reduced in the aging group compared with those in control and Ica-treated groups (P < 0.01). In contrast, Bax expression was significantly higher in the aging group compared with all other groups (P < 0.01), and the Bcl-2/Bax ratio was markedly reduced in aging group compared with control, Ica 100 and 200 groups (P < 0.01), and Ica 50 group (P < 0.05). Ovarian expression of AMH protein was elevated in the Ica 100 group compared with the aging, control and Ica 50 groups (P < 0.01) and Ica 200 group (P < 0.05). Ovarian Bcl-2 protein levels and the Bcl-2/Bax ratio were significantly higher in the Ica 100 group than those in the Ica 50, 200 and aging groups (P < 0.05), and were similar or reduced (P < 0.05), respectively, compared to those in control group. Ovarian Bax expression was similar in each group. These findings suggest that Ica can improve ovarian follicular development, inhibit follicular atresia, decrease FSH and LH levels and increase E2, upregulate ovarian AMH expression and increase the Bcl-2/Bax ratio in aging mice. Therefore, Ica can partially restore ovarian function of aging mice and enhance their fertility. Optimal reproductive effects were obtained with the Ica 100 group.
Collapse
Affiliation(s)
- Jun-Li Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Bing Liu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Chao Zhang
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xin-Mei Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Di Zhen
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Xiao-Meng Huang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Wu Chen
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| | - Jian-Ming Gao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| |
Collapse
|
9
|
Riccetti L, Sperduti S, Lazzaretti C, Casarini L, Simoni M. The cAMP/PKA pathway: steroidogenesis of the antral follicular stage. ACTA ACUST UNITED AC 2018; 70:516-524. [PMID: 30160084 DOI: 10.23736/s0026-4784.18.04282-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pituitary gonadotropins, follicle-stimulating (FSH) and luteinizing hormone (LH) promote follicular recruitment and support antral follicle growth, maturation and selection, resulting in ovulation of the dominant follicle. FSH and LH biological functions are mediated by G protein-coupled receptors, FSHR and LHCGR, resulting in the activation of a number of signaling cascades, such as the cyclic AMP/protein kinase A (cAMP/PKA) pathway. Some in-vitro data are consistent with the dual, proliferative and pro-apoptotic role of cAMP, leaving unanswered questions on how cAMP/PKA signaling is linked to the follicle fate. Progression of the antral stage is characterized by the presence of dynamic serum gonadotropin and estrogen levels, accompanying proliferation and steroidogenesis of growing as well as apoptosis of atretic follicles. These events are parallel to changes of FSHR and LHCGR density at the cell surface occurring throughout the antral stage, reasonably modulating the cAMP/PKA activation pattern, cell metabolism and functions. Understanding whether gonadotropins and receptor expression levels impact on the steroidogenic pathway and play a role in determining the follicular fate, may put new light on molecular mechanisms regulating human reproduction. The aim of the present review is to update the role of major players modulating the cAMP/PKA pathway and regulating the balance between proliferative, differentiating and pro-apoptotic signals.
Collapse
Affiliation(s)
- Laura Riccetti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy -
| | - Samantha Sperduti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Clara Lazzaretti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Livio Casarini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy.,Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Modena, Italy
| |
Collapse
|
10
|
Xu J, Xu F, Lawson MS, Tkachenko OY, Ting AY, Kahl CA, Park BS, Stouffer RR, Bishop CV. Anti-Müllerian hormone is a survival factor and promotes the growth of rhesus macaque preantral follicles during matrix-free culture. Biol Reprod 2018; 98:197-207. [PMID: 29293939 PMCID: PMC6248587 DOI: 10.1093/biolre/iox181] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 12/22/2017] [Indexed: 11/14/2022] Open
Abstract
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) nontargeting 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 nontargeting control shRNA-vector and AMH shRNA-vector were injected into macaque ovaries. Ovaries were collected 9 days postinjection 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.
Collapse
Affiliation(s)
- Jing Xu
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Fuhua Xu
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Maralee S Lawson
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Olena Y Tkachenko
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Alison Y Ting
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Christoph A Kahl
- Molecular Virology Support Core, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Byung S Park
- Oregon Health & Science University-Portland State University School of Public Health, Oregon Health & Science University, Portland, Oregon, USA
| | - Richard R Stouffer
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Cecily V Bishop
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| |
Collapse
|
11
|
Kim YJ, Park KE, Kim YY, Kim H, Ku SY, Suh CS, Kim SH, Choi YM. Effects of Estradiol on the Paracrine Regulator Expression of In Vitro Maturated Murine Ovarian Follicles. Tissue Eng Regen Med 2017; 14:31-38. [PMID: 30603459 PMCID: PMC6171573 DOI: 10.1007/s13770-016-0006-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 12/18/2022] Open
Abstract
The preservation of female germ cells is important in the individuals with ovarian dysfunction and failure. For this purpose, ovarian follicle in vitro maturation (OFIVM) is an important technology for the retrieval of mature oocytes. In the in vivo follicular development, paracrine factors such as angiotensin (AT) and anti-Müllerian hormone (AMH) play important roles. We attempted to add estrogen during the OFIVM and to assess their expression on the follicular cells. The ovaries and pre-antral follicles were collected from 13-day C57BL/6 mice and cultured in vitro with estradiol (E2) treatment for up to two weeks. In the whole ovaries, the expression of AT II was decreased and the expression of AMH was similar between control and E2-treated ovaries after in vitro culture. Although there was no difference in the survival, ovulation, maturation and fertilization rates between control and E2-treated groups, the expression of AT II in the follicular cells was down-regulated after E2 treatment at mRNA level, and AMH showed similar expression. In conclusion, adding E2 in OFIVM may regulate paracrine factors and their receptors that are related to follicular development. Further investigations are necessary to elucidate the roles of various sex hormones in the regulation of AT and AMH expression during the OFIVM.
Collapse
Affiliation(s)
- Yong Jin Kim
- Department of Obstetrics and Gynecology, Korea University Guro Hospital, Seoul, Korea
| | - Kyung Eui Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Yoon Young Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Korea
| | - Chang Suk Suh
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Korea
| | - Seok Hyun Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Korea
| | - Young Min Choi
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
12
|
Kristensen SG, Mamsen LS, Jeppesen JV, Bøtkjær JA, Pors SE, Borgbo T, Ernst E, Macklon KT, Andersen CY. Hallmarks of Human Small Antral Follicle Development: Implications for Regulation of Ovarian Steroidogenesis and Selection of the Dominant Follicle. Front Endocrinol (Lausanne) 2017; 8:376. [PMID: 29375481 PMCID: PMC5770355 DOI: 10.3389/fendo.2017.00376] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 12/20/2017] [Indexed: 12/03/2022] Open
Abstract
Regulation of human ovarian steroidogenesis differs from other species and precise knowledge on how human small antral follicles (hSAF) develop and acquire competence for continued growth and steroid output is still incomplete. The present study has characterized almost 1,000 normal hSAF collected in connection with cryopreservation of ovarian tissue for fertility preservation. The antral follicles (ranging from 3 to 13 mm) were generally aspirated from one ovary surgically removed during the natural cycle, and the follicular fluid (FF) and the granulosa cells (GC) were isolated and snap-frozen. In FF, the following hormones were measured: inhibin-B, inhibin-A, AMH, follistatin, PAPP-A, estradiol, progesterone, testosterone, and androstenedione. In GC, mRNA gene expressions using q-PCR were measured for the following genes: FSHR, AMH, CYP19, and AR. All samples in which one of the abovementioned parameters was measured were included, but typically multiple parameters were measured. Highly significant differences in concentration and follicular content in relation to follicular diameter were found for all measured hormones despite massive variability in-between follicles for any given diameter. The results demonstrate that profound changes take place in the hormonal microenvironment around follicular diameters of 8-11 mm corresponding to when follicular selection occurs. At this point, inhibin-B and inhibin-A showed distinct peaks concomitant with a significant reduction in both AMH protein and mRNA expression. Concentrations of inhibins, androgens, FSHR, and AR were intimately associated, and it is suggested that inhibin-B in combination with PAPP-A and thereby IGF2 activity exerts important paracrine signaling at follicular selection. At the same time upregulation of estradiol synthesis and CYP19 mRNA expression increased steroid output profoundly. Furthermore, the highly significant association between FSHR and AR mRNA gene expression enforces important functions of androgens in follicular development. Collectively, these data reintroduce the understanding of the follicular phase as two parted in which regulation of steroidogenesis differs. The profound changes taking place around follicular selection highlight important paracrine actions of TGF-β family members and IGFs for securing dominance of the selected follicle.
Collapse
Affiliation(s)
- Stine G. Kristensen
- Faculty of Health and Medical Sciences, Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Linn S. Mamsen
- Faculty of Health and Medical Sciences, Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Janni V. Jeppesen
- Faculty of Health and Medical Sciences, Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jane Alrø Bøtkjær
- Faculty of Health and Medical Sciences, Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Susanne E. Pors
- Faculty of Health and Medical Sciences, Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Tanni Borgbo
- Faculty of Health and Medical Sciences, Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Erik Ernst
- The Fertility Clinic, Aarhus University Hospital, Aarhus, Denmark
| | - Kirsten T. Macklon
- The Fertility Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Claus Yding Andersen
- Faculty of Health and Medical Sciences, Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Claus Yding Andersen,
| |
Collapse
|
13
|
Expression of genes involved in BMP and estrogen signaling and AMPK production can be important factors affecting total number of antral follicles in ewes. Theriogenology 2016; 91:36-43. [PMID: 28215684 DOI: 10.1016/j.theriogenology.2016.12.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 12/16/2016] [Accepted: 12/21/2016] [Indexed: 11/24/2022]
Abstract
Follicular growth and ovulation of healthy oocytes is a complicated process which is regulated by several endocrine and paracrine factors as well as cross-talk between the oocyte and its surrounding somatic cells. This study compared the expression profile of some candidate genes involved in BMP signaling as well as estrogen and AMPK production in cumulus-oocyte complex (COC) of small and large antral follicles and their associated somatic cell layers in ovaries from ewes with high- and low-antral follicle count (AFC). Expression of GDF9 was increased by increasing the size of antral follicles, while BMP15 expression was decreased by follicular size. It should be noteworthy that transcription of both GDF9 and BMP15 was also detected in the adjacent cellular layers under the follicles. There was a very strong positive correlation between BMP15 and BMPR2 in ovary tissues. Expression of GDF9 was highly correlated with BMP15, BMPR1B, and BMPR2 in large antral follicles. Expression of BMP7 in small antral follicles and BMPR2 in ovary tissues was significantly increased in the high-AFC group. Expression of ESR1 and ESR2 involved in estrogen production as well as PRKAA1 which involved in AMPK production were significantly greater in large antral follicles of high-AFC. There was a very high correlation between Cyp19 and ESR1 in large antral follicles and ovary tissues. Expression of Cyp19 and PRKAA1 were positively correlated with GDF9, BMP15, and BMP7 in large follicles. In conclusion, this study suggests that apart from the BMP signaling, genes involved in AMPK and estrogen production can be pivotal players in ewe's follicular development process. In addition, a strong cross-talk can exist among AMPK, BMP signaling, and estrogen synthesis systems in ewe ovary.
Collapse
|
14
|
Dewailly D, Robin G, Peigne M, Decanter C, Pigny P, Catteau-Jonard S. Interactions between androgens, FSH, anti-Müllerian hormone and estradiol during folliculogenesis in the human normal and polycystic ovary. Hum Reprod Update 2016; 22:709-724. [PMID: 27566840 DOI: 10.1093/humupd/dmw027] [Citation(s) in RCA: 312] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 07/12/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Androgens, FSH, anti-Müllerian hormone (AMH) and estradiol (E2) are essential in human ovarian folliculogenesis. However, the interactions between these four players is not fully understood. OBJECTIVES AND RATIONALE The purpose of this review is to highlight the chronological sequence of the appearance and function of androgens, FSH, AMH and E2 and to discuss controversies in the relationship between FSH and AMH. A better understanding of this interaction could supplement our current knowledge about the pathophysiology of the polycystic ovary syndrome (PCOS). SEARCH METHODS A literature review was performed using the following search terms: androgens, FSH, FSH receptor, anti-Mullerian hormone, AMHRII, estradiol, follicle, ovary, PCOS, aromatase, granulosa cell, oocyte. The time period searched was 1980-2015 and the databases interrogated were PubMed and Web of Science. OUTCOMES During the pre-antral ('gonadotropin-independent') follicle growth, FSH is already active and promotes follicle growth in synergy with theca cell-derived androgens. Conversely, AMH is inhibitory by counteracting FSH. We challenge the hypothesis that AMH is regulated by androgens and propose rather an indirect effect through an androgen-dependent amplification of FSH action on granulosa cells (GCs) from small growing follicles. This hypothesis implies that FSH stimulates AMH expression. During the antral ('gonadotropin-dependent') follicle growth, E2 production results from FSH-dependent activation of aromatase. Conversely, AMH is inhibitory but the decline of its expression, amplified by E2, allows full expression of aromatase, characteristic of the large antral follicles. We propose a theoretical scheme made up of two triangles that follow each other chronologically. In PCOS, pre-antral follicle growth is excessive (triangle 1) because of intrinsic androgen excess that renders GCs hypersensitive to FSH, with consequently excessive AMH expression. Antral follicle growth and differentiation are disturbed (triangle 2) because of the abnormally persisting inhibition of FSH effects by AMH that blocks aromatase. Beside anovulation, this scenario may also serve to explain the higher receptiveness to gonadotropin therapy and the increased risk of ovarian hyperstimulation syndrome (OHSS) in patients with PCOS. WIDER IMPLICATIONS Within GCs, the balance between FSH and AMH effects is pivotal in the shift from androgen- to oestrogen-driven follicles. Our two triangles hypothesis, based on updated data from the literature, offers a pedagogic template for the understanding of folliculogenesis in the normal and polycystic ovary. It opens new avenues for the treatment of anovulation due to PCOS.
Collapse
Affiliation(s)
- Didier Dewailly
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France .,Faculté de Médecine, Université Lille Nord de France, 59000 Lille, France
| | - Geoffroy Robin
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France
| | - Maëliss Peigne
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France
| | - Christine Decanter
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France
| | - Pascal Pigny
- Faculté de Médecine, Université Lille Nord de France, 59000 Lille, France.,CHU Lille, Laboratoire de Biochimie & Hormonologie, Centre de Biologie Pathologie, F-59037 Lille, France
| | - Sophie Catteau-Jonard
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France.,Faculté de Médecine, Université Lille Nord de France, 59000 Lille, France
| |
Collapse
|
15
|
Xu J, Bishop CV, Lawson MS, Park BS, Xu F. Anti-Müllerian hormone promotes pre-antral follicle growth, but inhibits antral follicle maturation and dominant follicle selection in primates. Hum Reprod 2016; 31:1522-30. [PMID: 27165618 DOI: 10.1093/humrep/dew100] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/18/2016] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION What are the direct effects and physiological role of anti-Müllerian hormone (AMH) during primate follicular development and function at specific stages of folliculogenesis? SUMMARY ANSWER 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. WHAT IS KNOWN ALREADY 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. STUDY DESIGN, SIZE, DURATION 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. PARTICIPANTS/MATERIALS, SETTING, METHODS 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. MAIN RESULTS AND THE ROLE OF CHANCE 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. LIMITATIONS, REASONS FOR CAUTION 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. WIDER IMPLICATIONS OF THE FINDINGS 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. STUDY FUNDING/COMPETING INTERESTS NIH NICHD R01HD082208, NIH ORWH/NICHD K12HD043488 (BIRCWH), NIH OD P51OD011092 (ONPRC), Collins Medical Trust. There are no conflicts of interest. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- J Xu
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - C V Bishop
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - M S Lawson
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - B S Park
- OHSU-PSU School of Public Health, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - F Xu
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| |
Collapse
|
16
|
Qi X, Pang Y, Qiao J. The role of anti-Müllerian hormone in the pathogenesis and pathophysiological characteristics of polycystic ovary syndrome. Eur J Obstet Gynecol Reprod Biol 2016; 199:82-7. [DOI: 10.1016/j.ejogrb.2016.01.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/05/2016] [Accepted: 01/29/2016] [Indexed: 02/01/2023]
|
17
|
Liang A, Salzano A, D'Esposito M, Comin A, Montillo M, Yang L, Campanile G, Gasparrini B. Anti-Mullerian hormone (AMH) concentration in follicular fluid and mRNA expression of AMH receptor type II and LH receptor in granulosa cells as predictive markers of good buffalo (Bubalus bubalis) donors. Theriogenology 2016; 86:963-970. [PMID: 27087535 DOI: 10.1016/j.theriogenology.2016.03.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 02/29/2016] [Accepted: 03/12/2016] [Indexed: 10/22/2022]
Abstract
High individual variability in follicular recruitment and hence in the number of embryos produced is a major factor limiting the application of reproductive technologies in buffalo. Therefore, the identification of reliable markers to select embryo donors is critical to enroll buffaloes in embryo production programs. Better understanding of factors involved in follicular growth is also necessary to improve the response to superovulation in this species. The aim of this work was thus to determine the anti-Mullerian hormone (AMH) concentration in follicular fluid (FF) recovered from different size follicles and evaluate the mRNA expression profiles of development-related (AMHR2, CYP19A1, FSHR, and LHR) and apoptosis-related genes (TP53INP1 and CASP3) in the corresponding granulosa cells (GCs) in buffalo. Another objective was to evaluate whether the AMH concentration in FF and gene expression of GCs is associated with the antral follicular count. Ovaries were collected at the slaughterhouse, and all follicles were counted and classified as small (3-5 mm), medium (5-8 mm), and large (>8 mm). Follicular fluid was recovered for AMH determination, and the mRNA expression of AMHR2, FSHR, LHR, CYP19A1, TP53INP1, and CASP3 was analyzed in GCs. The AMH concentration in FF decreased (P < 0.01) at increasing follicular diameter. The mRNA expression of AMHR2 and FSHR was higher (P < 0.05) in small follicles, whereas that of LHR and CYP19A1 was higher (P < 0.05) in large follicles. The intrafollicular AMH concentration was positively correlated with the antral follicular count (r = 0.31; P < 0.05). Interestingly, good donors (≥12 follicles) had a higher (P < 0.05) concentration of AMH and AMHR2 levels in small follicles and higher (P < 0.05) LHR levels in large follicles than bad donors (<12 follicles). These results suggest a potential use of AMH to select buffalo donors to enroll in embryo production programs, laying the basis for further investigations.
Collapse
Affiliation(s)
- Aixin Liang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Angela Salzano
- Department of Veterinary Medicine and Animal Production, Federico II University, Naples, Italy
| | - Maurizio D'Esposito
- Institute of Genetics and Biophysics ABT, National Research Council, Naples, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Antonella Comin
- Department of Food Science, University of Udine, Udine, Italy
| | - Marta Montillo
- Department of Food Science, University of Udine, Udine, Italy
| | - Liguo Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Giuseppe Campanile
- Department of Veterinary Medicine and Animal Production, Federico II University, Naples, Italy
| | - Bianca Gasparrini
- Department of Veterinary Medicine and Animal Production, Federico II University, Naples, Italy.
| |
Collapse
|
18
|
Borgbo T, Macek M, Chrudimska J, Jeppesen JV, Hansen LL, Andersen CY. Size matters: Associations between the androgen receptor CAG repeat length and the intrafollicular hormone milieu. Mol Cell Endocrinol 2016; 419:12-7. [PMID: 26404660 DOI: 10.1016/j.mce.2015.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 09/16/2015] [Accepted: 09/16/2015] [Indexed: 10/23/2022]
Abstract
Granulosa cell (GC) expressed androgen receptors (AR) and intrafollicular androgens are central to fertility. The transactivating domain of the AR contains a polymorphic CAG repeat sequence, which is linked to the transcriptional activity of AR and may influence the GC function. This study aims to evaluate the effects of the AR CAG repeat length on the intrafollicular hormone profiles, and the gene expression profiles of GC from human small antral follicles. In total, 190 small antral follicles (3-11 mm in diameter) were collected from 58 women undergoing ovarian cryopreservation for fertility preservation. The biallelic mean of the CAG repeat lengths were calculated for each woman, and grouped in three groups: Long CAG repeats (23-26 mean CAG); medium CAG repeats (20.5-22.5 mean CAG) and short CAG repeats (17.5-20.0 mean CAG). The following parameters were measured: follicle diameter, intrafollicular levels of Anti-Müllerian Hormone (AMH), progesterone, oestradiol, testosterone and androstenedione, and GC gene expression levels of FSHR, LHR, AR, CYP19A1, and AMH. The long CAG repeat lengths were associated with significantly decreased testosterone levels, as compared to medium CAG repeats (P = 0.05) and short CAG repeats (P = 0.003). Furthermore, in follicles 3-6 mm in diameter, the long CAG repeats were associated with significantly increased LHR and CYP19A1 gene expression levels compared to short CAG repeat lengths (P = 0.004 and P = 0.04 respectively), and significantly increased LHR expression compared to medium CAG repeat lengths (P = 0.03). In conclusion, long CAG repeat lengths in the AR were associated to significant attenuated levels of androgens and an increased conversion of testosterone into oestradiol, in human small antral follicles.
Collapse
Affiliation(s)
- T Borgbo
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark; Laboratory of Reproductive Biology, Rigshospitalet, Copenhagen, Denmark.
| | - M Macek
- Department of Biology and Medical Genetics, 2nd Faculty of Medicine Charles University, University Hospital Motol, Prague, Czech Republic
| | - J Chrudimska
- Department of Biology and Medical Genetics, 2nd Faculty of Medicine Charles University, University Hospital Motol, Prague, Czech Republic
| | - J V Jeppesen
- Laboratory of Reproductive Biology, Rigshospitalet, Copenhagen, Denmark
| | - L L Hansen
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - C Yding Andersen
- Laboratory of Reproductive Biology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
19
|
Sacchi S, D'Ippolito G, Sena P, Marsella T, Tagliasacchi D, Maggi E, Argento C, Tirelli A, Giulini S, La Marca A. The anti-Müllerian hormone (AMH) acts as a gatekeeper of ovarian steroidogenesis inhibiting the granulosa cell response to both FSH and LH. J Assist Reprod Genet 2015; 33:95-100. [PMID: 26631403 DOI: 10.1007/s10815-015-0615-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/08/2015] [Indexed: 01/12/2023] Open
Abstract
PURPOSE 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. METHODS 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. RESULT 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. CONCLUSIONS 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.
Collapse
Affiliation(s)
- Sandro Sacchi
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Policlinico di Modena, 41100, Modena, Italy
| | - Giovanni D'Ippolito
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Policlinico di Modena, 41100, Modena, Italy
| | - Paola Sena
- Department of Biomedical, Metabolic and Neural Sciences, Section of Human Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Tiziana Marsella
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Policlinico di Modena, 41100, Modena, Italy
| | - Daniela Tagliasacchi
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Policlinico di Modena, 41100, Modena, Italy
| | - Elena Maggi
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Policlinico di Modena, 41100, Modena, Italy
| | - Cindy Argento
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Policlinico di Modena, 41100, Modena, Italy
| | - Alessandra Tirelli
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Policlinico di Modena, 41100, Modena, Italy
| | - Simone Giulini
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Policlinico di Modena, 41100, Modena, Italy
| | - Antonio La Marca
- Mother-Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Policlinico di Modena, 41100, Modena, Italy.
| |
Collapse
|
20
|
Claes A, Ball BA, Troedsson MHT, Curry TE, Squires EL, Scoggin KE. Molecular changes in the equine follicle in relation to variations in antral follicle count and anti-Müllerian hormone concentrations. Equine Vet J 2015; 48:741-748. [PMID: 26403213 DOI: 10.1111/evj.12514] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 09/19/2015] [Indexed: 11/29/2022]
Abstract
REASONS FOR PERFORMING STUDY The wide variation in circulating anti-Müllerian hormone (AMH) concentrations between mares is attributed to differences in antral follicle count (AFC) which may reflect follicular function. There are few data regarding variations in AFC and associated regulatory factors for AMH in the equine follicle during follicular development. OBJECTIVES To examine molecular and hormonal differences in the equine follicle in relation to variations in AFC and circulating AMH concentrations during follicular development and to identify genes co-expressed with AMH in the equine follicle. STUDY DESIGN Observational study. METHODS Plasma AMH concentrations and AFC were determined in 30 cyclic mares. Granulosa cells, theca cells and follicular fluid were recovered from growing (n = 17) or dominant follicles (n = 13). The expression of several genes, known to be involved in folliculogenesis and steroidogenesis, was examined using real-time reverse transcriptase polymerase chain reaction and immunohistochemistry. Intrafollicular oestradiol and AMH concentrations were determined by immunoassay. RESULTS Within growing follicles, the expression of AMH, AMHR2, ESR2 and INHA in granulosa cells was positively correlated with AFC and plasma AMH concentrations. In addition, the expression of ESR1 and FSHR was positively associated with plasma AMH concentrations. No significant associations were detected in dominant follicles. Furthermore, there was no association between AMH or oestradiol concentrations in follicular fluid and variations in AFC. Finally, the expression of AMH and genes co-expressed with AMH (AMHR2, ESR2 and FSHR) in granulosa cells as well as intrafollicular AMH concentrations decreased during follicular development while intrafollicular oestradiol concentrations increased and were inversely related to intrafollicular AMH concentrations. CONCLUSIONS This study indicates that variations in AFC and circulating AMH concentrations are associated with molecular changes in the growing equine follicle.
Collapse
Affiliation(s)
- A Claes
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, USA
| | - B A Ball
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, USA.
| | - M H T Troedsson
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, USA
| | - T E Curry
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, USA
| | - E L Squires
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, USA
| | - K E Scoggin
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, USA
| |
Collapse
|
21
|
Bøtkjær JA, Jeppesen JV, Wissing ML, Kløverpris S, Oxvig C, Mason JI, Borgbo T, Andersen CY. Pregnancy-associated plasma protein A in human ovarian follicles and its association with intrafollicular hormone levels. Fertil Steril 2015; 104:1294-301.e1. [DOI: 10.1016/j.fertnstert.2015.07.1152] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/22/2015] [Accepted: 07/22/2015] [Indexed: 10/23/2022]
|
22
|
Borgbo T, Jeppesen J, Lindgren I, Lundberg Giwercman Y, Hansen L, Yding Andersen C. Effect of the FSH receptor single nucleotide polymorphisms (FSHR 307/680) on the follicular fluid hormone profile and the granulosa cell gene expression in human small antral follicles. ACTA ACUST UNITED AC 2014; 21:255-61. [DOI: 10.1093/molehr/gau106] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
23
|
Ribeiro E, Bisinotto R, Lima F, Greco L, Morrison A, Kumar A, Thatcher W, Santos J. Plasma anti-Müllerian hormone in adult dairy cows and associations with fertility. J Dairy Sci 2014; 97:6888-900. [DOI: 10.3168/jds.2014-7908] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 06/29/2014] [Indexed: 11/19/2022]
|
24
|
Lee CT, Wang JY, Chou KY, Hsu MI. 1,25-Dihydroxyvitamin D3 increases testosterone-induced 17beta-estradiol secretion and reverses testosterone-reduced connexin 43 in rat granulosa cells. Reprod Biol Endocrinol 2014; 12:90. [PMID: 25239217 PMCID: PMC4180350 DOI: 10.1186/1477-7827-12-90] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 09/13/2014] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Aromatase converts testosterone into 17beta-estradiol in granulosa cells, and the converted 17beta-estradiol contributes to follicular maturation. Additionally, excessive testosterone inhibits aromatase activity, which can lead to concerns regarding polycystic ovary syndrome (PCOS). Generally, 1,25-dihydroxyvitamin D3 (1,25D3) supplements help to improve the symptoms of PCOS patients who exhibit low blood levels of 1,25D3. Therefore, this study investigated the interaction effects of 1,25D3 and testosterone on estrogenesis and intercellular connections in rat granulosa cells. METHODS Primary cultures of granulosa cells were treated with testosterone or testosterone plus 1,25D3, or pre-treated with a calcium channel blocker or calcium chelator. Cell lysates were subjected to western blot analysis to determine protein and phosphorylation levels, and 17beta-estradiol secretion was examined using a radioimmunoassay technique. Cell viability was evaluated by MTT reduction assay. Connexin 43 (Cx43) mRNA and protein expression levels were assessed by qRT-PCR, western blot, and immunocytochemistry. RESULTS Testosterone treatment (0.1 and 1 microg/mL) increased aromatase expression and 17beta-estradiol secretion, and the addition of 1,25D3 attenuated testosterone (1 microg/mL)-induced aromatase expression but improved testosterone-induced 17beta-estradiol secretion. Furthermore, testosterone-induced aromatase phosphotyrosine levels increased at 10 min, 30 min and 1 h, whereas 1,25D3 increased the longevity of the testosterone effect to 6 h and 24 h. Within 18-24 h of treatment, 1,25D3 markedly enhanced testosterone-induced 17beta-estradiol secretion. Additionally, pre-treatment with a calcium channel blocker nifedipine or an intracellular calcium chelator BAPTA-AM reduced 1,25D3 and testosterone-induced 17beta-estradiol secretion. Groups that underwent testosterone treatment exhibited significantly increased estradiol receptor beta expression levels, which were not affected by 1,25D3. Neither testosterone nor 1,25D3 altered 1,25D3 receptor expression. Finally, at high doses of testosterone, Cx43 protein expression was decreased in granulosa cells, and this effect was reversed by co-treatment with 1,25D3. CONCLUSIONS These data suggest that 1,25D3 potentially increases testosterone-induced 17beta-estradiol secretion by regulating aromatase phosphotyrosine levels, and calcium increase is involved in both 1,25D3 and testosterone-induced 17beta-estradiol secretion. 1,25D3 reverses the inhibitory effect of testosterone on Cx43 expression in granulosa cells.
Collapse
Affiliation(s)
- Ching-Tien Lee
- Department of Nursing, Hsin Sheng College of Medical Care and Management, Taoyuan, Taiwan
| | - Jiz-Yuh Wang
- Department of Neurology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kuang-Yi Chou
- General Education Center, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Ming-I Hsu
- Department of Obstetrics and Gynecology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
25
|
Weintraub A, Margalioth EJ, Chetrit AB, Gal M, Goldberg D, Alerhand S, Eldar-Geva T. The dynamics of serum anti-Mullerian-hormone levels during controlled ovarian hyperstimulation with GnRH-antagonist short protocol in polycystic ovary syndrome and low responders. Eur J Obstet Gynecol Reprod Biol 2014; 176:163-7. [DOI: 10.1016/j.ejogrb.2014.02.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 01/09/2014] [Accepted: 02/09/2014] [Indexed: 10/25/2022]
|
26
|
Can anti-Müllerian hormone concentrations be used to determine gonadotrophin dose and treatment protocol for ovarian stimulation? Reprod Biomed Online 2013; 26:431-9. [DOI: 10.1016/j.rbmo.2012.02.027] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 02/22/2012] [Accepted: 02/23/2012] [Indexed: 11/19/2022]
|
27
|
Jeppesen JV, Anderson RA, Kelsey TW, Christiansen SL, Kristensen SG, Jayaprakasan K, Raine-Fenning N, Campbell BK, Yding Andersen C. Which follicles make the most anti-Mullerian hormone in humans? Evidence for an abrupt decline in AMH production at the time of follicle selection. Mol Hum Reprod 2013; 19:519-27. [DOI: 10.1093/molehr/gat024] [Citation(s) in RCA: 244] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
28
|
Lan KC, Chang SY, Huang FJ, Lin HJ, Lin CY, Huang KE, Kang HY. Analysis of androgen receptor and anti-Müllerian hormone pathways in human granulosa cells under luteinizing hormone treatment. Reprod Biol Endocrinol 2013; 11:11. [PMID: 23433069 PMCID: PMC3599510 DOI: 10.1186/1477-7827-11-11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 02/13/2013] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The objective of this study was to determine the gene expression profiles of the androgen/androgen receptor (AR) and anti-Müllerian hormone (AMH)/ Sry-related high-mobility group box 9 (SOX9) pathways in granulosa-luteal cells from patients undergoing standard in vitro fertilization (IVF) with or without recombinant luteinizing hormone (rLH) therapy. METHODS Levels of reproductive hormones in the pre-ovulatory follicular fluid and the expression levels of LHR (luteinizing hormone receptor), AR, SOX9, AMH, AR-associated protein 54(ARA54)and ARA70 were determined in granulosa-luteal cells by real-time reverse-transcription PCR. The effects of androgen and rLH treatments on AR and AMH expression levels were also tested in vitro using HO23 cells. RESULTS We collected 35 an 70 granulosa cell samples from patients cycled with and without rLH supplementation, respectively. The clinical outcomes were similar in patients who received rLH therapy and those who did not, though the pre-ovulatory follicular fluid levels of androstenedione, testosterone, and estradiol were significantly higher and progesterone was lower in the rLH supplementation group. Moreover, granulosa-luteal cell mRNA levels of LHR, AR, AMH, and SOX9 were significantly higher in the rLH supplementation group relative to the group that did not receive rLH supplementation. In addition, we observed significant correlations between LHR and AR mRNA expression and among AR, AMH, and SOX9 mRNA expression in granulosa-luteal cells from patients undergoing standard IVF treatment. CONCLUSIONS Increased expression of LHR, AR, AMH, and SOX9 is characteristic of granulosa-luteal cells from IVF/ intracytoplasmic sperm injection (ICSI) patients receiving rLH supplementation.
Collapse
Affiliation(s)
- Kuo-Chung Lan
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Kaohsiung, Taiwan
- Center for Menopause and Reproductive Medicine Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | - Fu-Jen Huang
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsin-Jung Lin
- Center for Menopause and Reproductive Medicine Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ching-Yuang Lin
- College of Medicine, China Medical University, Clinical Immunology Center, China Medical University Hospital, Taichung, Taiwan
| | - Ko-En Huang
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Center for Menopause and Reproductive Medicine Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hong-Yo Kang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Kaohsiung, Taiwan
- Center for Menopause and Reproductive Medicine Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
29
|
Monniaux D, Drouilhet L, Rico C, Estienne A, Jarrier P, Touzé JL, Sapa J, Phocas F, Dupont J, Dalbiès-Tran R, Fabre S. Regulation of anti-Müllerian hormone production in domestic animals. Reprod Fertil Dev 2013; 25:1-16. [DOI: 10.1071/rd12270] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In mammals, anti-Müllerian hormone (AMH) expression is detected in the granulosa cells of all growing follicles and is highest in healthy small antral follicles, which contribute most significantly to AMH endocrine levels. AMH is a reliable endocrine marker of this population of gonadotrophin-responsive follicles in ruminants and, over the longer term, plasma AMH concentrations are characteristic of individual animals. In the cow, plasma AMH concentrations follow specific dynamic profiles throughout the prepubertal period, the oestrous cycle and the change from gestation to the post partum period, with the alterations most likely reflecting numerical changes in the population of high AMH-producing follicles. In granulosa cells, bone morphogenetic proteins (BMP) enhance AMH gene expression and AMH synthesis, with these effects antagonised by FSH. BMP could both support follicular growth and contribute significantly to the induction and/or maintenance of AMH expression in small growing follicles. AMH expression decreases sharply in large follicles when they become oestrogenic, suggesting a role for FSH and/or oestradiol in these changes, but the underlying mechanisms remain hypothetical. A better understanding of the factors and mechanisms regulating AMH production is needed to propose new strategies for managing the reserve of primordial and small growing follicles, as well as for improving embryo production.
Collapse
|
30
|
Grynberg M, Pierre A, Rey R, Leclerc A, Arouche N, Hesters L, Catteau-Jonard S, Frydman R, Picard JY, Fanchin R, Veitia R, di Clemente N, Taieb J. Differential regulation of ovarian anti-müllerian hormone (AMH) by estradiol through α- and β-estrogen receptors. J Clin Endocrinol Metab 2012; 97:E1649-57. [PMID: 22689696 DOI: 10.1210/jc.2011-3133] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
BACKGROUND Anti-müllerian hormone (AMH) is a member of the TGF-β family, which limits follicle maturation. Recently serum AMH has been recognized as a useful diagnostic and prognostic tool in human reproductive endocrinology. OBJECTIVE The aim of this study was to investigate the regulation of human ovarian AMH by estradiol and FSH. METHODS AMH mRNA were quantified by real time RT-PCR in human granulosa cells (GC). AMH transcription was studied in KK1 GC cotransfected with estrogen receptors (ER)-β or ERα, and normal human AMH promoter-luciferase construct (hAMH-luc) or mutated AMH promoter reporter constructs. Binding sites for estradiol (estrogen response element half-site) and steroidogenic factor 1 were disrupted by targeted mutagenesis. The level of ER in GC was determined by quantitative RT-PCR and Western blotting. RESULTS In KK1 cells, estradiol up-regulated and inhibited hAMH-luc in the presence of ERα and ERβ respectively. Disruption of estrogen response element half-site and/or steroidogenic factor 1 binding sites did not modify ERβ-mediated effect of estradiol on hAMH-luc, whereas it affected that conveyed by ERα. The FSH enhancement of hAMH-luc was abolished by estradiol in cells overexpressing ERβ. When both ER were transfected, estradiol inhibited hAMH-luc or had no effect. Estradiol repressed AMH mRNAs in human GC, which express a little more ERα than ERβ mRNA. CONCLUSIONS Our results show that AMH expression can be differentially regulated by estradiol depending on the ER and suggest that its decrease in GC of growing follicles, which mainly express ERβ, and during controlled ovarian hyperstimulation is due to the effect of estradiol.
Collapse
Affiliation(s)
- Michaël Grynberg
- Institut National de la Santé et de la Recherche Médicale Unité 782, 32 Rue des Carnets, Clamart F-92140, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Jeppesen JV, Nielsen ME, Kristensen SG, Yding Andersen C. Concentration of activin A and follistatin in follicular fluid from human small antral follicles associated to gene expression of the corresponding granulosa cells. Mol Cell Endocrinol 2012; 356:48-54. [PMID: 21846490 DOI: 10.1016/j.mce.2011.07.051] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 06/20/2011] [Accepted: 07/29/2011] [Indexed: 11/17/2022]
Abstract
The present study correlated concentrations of activin A and follistatin in follicular fluid (FF) from human small antral follicles to FF concentrations of AMH, inhibin B, progesterone, and oestradiol and to the mRNA expression of FSH-receptor (FSHR), LH-receptor (LHR), AMH-receptor2 (AMHR2), CYP19a, and androgen-receptor (AR) in the corresponding granulosa cells (GC). FF from 144 follicles (3-12 mm in diameter) was included whereas mRNA expression profiles were established in GC from 66 of the 144 follicles. Levels of follistatin remained constant in relation to follicular diameter, whereas activin A levels increased in follicles exceeding 10 mm in diameter. Levels of activin A and inhibin B showed a highly significant inverse association. Follistatin showed highly significant positive associations with AMH and inhibin B levels and with FSHR and AR gene expression in GC. This study revealed unexpected associations that probably reflect the complicated regulatory mechanisms governing human folliculogenesis.
Collapse
Affiliation(s)
- J V Jeppesen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction - Copenhagen University Hospital, Copenhagen University, Denmark.
| | | | | | | |
Collapse
|
32
|
Kedem-Dickman A, Maman E, Yung Y, Yerushalmi GM, Hemi R, Hanochi M, Dor J, Hourvitz A. Anti-Müllerian hormone is highly expressed and secreted from cumulus granulosa cells of stimulated preovulatory immature and atretic oocytes. Reprod Biomed Online 2012; 24:540-6. [PMID: 22421733 DOI: 10.1016/j.rbmo.2012.01.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 01/23/2012] [Accepted: 01/24/2012] [Indexed: 10/14/2022]
Abstract
This study investigated anti-Müllerian hormone (AMH) expression and secretion from cumulus granulosa cells (GC) and steroidogenesis in follicular fluids (FF) with relation to oocyte maturational stages and fertilization capacity in large preovulatory follicles. This prospective study included 53 ovulatory women undergoing intracytoplasmic sperm injection. FF and cumulus GC from 140 large preovulatory follicles were individually obtained during oocyte retrieval. Main outcome measures were oocyte maturation, fertilization and embryo quality. FF were assayed for AMH, progesterone, 17β-oestradiol and testosterone. Cumulus GC were assayed for AMH mRNA expression. AMH mRNA expression and secretion in cumulus GC in preovulatory follicles containing germinal-vesicle (GV) and metaphase-I (MI) oocytes were significantly higher than follicles containing MII oocytes (P<0.01 and P<0.0001, respectively). In addition, FF AMH concentrations from atretic oocytes were significantly higher than from MII oocytes. No correlation was found between AMH expression and secretion to fertilization or embryo quality. FF of MI and GV oocytes had higher concentrations of testosterone and lower progesterone/oestradiol ratios than MII oocytes, and FF of atretic oocytes contained higher testosterone concentrations than FF of MII oocytes. AMH is highly expressed in and secreted from cumulus GC of preovulatory follicles containing premature and atretic oocytes. Anti-Müllerian hormone (AMH) is produced in the female exclusively by granulosa cells. AMH has recently been shown to be one of the most important markers of ovarian reserve and it is highly associated with ovarian follicular development. This study investigates AMH expression and secretion from cumulus granulosa cells (GC) and steroidogenesis in the follicular fluids (FF) with relation to oocyte maturational stages, and fertilization capacity in large preovulatory follicles. We conducted a prospective study with 53 ovulatory women undergoing intracytoplasmic sperm injection. FF and cumulus GC from 140 large preovulatory follicles were individually obtained during oocyte retrieval. The main outcome measures were oocyte maturation, fertilization and embryo quality. FF were assayed for AMH, progesterone, 17β-oestradiol and testosterone. Cumulus GC were assayed for AMH mRNA expression. AMH mRNA expression in cumulus GC and AMH concentrations in FF of preovulatory follicles containing premature oocytes (germinal vesicle (GV) and metaphase I (MI)) were significantly higher than preovulatory follicles containing mature oocytes (MII oocytes). In addition, FF AMH concentrations of atretic oocytes were significantly higher than FF AMH of MII oocytes. No correlation was found between AMH expression and secretion for fertilization or embryo quality. FF of preovulatory MI and GV oocytes had higher levels of testosterone and lower progesterone/oestradiol ratios than MII oocytes, and FF of atretic oocytes contained higher testosterone levels than FF of MII oocytes. This study shows that AMH is highly expressed in and secreted from cumulus GC of preovulatory follicles containing premature and atretic oocytes.
Collapse
Affiliation(s)
- Alon Kedem-Dickman
- IVF Unit and Fertility Research Laboratory, Department of Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, Affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Kristensen SL, Ramlau-Hansen CH, Andersen CY, Ernst E, Olsen SF, Bonde JP, Vested A, Toft G. The association between circulating levels of antimüllerian hormone and follicle number, androgens, and menstrual cycle characteristics in young women. Fertil Steril 2012; 97:779-85. [PMID: 22244782 DOI: 10.1016/j.fertnstert.2011.12.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 12/07/2011] [Accepted: 12/14/2011] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To investigate the association between serum antimüllerian hormone (AMH) and other reproductive parameters in young women. DESIGN Cross-sectional study. SETTING University hospital. PATIENT(S) Population-based cohort of 256 women: 180 were users and 76 were nonusers of hormonal contraceptives. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Antral follicles, androgens, age at menarche, and duration and regularity of menstrual cycle. RESULT(S) AMH levels were lower among users of hormonal contraceptives compared to nonusers. Among nonusers, women with AMH levels in the upper tertile had 55% (95% confidence interval [CI]: 22%-99%) higher levels of total T and 8% (95% CI = 2%-15%) longer menstrual cycles than women with AMH levels in the lower tertile. An increase of 1 ng/mL in AMH was associated with 45% (95% CI = 6%-97%) higher prevalence of irregular menstrual cycles. These associations were not seen among users of hormonal contraceptives. A strong relationship between AMH and follicle number was found in both users and nonusers. CONCLUSION(S) AMH measurements were found to be applicable in evaluation of the reproductive function of young women. However, there may be differences in the way that serum AMH levels can be interpreted depending on whether the woman uses hormonal contraceptives or not.
Collapse
|
34
|
Baerwald AR, Adams GP, Pierson RA. Ovarian antral folliculogenesis during the human menstrual cycle: a review. Hum Reprod Update 2011; 18:73-91. [DOI: 10.1093/humupd/dmr039] [Citation(s) in RCA: 282] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
35
|
Pellatt L, Rice S, Dilaver N, Heshri A, Galea R, Brincat M, Brown K, Simpson ER, Mason HD. Anti-Müllerian hormone reduces follicle sensitivity to follicle-stimulating hormone in human granulosa cells. Fertil Steril 2011; 96:1246-51.e1. [DOI: 10.1016/j.fertnstert.2011.08.015] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 07/13/2011] [Accepted: 08/10/2011] [Indexed: 10/17/2022]
|
36
|
Rico C, Médigue C, Fabre S, Jarrier P, Bontoux M, Clément F, Monniaux D. Regulation of Anti-Müllerian Hormone Production in the Cow: A Multiscale Study at Endocrine, Ovarian, Follicular, and Granulosa Cell Levels1. Biol Reprod 2011; 84:560-71. [DOI: 10.1095/biolreprod.110.088187] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
37
|
Grøndahl ML, Nielsen ME, Dal Canto MB, Fadini R, Rasmussen IA, Westergaard LG, Kristensen SG, Yding Andersen C. Anti-Müllerian hormone remains highly expressed in human cumulus cells during the final stages of folliculogenesis. Reprod Biomed Online 2010; 22:389-98. [PMID: 21353640 DOI: 10.1016/j.rbmo.2010.12.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 12/09/2010] [Accepted: 12/14/2010] [Indexed: 11/17/2022]
Abstract
This study evaluated whether anti-Müllerian 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.
Collapse
Affiliation(s)
- M L Grøndahl
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, Copenhagen University, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Hillier SG, Smitz J, Eichenlaub-Ritter U. Folliculogenesis and oogenesis: from basic science to the clinic. Mol Hum Reprod 2010; 16:617-20. [DOI: 10.1093/molehr/gaq068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|