101
|
Froment P, Plotton I, Giulivi C, Fabre S, Khoueiry R, Mourad NI, Horman S, Ramé C, Rouillon C, Grandhaye J, Bigot Y, Chevaleyre C, Le Guevel R, Mallegol P, Andriantsitohaina R, Guerif F, Tamburini J, Viollet B, Foretz M, Dupont J. At the crossroads of fertility and metabolism: the importance of AMPK-dependent signaling in female infertility associated with hyperandrogenism. Hum Reprod 2022; 37:1207-1228. [PMID: 35459945 DOI: 10.1093/humrep/deac067] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 03/01/2022] [Indexed: 03/25/2024] Open
Abstract
STUDY QUESTION What biological processes are linked to the signaling of the energy sensor 5'-AMP-activated protein kinase (AMPK) in mouse and human granulosa cells (GCs)? SUMMARY ANSWER The lack of α1AMPK in GCs impacted cell cycle, adhesion, lipid metabolism and induced a hyperandrogenic response. WHAT IS KNOWN ALREADY AMPK is expressed in the ovarian follicle, and its activation by pharmacological medications, such as metformin, inhibits the production of steroids. Polycystic ovary syndrome (PCOS) is responsible for infertility in approximately 5-20% of women of childbearing age and possible treatments include reducing body weight, improving lifestyle and the administration of a combination of drugs to improve insulin resistance, such as metformin. STUDY DESIGN, SIZE, DURATION AMPK signaling was evaluated by analyzing differential gene expression in immortalized human granulosa cells (KGNs) with and without silencing α1AMPK using CRISPR/Cas9. In vivo studies included the use of a α1AMPK knock-out mouse model to evaluate the role of α1AMPK in folliculogenesis and fertility. Expression of α1AMPK was evaluated in primary human granulosa-luteal cells retrieved from women undergoing IVF with and without a lean PCOS phenotype (i.e. BMI: 18-25 kg/m2). PARTICIPANTS/MATERIALS, SETTING, METHODS α1AMPK was disrupted in KGN cells and a transgenic mouse model. Cell viability, proliferation and metabolism were evaluated. Androgen production was evaluated by analyzing protein levels of relevant enzymes in the steroid pathway by western blots, and steroid levels obtained from in vitro and in vivo models by mass spectrometry. Differential gene expression in human GC was obtained by RNA sequencing. Analysis of in vivo murine folliculogenesis was performed by histology and immunochemistry, including evaluation of the anti-Müllerian hormone (AMH) marker. The α1AMPK gene expression was evaluated by quantitative RT-PCR in primary GCs obtained from women with the lean PCOS phenotype (n = 8) and without PCOS (n = 9). MAIN RESULTS AND THE ROLE OF CHANCE Silencing of α1AMPK in KGN increased cell proliferation (P < 0.05 versus control, n = 4), promoted the use of fatty acids over glucose, and induced a hyperandrogenic response resulting from upregulation of two of the enzymes involved in steroid production, namely 3β-hydroxysteroid dehydrogenase (3βHSD) and P450 side-chain cleavage enzyme (P450scc) (P < 0.05, n = 3). Female mice deficient in α1AMPK had a 30% decrease in their ovulation rate (P < 0.05, n = 7) and litter size, a hyperandrogenic response (P < 0.05, n = 7) with higher levels of 3βHSD and p450scc levels in the ovaries, and an increase in the population of antral follicles (P < 0.01, n = 10) compared to controls. Primary GCs from lean women with PCOS had lower α1AMPK mRNA expression levels than the control group (P < 0.05, n = 8-9). LARGE SCALE DATA The FastQ files and metadata were submitted to the European Nucleotide Archive (ENA) at EMBL-EBI under accession number PRJEB46048. LIMITATIONS, REASONS FOR CAUTION The human KGN is a not fully differentiated, transformed cell line. As such, to confirm the role of AMPK in GC and the PCOS phenotype, this model was compared to two others: an α1AMPK transgenic mouse model and primary differentiated granulosa-lutein cells from non-obese women undergoing IVF (with and without PCOS). A clear limitation is the small number of patients with PCOS utilized in this study and that the collection of human GCs was performed after hormonal stimulation. WIDER IMPLICATIONS OF THE FINDINGS Our results reveal that AMPK is directly involved in steroid production in human GCs. In addition, AMPK signaling was associated with other processes frequently reported as dysfunctional in PCOS models, such as cell adhesion, lipid metabolism and inflammation. Silencing of α1AMPK in KGN promoted folliculogenesis, with increases in AMH. Evaluating the expression of the α1AMPK subunit could be considered as a marker of interest in infertility cases related to hormonal imbalances and metabolic disorders, including PCOS. STUDY FUNDING/COMPETING INTEREST(S) This study was financially supported by the Institut National de la Recherche Agronomique (INRA) and the national programme « FERTiNERGY » funded by the French National Research Agency (ANR). The authors report no intellectual or financial conflicts of interest related to this work. R.K. is identified as personnel of the International Agency for Research on Cancer/World Health Organization. R.K. alone is responsible for the views expressed in this article and she does not necessarily represent the decisions, policy or views of the International Agency for Research on Cancer/World Health Organization. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Pascal Froment
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Ingrid Plotton
- Molecular Endocrinology and Rare Diseases, University Hospital, Claude Bernard Lyon 1 University, Bron, France
| | - Cecilia Giulivi
- Department of Molecular Biosciences, University of California Davis, School of Veterinary Medicine, Davis, CA, USA
- The MIND Institute, University of California Davis Medical Center, Sacramento, CA, USA
| | - Stephane Fabre
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Rita Khoueiry
- Epigenetics Group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Nizar I Mourad
- Pôle de Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Brussels, Belgium
| | - Sandrine Horman
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Christelle Ramé
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | | | | - Yves Bigot
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | | - Remy Le Guevel
- Plate-forme ImPACcell, Université de Rennes 1, Rennes, France
| | - Patricia Mallegol
- SOPAM, U1063, INSERM, UNIV Angers, Angers, France
- Federative Structure of Research Cellular Interactions and Therapeutic Applications, SFR 4208 ICAT, Univ Angers, Angers, France
| | - Ramaroson Andriantsitohaina
- SOPAM, U1063, INSERM, UNIV Angers, Angers, France
- Federative Structure of Research Cellular Interactions and Therapeutic Applications, SFR 4208 ICAT, Univ Angers, Angers, France
| | | | - Jérôme Tamburini
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Benoit Viollet
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Marc Foretz
- Université de Paris, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Joelle Dupont
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| |
Collapse
|
102
|
McCartney CR, Campbell RE, Marshall JC, Moenter SM. The role of gonadotropin-releasing hormone neurons in polycystic ovary syndrome. J Neuroendocrinol 2022; 34:e13093. [PMID: 35083794 PMCID: PMC9232905 DOI: 10.1111/jne.13093] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/21/2021] [Accepted: 01/11/2022] [Indexed: 01/28/2023]
Abstract
Given the critical central role of gonadotropin-releasing hormone (GnRH) neurons in fertility, it is not surprising that the GnRH neural network is implicated in the pathology of polycystic ovary syndrome (PCOS), the most common cause of anovulatory infertility. Although many symptoms of PCOS relate most proximately to ovarian dysfunction, the central reproductive neuroendocrine system ultimately drives ovarian function through its regulation of anterior pituitary gonadotropin release. The typical cyclical changes in frequency of GnRH release are often absent in women with PCOS, resulting in a persistent high-frequency drive promoting gonadotropin changes (i.e., relatively high luteinizing hormone and relatively low follicle-stimulating hormone concentrations) that contribute to ovarian hyperandrogenemia and ovulatory dysfunction. However, the specific mechanisms underpinning GnRH neuron dysfunction in PCOS remain unclear. Here, we summarize several preclinical and clinical studies that explore the causes of aberrant GnRH secretion in PCOS and the role of disordered GnRH secretion in PCOS pathophysiology.
Collapse
Affiliation(s)
- Christopher R. McCartney
- Center for Research in Reproduction and Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of PhysiologySchool of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - John C. Marshall
- Center for Research in Reproduction and Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - Suzanne M. Moenter
- Departments of Molecular & Integrative PhysiologyInternal MedicineObstetrics and GynecologyUniversity of MichiganAnn ArborMIUSA
| |
Collapse
|
103
|
Prevot V, Sharif A. The polygamous GnRH neuron: Astrocytic and tanycytic communication with a neuroendocrine neuronal population. J Neuroendocrinol 2022; 34:e13104. [PMID: 35233849 DOI: 10.1111/jne.13104] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/12/2022] [Accepted: 01/30/2022] [Indexed: 11/28/2022]
Abstract
To ensure the survival of the species, hypothalamic neuroendocrine circuits controlling fertility, which converge onto neurons producing gonadotropin-releasing hormone (GnRH), must respond to fluctuating physiological conditions by undergoing rapid and reversible structural and functional changes. However, GnRH neurons do not act alone, but through reciprocal interactions with multiple hypothalamic cell populations, including several glial and endothelial cell types. For instance, it has long been known that in the hypothalamic median eminence, where GnRH axons terminate and release their neurohormone into the pituitary portal blood circulation, morphological plasticity displayed by distal processes of tanycytes modifies their relationship with adjacent neurons as well as the spatial properties of the neurohemal junction. These alterations not only regulate the capacity of GnRH neurons to release their neurohormone, but also the activation of discrete non-neuronal pathways that mediate feedback by peripheral hormones onto the hypothalamus. Additionally, a recent breakthrough has demonstrated that GnRH neurons themselves orchestrate the establishment of their neuroendocrine circuitry during postnatal development by recruiting an entourage of newborn astrocytes that escort them into adulthood and, via signalling through gliotransmitters such as prostaglandin E2, modulate their activity and GnRH release. Intriguingly, several environmental and behavioural toxins perturb these neuron-glia interactions and consequently, reproductive maturation and fertility. Deciphering the communication between GnRH neurons and other neural cell types constituting hypothalamic neuroendocrine circuits is thus critical both to understanding physiological processes such as puberty, oestrous cyclicity and aging, and to developing novel therapeutic strategies for dysfunctions of these processes, including the effects of endocrine disruptors.
Collapse
Affiliation(s)
- Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, Lille, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, Lille, France
| |
Collapse
|
104
|
Fan L, Tang N, Yao C, Wei X, Tang Y, Li J, Huang W. Association Between Fresh Embryo Transfers and Frozen-Thawed Embryo Transfers Regarding Live Birth Rates Among Women Undergoing Long Gonadotropin-Releasing Hormone Antagonist Protocols. Front Cell Dev Biol 2022; 10:884677. [PMID: 35573703 PMCID: PMC9096096 DOI: 10.3389/fcell.2022.884677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background: The availability and use of frozen-thawed embryos after controlled ovarian hyperstimulation for assisted reproduction have increased with improvements in vitrification techniques and the rise of gonadotropin-releasing hormone (GnRH) antagonist protocols. Although evidence has shown that frozen-thawed embryo transfers (FETs) result in higher live birth rates than fresh embryo transfers, it is uncertain whether this association exists in cycles employing the GnRH antagonist protocol. Objective: To test the hypothesis that FETs are more likely to result in a live birth than fresh embryo transfers in a GnRH antagonist protocol cycle and to investigate whether frozen blastocyst transfer increases live birth rates compared to fresh blastocyst transfer. Design: A retrospective historical cohort study was conducted using data collected from the Department of Reproductive Medicine of Liuzhou Maternity and Child Healthcare Hospital for 1,437 patients who underwent the GnRH antagonist protocol between 1 January 2015, and 31 December 2020. The primary outcome was the live birth rate, which was compared between fresh embryo transfer and FET, and the secondary outcomes were clinical pregnancy rate and miscarriage rate, which were compared between the two groups. Analyses were adjusted to account for the age of the patient, number of embryo transfers, day of embryo transfer, and type of infertility. Results: Fresh embryo transfers accounted for 1,026 (71.4%) of the 1,437 patients who underwent the GnRH antagonist protocol in our analysis, while FETs accounted for 411 (28.6%). Patients with fresh and frozen-thawed embryos had comparable median body mass index (body mass index; 22.3 [IQR, 24.6-20.0] vs. 22.0 [IQR, 24.5-19.9]). There was a significant difference in the median age of the fresh embryo transfer group (34.0 [IQR, 39.0-30.0]) and the Frozen-thawed embryo transfer group (32.0 [IQR, 37.0-29.0]). Blastocysts were transferred in 14.6% of the fresh embryo transfer cycles and 45.5% of the FET cycles, whereas they account for 10.4% and 13.0% of all patients, respectively. The mean number of embryos transferred was 2 (IQR, 2.0-1.0) for the fresh embryo transfer group and 1 (IQR, 2.0-1.0) for the FET group, with a significant difference in the mean number of embryos transferred. The live birth rate after fresh embryo transfer vs. FET was 28.7% vs. 34.5% (absolute difference, 5.9%; adjusted relative risk [aRR], 1.15 [95% CI, 0.88-1.51]). The clinical pregnancy rates were 39.9% vs. 46.0%, respectively (absolute difference, 6.1%; aRR, 1.10 [95% CI, 0.85-1.43]). The miscarriage rates were 22.5% vs. 23.8%, respectively (absolute difference, 1.3%; aRR, 1.13 [95% CI, 0.75-1.70]). Conclusion: In this retrospective study of women who underwent assisted reproduction using GnRH antagonists, FETs resulted in a higher live birth rates and clinical pregnancy rates than fresh embryo transfers, which parts of these differences were attributable to embryo stage. However, the interpretation of the findings is limited by the possibility of selection and confounding biases.
Collapse
Affiliation(s)
| | | | | | | | | | - Jingjing Li
- Department of Reproductive Medicine, Liuzhou Maternity and Child Healthcare Hospital Affiliated with Women and Children’s Hospital of Guangxi University of Science and Technology, Liuzhou, China
| | - Wenjie Huang
- Department of Reproductive Medicine, Liuzhou Maternity and Child Healthcare Hospital Affiliated with Women and Children’s Hospital of Guangxi University of Science and Technology, Liuzhou, China
| |
Collapse
|
105
|
Abruzzese GA, Silva AF, Velazquez ME, Ferrer MJ, Motta AB. Hyperandrogenism and Polycystic ovary syndrome: Effects in pregnancy and offspring development. WIREs Mech Dis 2022; 14:e1558. [PMID: 35475329 DOI: 10.1002/wsbm.1558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/18/2022] [Accepted: 04/01/2022] [Indexed: 11/10/2022]
Abstract
Polycystic ovary syndrome (PCOS) is one of the major endocrine disorders affecting women of reproductive age. Its etiology remains unclear. It is suggested that environmental factors, and particularly the intrauterine environment, play key roles in PCOS development. Besides the role of androgens in PCOS pathogenesis, exposure to endocrine disruptors, as is Bisphenol A, could also contribute to its development. Although PCOS is considered one of the leading causes of ovarian infertility, many PCOS patients can get pregnant. Some of them by natural conception and others by assisted reproductive technique treatments. As hyperandrogenism (one of PCOS main features) affects ovarian and uterine functions, PCOS women, despite reaching pregnancy, could present high-risk pregnancies, including implantation failure, an increased risk of gestational diabetes, preeclampsia, and preterm birth. Moreover, hyperandrogenism may also be maintained in these women during pregnancy. Therefore, as an altered uterine milieu, including hormonal imbalance, could affect the developing organisms, monitoring these patients throughout pregnancy and their offspring development is highly relevant. The present review focuses on the impact of androgenism and PCOS on fertility issues and pregnancy-related outcomes and offspring development. The evidence suggests that the increased risk of pregnancy complications and adverse offspring outcomes of PCOS women would be due to the factors involved in the syndrome pathogenesis and the related co-morbidities. A better understanding of the involved mechanisms is still needed and could contribute to a better management of these women and their offspring. This article is categorized under: Reproductive System Diseases > Molecular and Cellular Physiology Reproductive System Diseases > Environmental Factors.
Collapse
Affiliation(s)
- Giselle A Abruzzese
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Aimé F Silva
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariela E Velazquez
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria-José Ferrer
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alicia B Motta
- Laboratorio de Fisiopatología Ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFyBO), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
106
|
Coyle CS, Prescott M, Handelsman DJ, Walters KA, Campbell RE. Chronic androgen excess in female mice does not impact luteinizing hormone pulse frequency or putative GABAergic inputs to GnRH neurons. J Neuroendocrinol 2022; 34:e13110. [PMID: 35267218 PMCID: PMC9286661 DOI: 10.1111/jne.13110] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 11/28/2022]
Abstract
Polycystic ovary syndrome (PCOS) is associated with androgen excess and, frequently, hyperactive pulsatile luteinizing hormone (LH) secretion. Although the origins of PCOS are unclear, evidence from pre-clinical models implicates androgen signalling in the brain in the development of PCOS pathophysiology. Chronic exposure of female mice to dihydrotestosterone (DHT) from 3 weeks of age drives both reproductive and metabolic impairments that are ameliorated by selective androgen receptor (AR) loss from the brain. This suggests centrally driven mechanisms in hyperandrogen-mediated PCOS-like pathophysiology that remain to be defined. Acute prenatal DHT exposure can also model the hyperandrogenism of PCOS, and this is accompanied by increased LH pulse frequency and increased GABAergic innervation of gonadotrophin-releasing hormone (GnRH) neurons. We aimed to determine the impact of chronic exposure of female mice to DHT, which models the hyperandrogenism of PCOS, on pulsatile LH secretion and putative GABAergic input to GnRH neurons. To do this, GnRH-green fluorescent protein (GFP) female mice received either DHT or blank capsules for 90 days from postnatal day 21 (n = 6 or 7 per group). Serial tail-tip blood sampling was used to measure LH dynamics and perfusion-fixed brains were collected and immunolabelled for vesicular GABA transporter (VGAT) to assess putative GABAergic terminals associated with GFP-labelled GnRH neurons. As expected, chronic DHT resulted in acyclicity and significantly increased body weight. However, no differences in LH pulse frequency or the density of VGAT appositions to GnRH neurons were identified between ovary-intact DHT-treated females and controls. Chronic DHT exposure significantly increased the number of AR expressing cells in the hypothalamus, whereas oestrogen receptor α-expressing neuron number was unchanged. Therefore, although chronic DHT exposure from 3 weeks of age increases AR expressing neurons in the brain, the GnRH neuronal network changes and hyperactive LH secretion associated with prenatal androgen excess are not evident. These findings suggest that unique central mechanisms are involved in the reproductive impairments driven by exposure to androgen excess at different developmental stages.
Collapse
Affiliation(s)
- Chris S. Coyle
- Centre for Neuroendocrinology and Department of PhysiologySchool of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - Melanie Prescott
- Centre for Neuroendocrinology and Department of PhysiologySchool of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - David J Handelsman
- Andrology LaboratoryANZAC Research InstituteConcord HospitalUniversity of SydneySydneyNSWAustralia
| | - Kirsty A. Walters
- Fertility and Research CentreSchool of Women’s and Children’s HealthUniversity of New South WalesSydneyNSWAustralia
| | - Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of PhysiologySchool of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| |
Collapse
|
107
|
Silva MSB, Campbell RE. Polycystic Ovary Syndrome and the Neuroendocrine Consequences of Androgen Excess. Compr Physiol 2022; 12:3347-3369. [PMID: 35578968 DOI: 10.1002/cphy.c210025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a major endocrine disorder strongly associated with androgen excess and frequently leading to female infertility. Although classically considered an ovarian disease, altered neuroendocrine control of gonadotropin-releasing hormone (GnRH) neurons in the brain and abnormal gonadotropin secretion may underpin PCOS presentation. Defective regulation of GnRH pulse generation in PCOS promotes high luteinizing hormone (LH) pulsatile secretion, which in turn overstimulates ovarian androgen production. Early and emerging evidence from preclinical models suggests that maternal androgen excess programs abnormalities in developing neuroendocrine circuits that are associated with PCOS pathology, and that these abnormalities are sustained by postpubertal elevation of endogenous androgen levels. This article will discuss experimental evidence, from the clinic and in preclinical animal models, that has significantly contributed to our understanding of how androgen excess influences the assembly and maintenance of neuroendocrine impairments in the female brain. Abnormal central gamma-aminobutyric acid (GABA) signaling has been identified in both patients and preclinical models as a possible link between androgen excess and elevated GnRH/LH secretion. Enhanced GABAergic innervation and drive to GnRH neurons is suspected to contribute to the pathogenesis and early manifestation of neuroendocrine derangement in PCOS. Accordingly, this article also provides an overview of GABA regulation of GnRH neuron function from prenatal development to adulthood to discuss possible avenues for future discovery research and therapeutic interventions. © 2022 American Physiological Society. Compr Physiol 12:3347-3369, 2022.
Collapse
Affiliation(s)
- Mauro S B Silva
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
108
|
Man L, Lustgarten Guahmich N, Kallinos E, Caiazza B, Khan M, Liu ZY, Patel R, Torres C, Pepin D, Yang HS, Bodine R, Zaninovic N, Schattman G, Rosenwaks Z, James D. Chronic superphysiologic AMH promotes premature luteinization of antral follicles in human ovarian xenografts. SCIENCE ADVANCES 2022; 8:eabi7315. [PMID: 35263130 PMCID: PMC8906729 DOI: 10.1126/sciadv.abi7315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 01/14/2022] [Indexed: 05/19/2023]
Abstract
Anti-Müllerian hormone (AMH) is produced by growing ovarian follicles and provides a diagnostic measure of reproductive reserve in women; however, the impact of AMH on folliculogenesis is poorly understood. We cotransplanted human ovarian cortex with control or AMH-expressing endothelial cells in immunocompromised mice and recovered antral follicles for purification and downstream single-cell RNA sequencing of granulosa and theca/stroma cell fractions. A total of 38 antral follicles were observed (19 control and 19 AMH) at long-term intervals (>10 weeks). In the context of exogenous AMH, follicles exhibited a decreased ratio of primordial to growing follicles and antral follicles of increased diameter. Transcriptomic analysis and immunolabeling revealed a marked increase in factors typically noted at more advanced stages of follicle maturation, with granulosa and theca/stroma cells also displaying molecular hallmarks of luteinization. These results suggest that superphysiologic AMH alone may contribute to ovulatory dysfunction by accelerating maturation and/or luteinization of antral-stage follicles.
Collapse
Affiliation(s)
- Limor Man
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nicole Lustgarten Guahmich
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Eleni Kallinos
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Barbara Caiazza
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Monica Khan
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Zong-Ying Liu
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ritaben Patel
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Carmen Torres
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - David Pepin
- Department of Surgery, Harvard Medical School, Boston, MA 02215, USA
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02214, USA
| | - He S. Yang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Richard Bodine
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nikica Zaninovic
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
- Tri-Institutional Stem Cell Derivation Laboratory, Weill Cornell Medicine, New York, NY 10065, USA
| | - Glenn Schattman
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Zev Rosenwaks
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Daylon James
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
- Tri-Institutional Stem Cell Derivation Laboratory, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
109
|
Şahin B, Cura Şahin G, Tinelli A. Anti-Mullerian hormone levels in spontaneous pregnancies with hyperemesis gravidarum. J OBSTET GYNAECOL 2022; 42:2255-2259. [PMID: 35264073 DOI: 10.1080/01443615.2022.2036973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We investigated the serum anti-Mullerian hormone (AMH), oestrogen and progesterone levels in spontaneously conceiving pregnant women, diagnosed for hyperemesis gravidarum (HG), in the first trimester. Pregnant women admitted at Hospital between 5 and 12 gestational weeks and treated for HG were compared, in this prospective case-control study, to a control group. The serum AMH, oestradiol, and progesterone values of both groups were evaluated and compared, and potential correlations were calculated. The serum AMH values were significantly higher in the HG group than the control group (3.15 ± 1.51 ng/mL vs. 2.27 ± 1.20 ng/mL; p = 0.012). No significant difference in the two groups was about serum oestradiol and progesterone values (p = 0.264, p = 0.235). In the HG group, there was no significant correlation between AMH and oestradiol (p = 0.570). However, a significant positive correlation was determined between AMH and progesterone (p = 0.050). The area under the ROC curve (AUC) was 0.669 and the cut-off value was 2.65 ng/ml with sensitivity of 58.3% and specificity of 66.7% for AMH in pregnant women with HG. A correlation between AMH high values and unexplained HG was detected in spontaneous pregnancies in the first trimester. High AMH levels may play a role in HG etiopathogenesis, especially in pregnant women with low progesterone levels.Impact statementWhat is already known on this subject? Aetiological causes of Hyperemesis gravidarum (HG) include hormonal, immunological, genetic, and psychological issues. However, no conclusive etiopathogenesis to explain the pathophysiological mechanism of HG has been stated.What do the results of this study add? Authors focussed on investigation a possible correlation between pregnant hormones and the HG pathogenesis, prevalent in the first trimester, when hormones are produced by either the placenta or corpus luteum. High anti-Mullerian hormone (AMH) levels may play a role in HG etiopathogenesis, especially in pregnant women with low progesterone levels.What are the implications of these findings for clinical practice and/or further research? A serum AMH level of ≥2.65 mg/mL could be predictive for the development of HG in the first trimester of a spontaneous pregnancy. Basing on these findings, more selective pharmacological treatments could be administrated to these patients.
Collapse
Affiliation(s)
- Banuhan Şahin
- Gynecology and Obstetrics Department, Amasya University Sabuncuoglu Serefeddin Training and Research Hospital, Amasya, Turkey
| | - Gizem Cura Şahin
- Obstetrics and Gynecology Department, Tokat State Hospital, Tokat, Turkey
| | - Andrea Tinelli
- Obstetrics and Gynecology Department, Veris delli Ponti Hospital, Scorrano, Lecce, Italy.,Division of Experimental Endoscopic Surgery, Imaging, Technology and Minimally Invasive Therapy, Vito Fazzi Hospital, Lecce, Italy
| |
Collapse
|
110
|
Fornes R, Simin J, Nguyen MH, Cruz G, Crisosto N, van der Schaaf M, Engstrand L, Brusselaers N. Pregnancy, perinatal and childhood outcomes in women with and without polycystic ovary syndrome and metformin during pregnancy: a nationwide population-based study. Reprod Biol Endocrinol 2022; 20:30. [PMID: 35130922 PMCID: PMC8819934 DOI: 10.1186/s12958-022-00905-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/30/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Polycystic Ovary Syndrome (PCOS) is an endocrine disorder that affects women in reproductive age and represents an unfavourable risk factor for several pregnancy and perinatal outcomes. Despite, no guidelines or pharmaceutical strategies for treating PCOS during pregnancy are available. The aim of this study is to determine the association between polycystic ovary syndrome with or without metformin and the pregnancy, perinatal outcomes as well as the risk of obesity in children born to these mothers. METHODS In this nationwide population-based cohort study based in Swedish population, all singleton births (n = 1,016,805) from 686,847 women since 2006 up to 2016 were included. Multivariable logistic and Cox regression modelling with odds ratios (OR) and hazard ratios (HR) and 95% confidence intervals were used to study the association between the exposure of maternal PCOS, metformin during pregnancy (or the combination of both) and: 1) Pregnancy outcomes: preeclampsia, gestational diabetes, caesarean section, and acute caesarean section, 2) Perinatal outcomes: preterm birth, stillbirth, low birth weight, macrosomia, Apgar < 7 at 5 min, small for gestational age and large for gestational age, and 3) Childhood Obesity. RESULTS PCOS in women without metformin use during pregnancy was associated with higher risks of preeclampsia (OR = 1.09, 1.02-1.17), gestational diabetes (OR = 1.71, 1.53-1.91) and caesarean section (OR = 1.08, 1.04-1.12), preterm birth (OR = 1.30, 1.23-1.38), low birth weight (OR = 1.29, 1.20-1.38), low Apgar scores (OR = 1.17, 1.05-1.31) and large for gestational age (OR = 1.11, 1.03-1.20). Metformin use during pregnancy (in women without PCOS) was associated with a 29% lower risks of preeclampsia (OR = 0.71, 0.51-0.97), macrosomia and large for gestational age. Obesity was more common among children born to mothers with PCOS without metformin (HR = 1.61, 1.44-1.81); and those with metformin without PCOS (HR = 1.67, 1.05-2.65). PCOS with metformin was not associated with any adverse outcome. CONCLUSION PCOS was associated with increased risks of adverse pregnancy and perinatal outcomes and childhood obesity. Metformin appears to reduce these risks in mothers with polycystic ovary syndrome and their children; but may increase the risk of childhood-obesity in children form women without PCOS.
Collapse
Affiliation(s)
- Romina Fornes
- Centre for Translational Microbiome Research (CTMR), Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Biomedicum kvarter 8A, Tomtebodavägen 16, SE-171 65, Stockholm, Sweden
| | - Johanna Simin
- Centre for Translational Microbiome Research (CTMR), Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Biomedicum kvarter 8A, Tomtebodavägen 16, SE-171 65, Stockholm, Sweden
| | - Minh Hanh Nguyen
- I-BioStat, Data Science Institute, Hasselt University, Hasselt, Belgium
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Nicolás Crisosto
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Laboratory of Endocrinology and Metabolism, West Division, Faculty of Medicine, University of Chile, Santiago, Chile
- Endocrinology Unit, Clínica Las Condes, Las Condes, Chile
| | | | - Lars Engstrand
- Centre for Translational Microbiome Research (CTMR), Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Biomedicum kvarter 8A, Tomtebodavägen 16, SE-171 65, Stockholm, Sweden
| | - Nele Brusselaers
- Centre for Translational Microbiome Research (CTMR), Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Biomedicum kvarter 8A, Tomtebodavägen 16, SE-171 65, Stockholm, Sweden.
- Global Health Institute, Antwerp University, Antwerpen, Belgium.
| |
Collapse
|
111
|
Azumah R, Hummitzsch K, Hartanti MD, St. John JC, Anderson RA, Rodgers RJ. Analysis of Upstream Regulators, Networks, and Pathways Associated With the Expression Patterns of Polycystic Ovary Syndrome Candidate Genes During Fetal Ovary Development. Front Genet 2022; 12:762177. [PMID: 35197999 PMCID: PMC8860493 DOI: 10.3389/fgene.2021.762177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 11/25/2021] [Indexed: 12/31/2022] Open
Abstract
Polycystic Ovary Syndrome (PCOS) is a multifactorial syndrome with reproductive, endocrine, and metabolic symptoms, affecting about 10% women of reproductive age. Pathogenesis of the syndrome is poorly understood with genetic and fetal origins being the focus of the conundrum. Genetic predisposition of PCOS has been confirmed by candidate gene studies and Genome-Wide Association Studies (GWAS). Recently, the expression of PCOS candidate genes across gestation has been studied in human and bovine fetal ovaries. The current study sought to identify potential upstream regulators and mechanisms associated with PCOS candidate genes. Using RNA sequencing data of bovine fetal ovaries (62-276 days, n = 19), expression of PCOS candidate genes across gestation was analysed using Partek Flow. A supervised heatmap of the expression data of all 24,889 genes across gestation was generated. Most of the PCOS genes fell into one of four clusters according to their expression patterns. Some genes correlated negatively (early genes; C8H9orf3, TOX3, FBN3, GATA4, HMGA2, and DENND1A) and others positively (late genes; FDFT1, LHCGR, AMH, FSHR, ZBTB16, and PLGRKT) with gestational age. Pathways associated with PCOS candidate genes and genes co-expressed with them were determined using Ingenuity pathway analysis (IPA) software as well as DAVID Bioinformatics Resources for KEGG pathway analysis and Gene Ontology databases. Genes expressed in the early cluster were mainly involved in mitochondrial function and oxidative phosphorylation and their upstream regulators included PTEN, ESRRG/A and MYC. Genes in the late cluster were involved in stromal expansion, cholesterol biosynthesis and steroidogenesis and their upstream regulators included TGFB1/2/3, TNF, ERBB2/3, VEGF, INSIG1, POR, and IL25. These findings provide insight into ovarian development of relevance to the origins of PCOS, and suggest that multiple aetiological pathways might exist for the development of PCOS.
Collapse
Affiliation(s)
- Rafiatu Azumah
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Katja Hummitzsch
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Monica D. Hartanti
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Faculty of Medicine, Universitas Trisakti, Jakarta, Indonesia
| | - Justin C. St. John
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Raymond J. Rodgers
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
112
|
Incidence Of Congenital Uterine Abnormalities In Polycystic Ovarian Syndrome. Eur J Obstet Gynecol Reprod Biol 2022; 271:183-188. [DOI: 10.1016/j.ejogrb.2022.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/23/2021] [Accepted: 02/06/2022] [Indexed: 11/18/2022]
|
113
|
Dumesic DA, Padmanabhan V, Chazenbalk GD, Abbott DH. Polycystic ovary syndrome as a plausible evolutionary outcome of metabolic adaptation. Reprod Biol Endocrinol 2022; 20:12. [PMID: 35012577 PMCID: PMC8744313 DOI: 10.1186/s12958-021-00878-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/10/2021] [Indexed: 12/22/2022] Open
Abstract
As a common endocrinopathy of reproductive-aged women, polycystic ovary syndrome (PCOS) is characterized by hyperandrogenism, oligo-anovulation and polycystic ovarian morphology. It is linked with insulin resistance through preferential abdominal fat accumulation that is worsened by obesity. Over the past two millennia, menstrual irregularity, male-type habitus and sub-infertility have been described in women and confirm that these clinical features of PCOS were common in antiquity. Recent findings in normal-weight hyperandrogenic PCOS women show that exaggerated lipid accumulation by subcutaneous (SC) abdominal stem cells during development to adipocytes in vitro occurs in combination with reduced insulin sensitivity and preferential accumulation of highly-lipolytic intra-abdominal fat in vivo. This PCOS phenotype may be an evolutionary metabolic adaptation to balance energy storage with glucose availability and fatty acid oxidation for optimal energy use during reproduction. This review integrates fundamental endocrine-metabolic changes in healthy, normal-weight PCOS women with similar PCOS-like traits present in animal models in which tissue differentiation is completed during fetal life as in humans to support the evolutionary concept that PCOS has common ancestral and developmental origins.
Collapse
Affiliation(s)
- Daniel A. Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Room 22-178 CHS, Los Angeles, CA 90095 USA
| | | | - Gregorio D. Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Room 22-178 CHS, Los Angeles, CA 90095 USA
| | - David H. Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin and Wisconsin National Primate Research Center, 1223 Capitol Court, Madison, WI 53715 USA
| |
Collapse
|
114
|
Bar-Sadeh B, Amichai OE, Pnueli L, Begum K, Leeman G, Emes RD, Stöger R, Bentley GR, Melamed P. Epigenetic regulation of 5α reductase-1 underlies adaptive plasticity of reproductive function and pubertal timing. BMC Biol 2022; 20:11. [PMID: 34996447 PMCID: PMC8742331 DOI: 10.1186/s12915-021-01219-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 12/16/2021] [Indexed: 12/30/2022] Open
Abstract
Background Women facing increased energetic demands in childhood commonly have altered adult ovarian activity and shorter reproductive lifespan, possibly comprising a strategy to optimize reproductive success. Here, we sought to understand the mechanisms of early-life programming of reproductive function, by integrating analysis of reproductive tissues in an appropriate mouse model with methylation analysis of proxy tissue DNA in a well-characterized population of Bangladeshi migrants in the UK. Bangladeshi women whose childhood was in Bangladesh were found to have later pubertal onset and lower age-matched ovarian reserve than Bangladeshi women who grew-up in England. Subsequently, we aimed to explore the potential relevance to the altered reproductive phenotype of one of the genes that emerged from the screens. Results Of the genes associated with differential methylation in the Bangladeshi women whose childhood was in Bangladesh as compared to Bangladeshi women who grew up in the UK, 13 correlated with altered expression of the orthologous gene in the mouse model ovaries. These mice had delayed pubertal onset and a smaller ovarian reserve compared to controls. The most relevant of these genes for reproductive function appeared to be SRD5A1, which encodes the steroidogenic enzyme 5α reductase-1. SRD5A1 was more methylated at the same transcriptional enhancer in mice ovaries as in the women’s buccal DNA, and its expression was lower in the hypothalamus of the mice as well, suggesting a possible role in the central control of reproduction. The expression of Kiss1 and Gnrh was also lower in these mice compared to controls, and inhibition of 5α reductase-1 reduced Kiss1 and Gnrh mRNA levels and blocked GnRH release in GnRH neuronal cell cultures. Crucially, we show that inhibition of this enzyme in female mice in vivo delayed pubertal onset. Conclusions SRD5A1/5α reductase-1 responds epigenetically to the environment and its downregulation appears to alter the reproductive phenotype. These findings help to explain diversity in reproductive characteristics and how they are shaped by early-life environment and reveal novel pathways that might be targeted to mitigate health issues caused by life-history trade-offs. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01219-6.
Collapse
Affiliation(s)
- Ben Bar-Sadeh
- Faculty of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Or E Amichai
- Faculty of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Lilach Pnueli
- Faculty of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Khurshida Begum
- Department of Anthropology, Durham University, Durham, DH1 3LE, UK
| | - Gregory Leeman
- School of Biosciences, University of Nottingham, Nottingham, LE12 5RD, UK
| | - Richard D Emes
- School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham, LE12 5RD, UK
| | - Reinhard Stöger
- School of Biosciences, University of Nottingham, Nottingham, LE12 5RD, UK
| | | | - Philippa Melamed
- Faculty of Biology, Technion-Israel Institute of Technology, 32000, Haifa, Israel.
| |
Collapse
|
115
|
Antimullerian Hormone Levels and Association with Abortion and Preterm Delivery in Patients with Polycystic Ovary Syndrome Who Conceived with Assisted Reproductive Techniques. J Obstet Gynaecol India 2022; 72:295-298. [PMID: 35928099 PMCID: PMC9343493 DOI: 10.1007/s13224-021-01506-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 05/24/2021] [Indexed: 10/19/2022] Open
Abstract
Background Pregnant PCOS patients were found to suffer from many adverse outcomes of pregnancy. Prediction of the fate of pregnancy and labor in PCOS patients was highly needed. There were recently discovered roles of serum AMH in those patients who were seeking for pregnancy and who were conceiving with assisted reproductive techniques. Aim To analyze the predictive roles of measuring serum levels of AMH in patients with PCOS who became pregnant spontaneously or used assisted reproductive techniques regarding abortion and preterm delivery. Patients and Study Design A total of 100 females with PCOS were included in the study and they underwent assisted reproductive techniques were included in the study and underwent measurement of AMH levels regularly. Results We found that a total of 70 patients had a term delivery, and 30 patients had a preterm delivery. We found no statistically significant differences between both groups regarding their age or body mass index (BMI). We showed that serum AMH levels were higher in the group of PCOS patients who had preterm delivery than in the group of patients with term delivery (p < .0.001). Conclusions High serum AMH levels were found to be associated with higher risks of occurrence of preterm labor in patients with PCOS who underwent assisted reproductive techniques. Our results gave a clue to clinicians for better management of the pregnancy process in these patients.
Collapse
|
116
|
Wang K, Xu F, Maylie J, Xu J. Anti-Müllerian Hormone Regulation of Synaptic Transmission in the Hippocampus Requires MAPK Signaling and Kv4.2 Potassium Channel Activity. Front Neurosci 2022; 15:772251. [PMID: 34975379 PMCID: PMC8716599 DOI: 10.3389/fnins.2021.772251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/01/2021] [Indexed: 11/13/2022] Open
Abstract
Anti-Müllerian hormone (AMH) is a paracrine factor generated peripherally by the gonads to regulate gonadal function in adult mammals. We recently reported that AMH and AMH-specific receptor Anti-Müllerian hormone receptor 2 (AMHR2) are expressed in the hippocampus, and exogenous AMH protein rapidly increased synaptic transmission and long-term synaptic plasticity at the CA3-CA1 synapses. Here we examined the cell-specific expression of AMHR2 and the cellular mechanism of rapid boosting effect of AMH on synaptic transmission in mouse hippocampus. Immunofluorescence staining showed that AMHR2 was specifically expressed in the soma and dendrites of hippocampal pyramidal neurons, but not glial cells. Electrophysiological recordings on acute hippocampal slices showed that AMH did not affect AMPAR-mediated or N-Methyl-D-aspartic acid receptor (NMDAR)-mediated excitatory postsynaptic currents at the CA3-CA1 synapses. The small-conductance Ca2+-activated K+ channel (SK2) and A-type K+ channel (Kv4.2) contribute to shaping excitatory postsynaptic potentials (EPSPs) at the CA3-CA1 synapses. Bath application of apamin to block SK2 did not alter AMH effect on increasing EPSPs, whereas blocking Kv4.2 channel with 4-aminopyridine, or chelating internal Ca2+ with BAPTA occluded the action of AMH on boosting EPSPs. Kv4.2 activity is regulated by p38 mitogen-activated kinase (MAPK). Blocking p38 MAPK with SB203580 occluded the effect of AMH on increasing EPSPs. These results show that Kv4.2 channel contributes to the rapid action of AMH on boosting synaptic transmission in a Ca2+- and p38 MAPK-dependent manner. Our findings provide functional evidence that AMH enhances synaptic transmission through Kv4.2 channel in the hippocampus, suggesting a possible role of Kv4.2 channel in AMH-regulated neuronal process underlying learning and memory.
Collapse
Affiliation(s)
- Kang Wang
- Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Fuhua Xu
- Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - James Maylie
- Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Jing Xu
- Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR, United States.,Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| |
Collapse
|
117
|
Neven ACH, Mousa A, Boyle JA, Teede HJ. Endocrine and metabolic interactions in healthy pregnancies and hyperinsulinemic pregnancies affected by polycystic ovary syndrome, diabetes and obesity. Front Endocrinol (Lausanne) 2022; 13:993619. [PMID: 36733795 PMCID: PMC9886898 DOI: 10.3389/fendo.2022.993619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 12/06/2022] [Indexed: 01/18/2023] Open
Abstract
During pregnancy, the fetoplacental unit is key in the pronounced physiological endocrine changes which support pregnancy, fetal development and survival, birth and lactation. In healthy women, pregnancy is characterized by changes in insulin sensitivity and increased maternal androgen levels. These are accompanied by a suite of mechanisms that support fetal growth, maintain glucose homeostasis and protect both mother and fetus from adverse effects of pregnancy induced insulin and androgen excess. In pregnancies affected by endocrine, metabolic disorders such as polycystic ovary syndrome (PCOS), diabetes and obesity, there is an imbalance of beneficial and adverse impacts of pregnancy induced endocrine changes. These inter-related conditions are characterized by an interplay of hyperinsulinemia and hyperandrogenism which influence fetoplacental function and are associated with adverse pregnancy outcomes including hypertensive disorders of pregnancy, macrosomia, preterm delivery and caesarean section. However, the exact underlying mechanisms and relationships of the endocrine and metabolic milieu in these disorders and the impact they have on the prenatal endocrine environment and developing fetus remain poorly understood. Here we aim to review the complex endocrine and metabolic interactions in healthy women during normal pregnancies and those in pregnancies complicated by hyperinsulinemic disorders (PCOS, diabetes and obesity). We also explore the relationships between these endocrine and metabolic differences and the fetoplacental unit, pregnancy outcomes and the developing fetus.
Collapse
Affiliation(s)
- Adriana C. H. Neven
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Clayton, VIC, Australia
| | - Aya Mousa
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Clayton, VIC, Australia
| | - Jacqueline A. Boyle
- Monash Department of Obstetrics and Gynaecology, Monash Health, Clayton, VIC, Australia
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
- *Correspondence: Jacqueline A. Boyle,
| | - Helena J. Teede
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, Clayton, VIC, Australia
| |
Collapse
|
118
|
Xu H, Feng G, Alpadi K, Han Y, Yang R, Chen L, Li R, Qiao J. A Model for Predicting Polycystic Ovary Syndrome Using Serum AMH, Menstrual Cycle Length, Body Mass Index and Serum Androstenedione in Chinese Reproductive Aged Population: A Retrospective Cohort Study. Front Endocrinol (Lausanne) 2022; 13:821368. [PMID: 35370993 PMCID: PMC8970043 DOI: 10.3389/fendo.2022.821368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/28/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND A clinical diagnosis of polycystic ovary syndrome (PCOS) can be tedious with many different required tests and examinations. Furthermore, women with PCOS have increased risks for several metabolic complications, which need long-term health management. Therefore, we attempted to establish an easily applicable model to identify such women at an early stage. OBJECTIVE To develop an easy-to-use tool for screening PCOS based on medical records from a large assisted reproductive technology (ART) center in China. MATERIALS AND METHODS A retrospective observational cohort from Peking University Third Hospital was used in the study. Least Absolute Shrinkage and Selection Operator (LASSO) logistic regression with 10-fold cross-validation was applied to construct the model. The area under the receiver operating characteristic curve (AUC), sensitivity, and specificity values were used to evaluate and compare the models. DESIGN SETTING AND PARTICIPANTS This retrospective cohort study included 21,219 ovarian stimulation cycle records from January to December 2019 in Peking University Third Hospital. MAIN OUTCOMES AND MEASURES The main outcome was whether there was a clinical diagnosis of PCOS. The independent variables included were age, body mass index (BMI), upper limit of menstrual cycle length (UML), basal serum levels of anti-Müllerian hormone (AMH), testosterone androstenedione, antral follicle counts et al. RESULTS We have established a new mathematical model for diagnosing PCOS using serum AMH and androstenedione levels, UML, and BMI, with AUC values of 0.855 (0.838-0.870), 0.848 (0.791-0.891), 0.846 (0.812-0.875) in the training, validation, and testing sets, respectively. The contribution of each predictor to this model were: AMH 41.2%; UML 35.2%; BMI 4.3%; and androstenedione 3.7%. The top 10 groups of women most predicted to develop PCOS were demonstrated. An online tool (http://121.43.113.123:8888/) has been developed to assist Chinese ART clinics. CONCLUSIONS The models and online tool we established here might be helpful for screening and identifying women with undiagnosed PCOS in Asian populations and could assist in the long-term management of related metabolic disorders.
Collapse
Affiliation(s)
- Huiyu Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Guoshuang Feng
- Center for Clinical Epidemiology and Evidence-Based Medicine, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | | | - Yong Han
- Hangzhou Qingguo Medical Technology Co. Ltd., Hangzhou, China
| | - Rui Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Lixue Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- *Correspondence: Rong Li,
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| |
Collapse
|
119
|
Harada M. Pathophysiology of polycystic ovary syndrome revisited: Current understanding and perspectives regarding future research. Reprod Med Biol 2022; 21:e12487. [PMID: 36310656 PMCID: PMC9601867 DOI: 10.1002/rmb2.12487] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/01/2022] [Accepted: 09/15/2022] [Indexed: 11/05/2022] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among reproductive-age women and has lifelong effects on health. Methods In this review, I discuss the pathophysiology of PCOS. First, I summarize our current understanding of the etiology and pathology of PCOS, then, discuss details of two representative environmental factors involved in the pathogenesis of PCOS. Finally, I present perspectives regarding the directions of future research. Main findings The pathophysiology of PCOS is heterogeneous and shaped by the interaction of reproductive dysfunction and metabolic disorders. Hyperandrogenism and insulin resistance exacerbate one another during the development of PCOS, which is also affected by dysfunction of the hypothalamus-pituitary-ovarian axis. PCOS is a highly heritable disorder, and exposure to certain environmental factors causes individuals with predisposing genetic factors to develop PCOS. The environmental factors that drive the development of PCOS pathophysiology make a larger contribution than the genetic factors, and may include the intrauterine environment during the prenatal period, the follicular microenvironment, and lifestyle after birth. Conclusion On the basis of this current understanding, three areas are proposed to be subjects for future research, with the ultimate goals of developing therapeutic and preventive strategies and providing appropriate lifelong management, including preconception care.
Collapse
Affiliation(s)
- Miyuki Harada
- Department of Obstetrics and Gynecology, Faculty of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
120
|
Mendelian randomization analyses for PCOS: evidence, opportunities, and challenges. Trends Genet 2022; 38:468-482. [DOI: 10.1016/j.tig.2022.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 12/21/2022]
|
121
|
Shi N, Ma HB. Global trends in polycystic ovary syndrome research: A 10-year bibliometric analysis. Front Endocrinol (Lausanne) 2022; 13:1027945. [PMID: 36699019 PMCID: PMC9868474 DOI: 10.3389/fendo.2022.1027945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is one of the most common reproductive metabolic disorders in women, significantly affecting the biological functionalities of ovaries. This disease has garnered enormous interest from researchers. However, there is a lack of a comprehensive research concerning assessing the current status and future trends in PCOS field. This study uses bibliometric tools to comprehensively analyze the PCOS-related research progress based on the literature in the past decade. METHODS The reported PCOS literature in the past decade is downloaded from the Web of Science database. The bibliometric software is applied to analyze the co-authorship, co-citation, and co-occurrence status. RESULTS A total of 9936 publications imported into bibliometric tools for analysis show a sharp increase in the annual citations. The USA is dominant in terms of contribution in the field of PCOS, while China is making a significant contribution to the advancement of this field. Monash University is the most prolific institution with the highest H-index value. The contribution of University of Adelaide must be acknowledged. Legro RS and Teede HJ are the most active and influential authors in recent times, while Azziz R is the most contributed pioneer in this field. The Journal of Clinical Endocrinology & Metabolism is the most active journal with the highest number of publications and citations. The pathogenesis of PCOS had been a long-term forefront of research. In recent years, the health management in PCOS prevention and long-term complications was attracting more and more attention. The keywords like "gut microbiota", "microRNAs", "apoptosis", "Myo-inositol", "TNF-alpha", "androgen receptor", and "Vitamin D-deficient" are considered the latest research topics. CONCLUSION The study comprehensively analyzes the current status and global trends in the PCOS field, providing a significant reference for researchers to explore this field effectively.
Collapse
Affiliation(s)
- Na Shi
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-bo Ma
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Hong-bo Ma,
| |
Collapse
|
122
|
Jamieson BB, Moore AM, Lohr DB, Thomas SX, Coolen LM, Lehman MN, Campbell RE, Piet R. Prenatal androgen treatment impairs the suprachiasmatic nucleus arginine-vasopressin to kisspeptin neuron circuit in female mice. Front Endocrinol (Lausanne) 2022; 13:951344. [PMID: 35992143 PMCID: PMC9388912 DOI: 10.3389/fendo.2022.951344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/06/2022] [Indexed: 01/13/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is associated with elevated androgen and luteinizing hormone (LH) secretion and with oligo/anovulation. Evidence indicates that elevated androgens impair sex steroid hormone feedback regulation of pulsatile LH secretion. Hyperandrogenemia in PCOS may also disrupt the preovulatory LH surge. The mechanisms through which this might occur, however, are not fully understood. Kisspeptin (KISS1) neurons of the rostral periventricular area of the third ventricle (RP3V) convey hormonal cues to gonadotropin-releasing hormone (GnRH) neurons. In rodents, the preovulatory surge is triggered by these hormonal cues and coincident timing signals from the central circadian clock in the suprachiasmatic nucleus (SCN). Timing signals are relayed to GnRH neurons, in part, via projections from SCN arginine-vasopressin (AVP) neurons to RP3VKISS1 neurons. Because rodent SCN cells express androgen receptors (AR), we hypothesized that these circuits are impaired by elevated androgens in a mouse model of PCOS. In prenatally androgen-treated (PNA) female mice, SCN Ar expression was significantly increased compared to that found in prenatally vehicle-treated mice. A similar trend was seen in the number of Avp-positive SCN cells expressing Ar. In the RP3V, the number of kisspeptin neurons was preserved. Anterograde tract-tracing, however, revealed reduced SCNAVP neuron projections to the RP3V and a significantly lower proportion of RP3VKISS1 neurons with close appositions from SCNAVP fibers. Functional assessments showed, on the other hand, that RP3VKISS1 neuron responses to AVP were maintained in PNA mice. These findings indicate that PNA changes some of the neural circuits that regulate the preovulatory surge. These impairments might contribute to ovulatory dysfunction in PNA mice modeling PCOS.
Collapse
Affiliation(s)
- Bradley B. Jamieson
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Aleisha M. Moore
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Dayanara B. Lohr
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Simone X. Thomas
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Lique M. Coolen
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Michael N. Lehman
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Richard Piet
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States
- *Correspondence: Richard Piet,
| |
Collapse
|
123
|
Azumah R, Liu M, Hummitzsch K, Bastian NA, Hartanti MD, Irving-Rodgers HF, Anderson RA, Rodgers RJ. OUP accepted manuscript. Hum Reprod 2022; 37:1244-1254. [PMID: 35413103 PMCID: PMC9156849 DOI: 10.1093/humrep/deac049] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/04/2022] [Indexed: 11/25/2022] Open
Abstract
STUDY QUESTION Could changes in transforming growth factor β (TGFβ) signalling during foetal ovary development alter the expression of polycystic ovary syndrome (PCOS) candidate genes leading to a predisposition to PCOS? SUMMARY ANSWER TGFβ signalling molecules are dynamically expressed during foetal ovary development and TGFβ1 inhibits expression of the androgen receptor (AR) and 7 (INSR, C8H9orf3, RAD50, ERBB3, NEIL2, IRF1 and ZBTB16) of the 25 PCOS candidate genes in foetal ovarian fibroblasts in vitro, whilst increasing expression of the AR cofactor TGFβ-induced transcript 1 (TGFB1I1 or Hic5). WHAT IS KNOWN ALREADY The ovarian stroma arises from the mesonephros during foetal ovary development. Changes in the morphology of the ovarian stroma are cardinal features of PCOS. The ovary is more fibrous and has more tunica and cortical and subcortical stroma. It is not known why this is and when this arises. PCOS has a foetal origin and perhaps ovarian stroma development is altered during foetal life to determine the formation of a polycystic ovary later in life. PCOS also has a genetic origin with 19 loci containing 25 PCOS candidate genes. In many adult tissues, TGFβ is known to stimulate fibroblast replication and collagen deposition in stroma, though it has the opposite effect in the non-scaring foetal tissues. Our previous studies showed that TGFβ signalling molecules [TGFβs and their receptors, latent TGFβ binding proteins (LTBPs) and fibrillins, which are extracellular matrix proteins that bind LTBPs] are expressed in foetal ovaries. Also, we previously showed that TGFβ1 inhibited expression of AR and 3 PCOS candidate genes (INSR, C8H9orf3 and RAD50) and stimulated expression of TGFB1I1 in cultured foetal ovarian fibroblasts. STUDY DESIGN, SIZE, DURATION We used Bos taurus for this study as we can ethically collect foetal ovaries from across the full 9-month gestational period. Foetal ovaries (62–276 days, n = 19) from across gestation were collected from pregnant B. taurus cows for RNA-sequencing (RNA-seq) analyses. Foetal ovaries from B. taurus cows were collected (160–198 days, n = 6) for culture of ovarian fibroblasts. PARTICIPANTS/MATERIALS, SETTING, METHODS RNA-seq transcriptome profiling was performed on foetal ovaries and the data on genes involved in TGFβ signalling were extracted. Cells were dispersed from foetal ovaries and fibroblasts cultured and treated with TGFβ1. The effects of TGFβ regulation on the remaining eight PCOS candidate genes not previously studied (ERBB3, MAPRE1, FDFT1, NEIL2, ARL14EP, PLGRKT, IRF1 and ZBTB16) were examined. MAIN RESULTS AND THE ROLE OF CHANCE Many TGFβ signalling molecules are expressed in the foetal ovary, and for most, their expression levels increased accross gestation (LTBP1/2/3/4, FBN1, TGFB2/3, TGFBR2/3 and TGFB1I1), while a few decreased (FBN3, TGFBR3L, TGFBI and TGFB1) and others remained relatively constant (TGFBRAP1, TGFBR1 and FBN2). TGFβ1 significantly decreased expression of PCOS candidate genes ERBB3, NEIL2, IRF1 and ZBTB16 in cultured foetal ovarian fibroblasts. LARGE SCALE DATA The FASTQ files, normalized data and experimental information have been deposited in the Gene Expression Omnibus (GEO) accessible by accession number GSE178450. LIMITATIONS, REASONS FOR CAUTION Regulation of PCOS candidate genes by TGFβ was carried out in vitro and further studies in vivo are required. This study was carried out in bovine where foetal ovaries from across all of the 9-month gestational period were available, unlike in the human where it is not ethically possible to obtain ovaries from the second half of gestation. WIDER IMPLICATIONS OF THE FINDINGS From our current and previous results we speculate that inhibition of TGFβ signalling in the foetal ovary is likely to (i) increase androgen sensitivity by enhancing expression of AR, (ii) increase stromal activity by stimulating expression of COL1A1 and COL3A1 and (iii) increase the expression of 7 of the 25 PCOS candidate genes. Thus inhibition of TGFβ signalling could be part of the aetiology of PCOS or at least the aetiology of polycystic ovaries. STUDY FUNDING/COMPETING INTEREST(S) Funding was received from Adelaide University China Fee Scholarship (M.L.), Australian Research Training Program (R.A.) and the Faculty of Health and Medical Science Divisional Scholarship (R.A.), Adelaide Graduate Research Scholarships (R.A. and N.A.B.), Australia Awards Scholarship (M.D.H.), Robinson Research Institute Career Development Fellowship (K.H.) and Building On Ideas Grant (K.H.), National Health and Medical Research Council of Australia Centre for Research Excellence in the Evaluation, Management and Health Care Needs of Polycystic Ovary Syndrome (N.A.B., M.D.H. and R.J.R.; GTN1078444) and the Centre for Research Excellence on Women’s Health in Reproductive life (R.A., R.J.R. and K.H.; GTN1171592) and the UK Medical Research Council (R.A.A.; grant no. G1100357). The funders did not play any role in the study design, data collection and analysis, decision to publish or preparation of the manuscript. The authors of this manuscript have nothing to declare and no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.
Collapse
Affiliation(s)
| | | | - Katja Hummitzsch
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Nicole A Bastian
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Monica D Hartanti
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
- Faculty of Medicine, Universitas Trisakti, Jakarta, Indonesia
| | - Helen F Irving-Rodgers
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
- School of Medical Science, Griffith University, Gold Coast Campus, Southport, QLD, Australia
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Raymond J Rodgers
- Correspondence address. Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia. E-mail:
| |
Collapse
|
124
|
Bertho S, Neyroud AS, Brun T, Jaillard S, Bonnet F, Ravel C. Anti-Müllerian hormone: A function beyond the Müllerian structures. Morphologie 2021; 106:252-259. [PMID: 34924282 DOI: 10.1016/j.morpho.2021.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 10/19/2022]
Abstract
The anti-Müllerian hormone (AMH) is a heterodimeric glycoprotein belonging to the TGFb superfamily implicated in human embryonic development. This hormone was first described as allowing regression of the epithelial embryonic Müllerian structures in males, which would otherwise differentiate into the uterus and fallopian tubes. It activates a signaling pathway mediated by two transmembrane receptors. Binding of AMH to its receptor induces morphological changes leading to the degeneration of Müllerian ducts. Recently, new data has shown the role played by this hormone on structures other than the genital tract. If testicular AMH expression decreases in humans over the course of a lifetime, synthesis may persist in other tissues in adulthood. The mechanisms underlying its production have been unveiled. The aim of this review is to describe the different pathways in which AMH has been identified and plays a pivotal role.
Collapse
Affiliation(s)
- S Bertho
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France.
| | - A S Neyroud
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France; Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France
| | - T Brun
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France
| | - S Jaillard
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France; Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France
| | - F Bonnet
- CHU Rennes, Service d'Endocrinologie, 35000 Rennes, France
| | - C Ravel
- CHU Rennes, Département de Gynécologie-Obstétrique-Reproduction-CECOS, 35000 Rennes, France; Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000 Rennes, France
| |
Collapse
|
125
|
Impact of intra-uterine life on future health. ANNALES D'ENDOCRINOLOGIE 2021; 83:54-58. [PMID: 34896342 DOI: 10.1016/j.ando.2021.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 12/25/2022]
Abstract
Since the emergence of the concept of developmental origins of health and disease (DOHaD), suggested by Barker in the 1980s, numerous epidemiological studies in humans have confirmed the relationship between maternal obesity during pregnancy and the risk of offspring developing various chronic adult illnesses. These effects of intrauterine life are independent of inheritance of disease susceptibility genes and/or socio-economic factors. Regarding potential mechanisms, recent data from animal models suggests a role of insulin resistance early in development. Another potential mechanism, in the case of maternal obesity, is increased placental nutrient transfer. The DOHaD concept also includes fetal exposure to environmental endocrine disruptors (EEDs). A Danish group for the first time recently analyzed EED passage across the placenta in humans throughout pregnancy. This study showed different levels of bioaccumulation depending on the fetal organ, with greater vulnerability in male than female fetuses. Recent clinical studies suggested an association between fetal exposure to particular EEDs and precocious puberty, increased incidence of cryptorchidism and impaired sperm quality in adulthood. These modifications of the in-utero environment also appear to be responsible for epigenetic changes that are transmittable over several generations. A recent example of this is the demonstration of the transmission of polycystic ovary syndrome (PCOS) in mice. In summary, an increasing number of examples of the impact of intrauterine life on the health of offspring have appeared in recent years, illustrating the important role that endocrinologists can play in preventing particular pathologies in future generations.
Collapse
|
126
|
Du M, Zhang J, Yu X, Guan Y. Elevated Anti-Müllerian Hormone Is an Independent Risk Factor for Preterm Birth Among Patients With Overweight Polycystic Ovary Syndrome. Front Endocrinol (Lausanne) 2021; 12:788000. [PMID: 34956097 PMCID: PMC8692368 DOI: 10.3389/fendo.2021.788000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/23/2021] [Indexed: 11/29/2022] Open
Abstract
Objective To explore whether elevated anti-Müllerian hormone (AMH) levels affect the rate of preterm birth (PTB) among PCOS patients with different BMIs. Methods In this retrospective cohort study, patients with PCOS who had undergone IVF/ICSI from January 2017 to December 2019 were included for potential evaluation. A total of 2368 singleton live births from PCOS patients were included. According to the BMI, all the PCOS patients were divided into two groups: BMI<24 kg/m2 and BMI≥24 kg/m2. In total, 1339 PCOS patients with a BMI<24 kg/m2 were grouped according to their serum AMH levels: ① <2.71 ng/ml (n=333), ② 2.71-4.08 ng/ml (n=330), ③ 4.09-6.45 ng/ml (n=351), and ④ >6.45 ng/ml (n=325). Additionally, 1029 cycles of patients with a BMI≥24 kg/m2 were grouped according to the serum AMH level: ① <2.71 ng/ml (n=255), ② 2.71-4.08 ng/ml (n=267), ③ 4.09-6.45 ng/ml (n=239), and ④ >6.45 ng/ml (n=268), with <2.71 ng/ml being considered the reference group. The grouping was based mainly on the interquartile range of serum AMH levels. The primary outcome of the study was PTB. The secondary outcomes were low birth weight (LBW), small for gestational age (SGA), macrosomia and large for gestational age (LGA). Results Regarding PCOS patients with a BMI<24 kg/m2, compared with the PTB rate of the AMH <2.71 ng/ml group, the PTB rates of the different groups were not significantly different (AMH 2.71-4.08, AOR (95% CI)=1.01 (0.52-2.00), P=0.99; AMH 4.09-6.45, AOR (95% CI)=0.93 (0.45-1.91), P=0.85; AMH>6.45, AOR (95% CI)=0.78 (0.35-1.73), P=0.54). Regarding PCOS patients with a BMI ≥24 kg/m2, compared with the PTB rate of the AMH <2.71 ng/ml group, the PTB rate of the AMH>6.45 ng/ml group was significantly higher (OR=2.47; 95% CI=1.34-4.55). After multiple logistic regression analysis, the risk of PTB in the AMH>6.45 ng/ml group was 2.1 times that in the AMH<2.71 ng/ml group (AOR=2.1, 95% CI=1.01-4.37, P=0.04). However, no statistically significant difference was found in the rate of SGA, LBW, macrosomia or LGA among patients in the different serum AMH groups. Conclusion For PCOS patients, a BMI≥24 kg/m2 plus serum AMH>6.45 ng/ml (75th percentile) is an independent risk factor for PTB.
Collapse
Affiliation(s)
| | | | | | - Yichun Guan
- The Reproductive Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
127
|
Ma X, Li X, Ma L, Chen Y, He S. Soy isoflavones alleviate polycystic ovary syndrome in rats by regulating NF- κB signaling pathway. Bioengineered 2021; 12:7215-7223. [PMID: 34546841 PMCID: PMC8806892 DOI: 10.1080/21655979.2021.1979864] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/02/2022] Open
Abstract
Soy isoflavones have been widely used in the treatment of clinical gynecological diseases. The aim of this study was to investigate the therapeutic effect and molecular mechanism of Soy isoflavones on rats with polycystic ovary syndrome (PCOS). Sprague-Dawley rats were orally administered 1 mg/kg letrozole for 21 consecutive days to induce the PCOS rat model. After PCOS induction, Soy isoflavones (100 mg/kg) or metformin (Positive control; 500 mg/kg) was administered continuously for 28 days. Then, H&E staining was used to observe the pathological changes of ovary. The serum hormone levels and the levels of antioxidant and inflammatory cytokines in ovarian tissue were detected. Additionally, the expression of NF-κB signaling pathway protein was detected by Western blot. Our results showed that soy isoflavones treatment significantly reduced the body weight, ovarian volume and weight, and improved estrous cycle in PCOS rats. H&E staining showed that the number of cystic dilated follicles and atretic follicles in ovarian tissue diminished, showing healthy follicles and corpus luteum. In addition, soy isoflavones treatment markedly decreased serum testosterone and luteinizing hormone (LH) levels, as well as oxidative stress levels and inflammation levels, and increased estradiol (E2) and follicle stimulating hormone (FSH) levels. At the same time, Soy isoflavones treatment inhibit the phosphorylation level of NF-κB p65 and increased the IκBα expression in ovarian tissues of PCOS rats. Overall, Soy isoflavones can improve ovarian morphology and hormone disorders in PCOS rats by inhibiting the activity of NF-κB pathway and enhancing anti-inflammatory and antioxidant capacity.
Collapse
Affiliation(s)
- Xiaohong Ma
- The Center for Reproductive Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiaorong Li
- The Center for Reproductive Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - LiLi Ma
- The Center for Reproductive Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yan Chen
- The Center for Reproductive Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Shengyu He
- Department of Gynecology and Obstetrics, Shizuishan Central Hospital, the Fifth People’ S Hospital of Ningxia, Shizuishan City, China
| |
Collapse
|
128
|
Ho EV, Shi C, Cassin J, He MY, Nguyen RD, Ryan GE, Tonsfeldt KJ, Mellon PL. Reproductive Deficits Induced by Prenatal Antimüllerian Hormone Exposure Require Androgen Receptor in Kisspeptin Cells. Endocrinology 2021; 162:6371276. [PMID: 34529765 PMCID: PMC8507963 DOI: 10.1210/endocr/bqab197] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Indexed: 11/19/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common reproductive disorder characterized by elevated androgens and antimüllerian hormone (AMH). These hormones remain elevated throughout pregnancy, and potential effects of hormone exposure on offspring from women with PCOS remain largely unexplored. Expanding on recent reports of prenatal AMH exposure in mice, we have fully characterized the reproductive consequences of prenatal AMH (pAMH) exposure throughout the lifespan of first- and second-generation offspring of both sexes. We also sought to elucidate mechanisms underlying pAMH-induced reproductive effects. There is a known reciprocal relationship between AMH and androgens, and in PCOS and PCOS-like animal models, androgen feedback is dysregulated at the level of the hypothalamus. Kisspeptin neurons express androgen receptors and play a critical role in sexual development and function. We therefore hypothesized that pAMH-induced reproductive phenotypes would be mediated by androgen signaling at the level of kisspeptin cells. We tested the pAMH model in kisspeptin-specific androgen receptor knockout (KARKO) mice and found that virtually all pAMH-induced phenotypes assayed are eliminated in KARKO offspring compared to littermate controls. By demonstrating the necessity of androgen receptor in kisspeptin cells to induce pAMH phenotypes, we have advanced understanding of the interactions between AMH and androgens in the context of prenatal exposure, which could have significant implications for children of women with PCOS.
Collapse
Affiliation(s)
- Emily V Ho
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Chengxian Shi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Jessica Cassin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Michelle Y He
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Ryan D Nguyen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Genevieve E Ryan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Karen J Tonsfeldt
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
- Correspondence: Pamela L. Mellon, PhD, Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0674, USA.
| |
Collapse
|
129
|
GnRH neurons recruit astrocytes in infancy to facilitate network integration and sexual maturation. Nat Neurosci 2021; 24:1660-1672. [PMID: 34795451 DOI: 10.1038/s41593-021-00960-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 10/08/2021] [Indexed: 12/25/2022]
Abstract
Neurons that produce gonadotropin-releasing hormone (GnRH), which control fertility, complete their nose-to-brain migration by birth. However, their function depends on integration within a complex neuroglial network during postnatal development. Here, we show that rodent GnRH neurons use a prostaglandin D2 receptor DP1 signaling mechanism during infancy to recruit newborn astrocytes that 'escort' them into adulthood, and that the impairment of postnatal hypothalamic gliogenesis markedly alters sexual maturation by preventing this recruitment, a process mimicked by the endocrine disruptor bisphenol A. Inhibition of DP1 signaling in the infantile preoptic region, where GnRH cell bodies reside, disrupts the correct wiring and firing of GnRH neurons, alters minipuberty or the first activation of the hypothalamic-pituitary-gonadal axis during infancy, and delays the timely acquisition of reproductive capacity. These findings uncover a previously unknown neuron-to-neural-progenitor communication pathway and demonstrate that postnatal astrogenesis is a basic component of a complex set of mechanisms used by the neuroendocrine brain to control sexual maturation.
Collapse
|
130
|
Anti-Müllerian Hormone in Pathogenesis, Diagnostic and Treatment of PCOS. Int J Mol Sci 2021; 22:ijms222212507. [PMID: 34830389 PMCID: PMC8619458 DOI: 10.3390/ijms222212507] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 01/19/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among reproductive-aged women. It is characterized by chronic anovulation, hyperandrogenism, and the presence of polycystic ovary in ultrasound examination. PCOS is specified by an increased number of follicles at all growing stages, mainly seen in the preantral and small antral follicles and an increased serum level of Anti-Müllerian Hormone (AMH). Because of the strong correlation between circulating AMH levels and antral follicle count on ultrasound, Anti-Müllerian Hormone has been proposed as an alternative marker of ovulatory dysfunction in PCOS. However, the results from the current literature are not homogeneous, and the specific threshold of AMH in PCOS and PCOM is, therefore, very challenging. This review aims to update the current knowledge about AMH, the pathophysiology of AMH in the pathogenesis of PCOS, and the role of Anti-Müllerian Hormone in the treatment of this syndrome.
Collapse
|
131
|
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: 81] [Impact Index Per Article: 20.3] [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
|
132
|
Moore AM, Lohr DB, Coolen LM, Lehman MN. Prenatal Androgen Exposure Alters KNDy Neurons and Their Afferent Network in a Model of Polycystic Ovarian Syndrome. Endocrinology 2021; 162:bqab158. [PMID: 34346492 PMCID: PMC8402932 DOI: 10.1210/endocr/bqab158] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Indexed: 02/08/2023]
Abstract
Polycystic ovarian syndrome (PCOS), the most common endocrinopathy affecting women worldwide, is characterized by elevated luteinizing hormone (LH) pulse frequency due to the impaired suppression of gonadotrophin-releasing hormone (GnRH) release by steroid hormone negative feedback. Although neurons that co-express kisspeptin, neurokinin B, and dynorphin (KNDy cells) were recently defined as the GnRH/LH pulse generator, little is understood about their role in the pathogenesis of PCOS. We used a prenatal androgen-treated (PNA) mouse model of PCOS to determine whether changes in KNDy neurons or their afferent network underlie altered negative feedback. First, we identified elevated androgen receptor gene expression in KNDy cells of PNA mice, whereas progesterone receptor and dynorphin gene expression was significantly reduced, suggesting elevated androgens in PCOS disrupt progesterone negative feedback via direct actions upon KNDy cells. Second, we discovered GABAergic and glutamatergic synaptic input to KNDy neurons was reduced in PNA mice. Retrograde monosynaptic tract-tracing revealed a dramatic reduction in input originates from sexually dimorphic afferents in the preoptic area, anteroventral periventricular nucleus, anterior hypothalamic area and lateral hypothalamus. These results reveal 2 sites of neuronal alterations potentially responsible for defects in negative feedback in PCOS: changes in gene expression within KNDy neurons, and changes in synaptic inputs from steroid hormone-responsive hypothalamic regions. How each of these changes contribute to the neuroendocrine phenotype seen in in PCOS, and the role of specific sets of upstream KNDy afferents in the process, remains to be determined.
Collapse
Affiliation(s)
- Aleisha M Moore
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Dayanara B Lohr
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Lique M Coolen
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Michael N Lehman
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
133
|
Arslanca T, Ecemis T, Kiseli M, Arslanoglu E, Kotanoğlu MS, Caglar GS. Pregnancy outcome of freeze thaw cycles of polycystic ovary syndrome patients regarding the anti-Müllerian hormone percentile. J OBSTET GYNAECOL 2021; 42:1319-1324. [PMID: 34698605 DOI: 10.1080/01443615.2021.1962819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Patients with polycystic ovary syndrome display increased levels of anti-Müllerian hormone. Frozen-thawed embryo transfer (FET) prevents ovarian hyperstimulation and results in better pregnancy outcome in PCOS patients. Therefore, we aimed to evaluate the effect of serum AMH levels on the pregnancy outcome of FET cycles in PCOS patients. 110 infertile women with PCOS who were recommended for embryo cryopreservation followed by FET. The patients' AMH levels were evaluated, and the age-related AMH percentiles were determined. The patients were then grouped according to AMH percentiles, namely, 75th-90th percentile (Group 1) and higher than the 90th percentile (Group 2).A total of 110 PCOS patients who conceived in Frozen Embryo Transfer (FET)-In-vitro Fertilisation (IVF) cycles were included in this study. The preterm delivery rates in FET cycles were higher in the group of PCOS patients with AMH levels greater than the 90th percentile than in patients in the 75th-90th percentile group (50% vs 28.8%, p = .024). In conclusion, PCOS patients with AMH levels higher than the 90th percentile had substantially higher preterm delivery rates than those with AMH levels at the 75th-90th percentile, suggesting the need for closer follow-up. Further studies are needed to elucidate the underlying mechanisms behind this correlation.IMPACT STATEMENTWhat is already known on this subject? The association of AMH levels with the risk of adverse pregnancy outcomes has been previously investigated. In women with PCOS, substantially elevated AMH levels were significantly associated with preterm birth.What do the results of this study add? Results showed that the PCOS patients with higher AMH levels and underwent assisted reproductive treatment demonstrated an increased risk of preterm labour than the PCOS patients with lower AMH levels.What are the implications of these findings for clinical practice and/or further research? In women with PCOS, substantially elevated AMH levels were significantly related to preterm birth, suggesting the need for closer follow-up in this population and the need for further studies to elucidate the underlying mechanisms behind this correlation.
Collapse
Affiliation(s)
- Tufan Arslanca
- Department of Obstetrics and Gynecology, Ufuk University, Ankara, Turkey
| | - Tolga Ecemis
- Private Gynecology and Obstetrics Clinic, Ankara, Turkey
| | - Mine Kiseli
- Department of Obstetrics and Gynecology, Ufuk University, Ankara, Turkey
| | | | - Mustafa S Kotanoğlu
- Department of Anesthesiology, Ankara Training and Research Hospital, Ankara, Turkey
| | - Gamze S Caglar
- Department of Obstetrics and Gynecology, Ufuk University, Ankara, Turkey
| |
Collapse
|
134
|
Kim KW. Unravelling Polycystic Ovary Syndrome and Its Comorbidities. J Obes Metab Syndr 2021; 30:209-221. [PMID: 34497157 PMCID: PMC8526288 DOI: 10.7570/jomes21043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/11/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a chronic multisystem endocrine disorder that affects women of reproductive age. In the ovary, the dynamic balance between dormant and growing follicles that culminates in ovulation becomes dysfunctional in the presence of excessive androgen production (ovarian/adrenal/peripheral). Moreover, hyperandrogenicity in pregnancy affects fetal development in utero and is linked to maternal pregnancy complications. Hormonal imbalance, ovarian dysfunction, and central obesity often emerge in these patients during adolescence. Once disordered physiological changes develop in PCOS, a vicious cycle ensues, leading to reproductive, metabolic, and psychological comorbidities. With the alarming increase of the number of young adults with a high degree of obesity in Korea, the prevalence of PCOS has also considerably increased. Timely and accurate screening, multicomponent healthy lifestyle modifications for both patients and family members, and comprehensive medical interventions based on international evidence-based guidelines are essential for curtailing PCOS and its comorbidities.
Collapse
Affiliation(s)
- Kyung-Wook Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Dongtan Jeil Women's Hospital and Sangwoon Medical Institute, Hwaseong, Korea.,Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
135
|
Mousa A, Joham A, Boyle J. Polycystic Ovary Syndrome. Semin Reprod Med 2021; 39:69-70. [PMID: 34530478 DOI: 10.1055/s-0041-1735506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Aya Mousa
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, VIC, Australia
| | - Anju Joham
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, VIC, Australia.,Diabetes and Vascular Medicine Unit, Monash Health, VIC, Australia
| | - Jacqueline Boyle
- Monash Centre for Health Research and Implementation (MCHRI), School of Public Health and Preventive Medicine, Monash University, VIC, Australia.,Women's and Children's, Monash Health, VIC, Australia
| |
Collapse
|
136
|
Mimouni NEH, Giacobini P. Polycystic ovary syndrome mouse model by prenatal exposure to high anti-Müllerian hormone. STAR Protoc 2021; 2:100684. [PMID: 34401772 PMCID: PMC8348292 DOI: 10.1016/j.xpro.2021.100684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Here, we describe a protocol that provides the steps required for the generation of a mouse model of polycystic ovary syndrome (PCOS) by exposing dams to elevated levels of anti-Müllerian hormone during late gestation. This protocol also describes the steps required to assess the PCOS-like equivalents of the Rotterdam PCOS diagnostic criteria in mice. For complete details on the use and execution of this protocol, please refer to Tata et al. (2018) and Mimouni et al. (2021). Protocol to induce PCOS traits in offspring using prenatal exposure to high AMH Procedure describing key steps for reliable assessment of PCOS-like reproductive traits Applicable to other established animal models of PCOS
Collapse
Affiliation(s)
- Nour El Houda Mimouni
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, 59000 Lille, France
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, 59000 Lille, France
| |
Collapse
|
137
|
GÖKÇE Ş, HERKİLOĞLU D. Cardiovascular risk factors in polycystic ovary syndrome; the relationship of dyslipidemia and obesity. JOURNAL OF HEALTH SCIENCES AND MEDICINE 2021. [DOI: 10.32322/jhsm.901791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
138
|
Piltonen TT, Arffman RK, Joham AE. Natural History of Polycystic Ovary Syndrome and New Advances in the Epidemiology. Semin Reprod Med 2021; 39:94-101. [PMID: 34464984 DOI: 10.1055/s-0041-1735211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrinopathy in women, affecting up to 15% of the female population. The natural history of the syndrome is complex, including both androgen exposure in early life and adiposity-driven dysfunction involving also dysregulated, also involving altered hypothalamus-ovarian crosstalk. The manifestations can arise as early as childhood or puberty onward, suggesting that genetic susceptibility is an important etiological factor. In adulthood, women with PCOS present with reproductive, metabolic, and psychological health issues at a population-based level. Epidemiological studies on large datasets offer an excellent opportunity to evaluate health effects and costs related to the syndrome. Hospital or insurance-based datasets are also available; however, the results are not representative of the female population in the community. More longitudinal studies spanning from early childhood to late adulthood are needed to assess the long-term health impact and early manifestations of PCOS. Moreover, the identification of women with PCOS from large datasets can be expensive. Self-reported symptoms or PCOS diagnosis may offer a feasible approach.
Collapse
Affiliation(s)
- Terhi T Piltonen
- PEDEGO Research Unit, Department of Obstetrics and Gynecology, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Riikka K Arffman
- PEDEGO Research Unit, Department of Obstetrics and Gynecology, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Anju E Joham
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia.,Department of Diabetes and Vascular Medicine, Monash Health, Melbourne, Australia
| |
Collapse
|
139
|
Stener-Victorin E, Deng Q. Epigenetic inheritance of polycystic ovary syndrome - challenges and opportunities for treatment. Nat Rev Endocrinol 2021; 17:521-533. [PMID: 34234312 DOI: 10.1038/s41574-021-00517-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the main cause of female infertility worldwide and is associated with a substantially increased lifetime risk of comorbidities, including type 2 diabetes mellitus, psychiatric disorders and gynaecological cancers. Despite its high prevalence (~15%) and substantial economic burden, the aetiology of PCOS remains elusive. The genetic loci linked to PCOS so far account for only ~10% of its heritability, which is estimated at 70%. However, growing evidence suggests that altered epigenetic and developmental programming resulting from hormonal dysregulation of the maternal uterine environment contributes to the pathogenesis of PCOS. Male as well as female relatives of women with PCOS are also at an increased risk of developing PCOS-associated reproductive and metabolic disorders. Although PCOS phenotypes are highly heterogenous, hyperandrogenism is thought to be the principal driver of this condition. Current treatments for PCOS are suboptimal as they can only alleviate some of the symptoms; preventative and targeted treatments are sorely needed. This Review presents an overview of the current understanding of the aetiology of PCOS and focuses on the developmental origin and epigenetic inheritance of this syndrome.
Collapse
Affiliation(s)
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
140
|
Oride A, Kanasaki H, Tumurbaatar T, Tumurgan Z, Okada H, Kyo S. Effect of anti-Müllerian hormone in hypothalamic Kiss-1- and GnRH-producing cell models. Gynecol Endocrinol 2021; 37:841-847. [PMID: 34236272 DOI: 10.1080/09513590.2021.1950134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Purpose: Anti-Müllerian hormone (AMH) is one of the local factors involved in follicle development. In addition, AMH and its receptor are broadly expressed throughout the body. In this study, we examined how AMH modifies gene expression of Kiss-1 and GnRH.Materials and methods: mHypoA-50 and mHypoA-55 cells were originated from the hypothalamic anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC), respectively, and these cells are known as Kiss-1 (which encodes kisspeptin) expressing cell models. These cells also express gonadotropin-releasing hormone (GnRH) genes. Our experiments were performed useing these cell models.Results: Both mHypoA-50 and mHypoA-55 hypothalamic cells expressed AMH and AMH receptor type 2 (AMHR2). Exogenous AMH failed to alter the expression levels of the Kiss-1 gene in both cell models but significantly increased GnRH gene expression by 1.73 ± 0.2-fold at 100 pM in mHypoA-50 AVPV cells and by 1.74 ± 0.17-fold at 1 nM in mHypoA-55 ARC cells. AMH also augmented GnRH protein expression in both cell models. Similar to the phenomenon observed in the hypothalamic cell lines, 100 pM AMH significantly increased GnRH, but not Kiss-1, mRNA expression in primary cultures of fetal rat brain cells. Kisspeptin-10 (KP10) increased Kiss-1 gene expression in mHypoA-55 ARC cells but this was blocked by AMH. AMH did not alter the expression of the kisspeptin receptor (Kiss1R) or that of neurokinin B or dynorphin A in mHypoA-55 ARC cells.Conclusions: It was demonstrated that AMH participates in hypothalamic-pituitary-gonadal axis control by stimulating GnRH expression. In addition, AMH might be a potent repressor of Kiss-1 gene expression induced by KP10.
Collapse
Affiliation(s)
- Aki Oride
- Department of Obstetrics and Gynecology, School of Medicine, Shimane University, Izumo, Japan
| | - Haruhiko Kanasaki
- Department of Obstetrics and Gynecology, School of Medicine, Shimane University, Izumo, Japan
| | - Tuvshintugs Tumurbaatar
- Department of Obstetrics and Gynecology, School of Medicine, Shimane University, Izumo, Japan
| | - Zolzaya Tumurgan
- Department of Obstetrics and Gynecology, School of Medicine, Shimane University, Izumo, Japan
| | - Hiroe Okada
- Department of Obstetrics and Gynecology, School of Medicine, Shimane University, Izumo, Japan
| | - Satoru Kyo
- Department of Obstetrics and Gynecology, School of Medicine, Shimane University, Izumo, Japan
| |
Collapse
|
141
|
Veikkolainen V, Ali N, Doroszko M, Kiviniemi A, Miinalainen I, Ohlsson C, Poutanen M, Rahman N, Elenius K, Vainio SJ, Naillat F. Erbb4 regulates the oocyte microenvironment during folliculogenesis. Hum Mol Genet 2021; 29:2813-2830. [PMID: 32716031 DOI: 10.1093/hmg/ddaa161] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/17/2020] [Accepted: 07/17/2020] [Indexed: 12/16/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders leading to infertility in women affecting reproductive, endocrine and metabolic systems. Recent genomewide association studies on PCOS cohorts revealed a single nucleotide polymorphism (SNP) in the ERBB4 receptor tyrosine kinase 4 gene, but its role in ovary development or during folliculogenesis remains poorly understood. Since no genetic animal models mimicking all PCOS reproductive features are available, we conditionally deleted Erbb4 in murine granulosa cells (GCs) under the control of Amh promoter. While we have demonstrated that Erbb4 deletion displayed aberrant ovarian function by affecting the reproductive function (asynchronous oestrous cycle leading to few ovulations and subfertility) and metabolic function (obesity), their ovaries also present severe structural and functional abnormalities (impaired oocyte development). Hormone analysis revealed an up-regulation of serum luteinizing hormone, hyperandrogenism, increased production of ovarian and circulating anti-Müllerian hormone. Our data implicate that Erbb4 deletion in GCs leads to defective intercellular junctions between the GCs and oocytes, causing changes in the expression of genes regulating the local microenvironment of the follicles. In vitro culture assays reducing the level of Erbb4 via shRNAs confirm that Erbb4 is essential for regulating Amh level. In conclusion, our results indicate a functional role for Erbb4 in the ovary, especially during folliculogenesis and its reduced expression plays an important role in reproductive pathophysiology, such as PCOS development.
Collapse
Affiliation(s)
- Ville Veikkolainen
- Institute of Biomedicine and MediCity Research Laboratory, University of Turku, FI-20520 Turku, Finland
| | - Nsrein Ali
- Organogenesis Laboratory, Department of Medical Biochemistry and Molecular Biology, Biocenter Oulu, University of Oulu, FI-90014 Oulu, Finland
| | - Milena Doroszko
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, FI-20520 Turku, Finland.,Department of Immunology Genetics and Pathology, Section for Neuro-oncology, Uppsala University, 752 36 Uppsala, Sweden
| | - Antti Kiviniemi
- Organogenesis Laboratory, Department of Medical Biochemistry and Molecular Biology, Biocenter Oulu, University of Oulu, FI-90014 Oulu, Finland
| | - Ilkka Miinalainen
- Electron Microscopy Unit, Biocenter Oulu, University of Oulu, FI-90220 Oulu, Finland
| | - Claes Ohlsson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-41345 Gothenburg, Sweden
| | - Matti Poutanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, FI-20520 Turku, Finland.,Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-41345 Gothenburg, Sweden
| | - Nafis Rahman
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, FI-20520 Turku, Finland
| | - Klaus Elenius
- Institute of Biomedicine and MediCity Research Laboratory, University of Turku, FI-20520 Turku, Finland.,Department of Oncology, Turku University Hospital, FI-20520 Turku, Finland
| | - Seppo J Vainio
- Department of Immunology Genetics and Pathology, Section for Neuro-oncology, Uppsala University, 752 36 Uppsala, Sweden.,InfoTech Oulu, Oulu University and Biobank Borealis of Northern Finland, Oulu University Hospital, University of Oulu, FI-90014 Oulu, FINLAND
| | - Florence Naillat
- Organogenesis Laboratory, Department of Medical Biochemistry and Molecular Biology, Biocenter Oulu, University of Oulu, FI-90014 Oulu, Finland
| |
Collapse
|
142
|
Fraser GL, Obermayer-Pietsch B, Laven J, Griesinger G, Pintiaux A, Timmerman D, Fauser BCJM, Lademacher C, Combalbert J, Hoveyda HR, Ramael S. Randomized Controlled Trial of Neurokinin 3 Receptor Antagonist Fezolinetant for Treatment of Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2021; 106:e3519-e3532. [PMID: 34000049 PMCID: PMC8372662 DOI: 10.1210/clinem/dgab320] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Indexed: 12/14/2022]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS), a highly prevalent endocrine disorder characterized by hyperandrogenism, is the leading cause of anovulatory infertility. OBJECTIVE This proof-of-concept study evaluated clinical efficacy and safety of the neurokinin 3 (NK3) receptor antagonist fezolinetant in PCOS. METHODS This was a phase 2a, randomized, double-blind, placebo-controlled, multicenter study (EudraCT 2014-004409-34). The study was conducted at 5 European clinical centers. Women with PCOS participated in the study. Interventions included fezolinetant 60 or 180 mg/day or placebo for 12 weeks. The primary efficacy end point was change in total testosterone. Gonadotropins, ovarian hormones, safety and tolerability were also assessed. RESULTS Seventy-three women were randomly assigned, and 64 participants completed the study. Adjusted mean (SE) changes in total testosterone from baseline to week 12 for fezolinetant 180 and 60 mg/day were -0.80 (0.13) and -0.39 (0.12) nmol/L vs -0.05 (0.10) nmol/L with placebo (P < .001 and P < .05, respectively). Adjusted mean (SE) changes from baseline in luteinizing hormone (LH) for fezolinetant 180 and 60 mg/d were -10.17 (1.28) and -8.21 (1.18) vs -3.16 (1.04) IU/L with placebo (P < .001 and P = .002); corresponding changes in follicle-stimulating hormone (FSH) were -1.46 (0.32) and -0.92 (0.30) vs -0.57 (0.26) IU/L (P = .03 and P = .38), underpinning a dose-dependent decrease in the LH-to-FSH ratio vs placebo (P < .001). Circulating levels of progesterone and estradiol did not change significantly vs placebo (P > .10). Fezolinetant was well tolerated. CONCLUSION Fezolinetant had a sustained effect to suppress hyperandrogenism and reduce the LH-to-FSH ratio in women with PCOS.
Collapse
Affiliation(s)
- Graeme L Fraser
- Correspondence: Graeme L. Fraser, PhD, EPICS Therapeutics, 47 Rue Adrienne Bolland, 6041 Gosselies, Belgium.
| | | | - Joop Laven
- Erasmus MC, 3015 Rotterdam, the Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Sati A, Prescott M, Holland S, Jasoni CL, Desroziers E, Campbell RE. Morphological evidence indicates a role for microglia in shaping the PCOS-like brain. J Neuroendocrinol 2021; 33:e12999. [PMID: 34216402 DOI: 10.1111/jne.12999] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/21/2022]
Abstract
Although polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility worldwide, the aetiology of the disorder remains poorly defined. Animal-based evidence highlights the brain as a prime suspect in both the development and maintenance of PCOS. Prenatally androgenised (PNA) models of PCOS exhibit excessive GABAergic wiring associated with PCOS-like reproductive deficits in adulthood, with aberrant brain wiring detected as early as postnatal day (P) 25, prior to disease onset, in the PNA mouse. The mechanisms underlying this aberrant brain wiring remain unknown. Microglia, the immune cells of the brain, are regulators of neuronal wiring across development, mediating both the formation and removal of neuronal inputs. Here, we tested the hypothesis that microglia play a role in the excessive GABAergic wiring that leads to PCOS-like features in the PNA brain. Using specific immunolabelling, microglia number and morphology associated with activation states were analysed in PNA and vehicle-treated controls across developmental timepoints, including embryonic day 17.5, P0, P25 and P60 (n = 7-14 per group), and in two regions of the hypothalamus implicated in fertility regulation. At P0, fewer amoeboid microglia were observed in the rostral preoptic area (rPOA) of PNA mice. However, the greatest changes were observed at P25, with PNA mice exhibiting fewer total microglia, and specifically fewer "sculpting" microglia, in the rPOA. Based on these findings, we assessed microglia-mediated refinement of GABAergic synaptic terminals at two developmental stages of peak synaptic refinement: P7 and P15 (n = 7 per group). PNA mice showed a reduction in the uptake of GABAergic synaptic material at P15. These findings reveal time-specific changes in the microglia population and refinement of GABAergic inputs in a mouse model of PCOS driven by prenatal androgen excess and suggest a role for microglia in shaping the atypical brain wiring associated with the development of PCOS features.
Collapse
Affiliation(s)
- Aisha Sati
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Melanie Prescott
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Sarah Holland
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Christine L Jasoni
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Elodie Desroziers
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
144
|
Sundström Poromaa I. Antimüllerian hormone and spontaneous preterm birth: more research is needed before we can change clinical guidelines. Fertil Steril 2021; 115:325. [PMID: 33579523 DOI: 10.1016/j.fertnstert.2020.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 10/22/2022]
|
145
|
Stener-Victorin E, Deng Q. Transmission of Polycystic Ovary Syndrome via Epigenetic Inheritance. Trends Mol Med 2021; 27:723-724. [PMID: 34127396 DOI: 10.1016/j.molmed.2021.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022]
Abstract
A recent study by Mimouni et al. shows that late gestation exposure to anti-Müllerian hormone (AMH) results in transgenerational transmission of a polycystic ovary syndrome (PCOS)-like phenotype in mice. Altered DNA methylation underlies transmission and was also observed in women with PCOS. Epigenetics-based therapy reversed some PCOS-like phenotypic traits when applied to F3 female mice.
Collapse
Affiliation(s)
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
146
|
Makolle S, Catteau-Jonard S, Robin G, Dewailly D. Revisiting the serum level of anti-Müllerian hormone in patients with functional hypothalamic anovulation. Hum Reprod 2021; 36:1043-1051. [PMID: 33580689 DOI: 10.1093/humrep/deab024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 12/20/2020] [Indexed: 12/24/2022] Open
Abstract
STUDY QUESTION Are serum levels of anti-Müllerian hormone (AMH) normal in patients with functional hypothalamic anovulation (FHA)? SUMMARY ANSWER Our study confirms that in the general FHA population, serum AMH levels are not decreased, but if patients with polycystic ovarian morphology (PCOM) are excluded, levels become significantly lower, as in other situations of gonadotropic insufficiency. WHAT IS KNOWN ALREADY In most situations of low LH (physiological, pharmacological or pathological), serum AMH levels are low. However, paradoxically, many publications have reported normal or even increased serum AMH levels in FHA patients. STUDY DESIGN, SIZE, DURATION Retrospective observational study conducted in an academic centre. The data concerning the study population was collected between 2006 and 2015 from a database including clinical, biological and ultrasound information. PARTICIPANTS/MATERIALS, SETTING, METHODS A total of 45 FHA patients were compared to 37 controls matched based on age and body mass index (BMI). Serum LH, FSH, androstenedione, total testosterone, prolactin and AMH levels were measured by immunoassay. We defined PCOM with strict criteria: a follicle number per ovary (FNPO) ≥ 12 or ≥ 19 per ovary, depending on the date on which the assessment was carried out and the ultrasound device. An AMH level ≥ 35 pmol/l could be a substitute for an excess FNPO. Controls meeting these criteria were not included in this study. MAIN RESULTS AND THE ROLE OF CHANCE There was no significant difference in the ranges of AMH levels between FHA and controls. Using strict criteria to define PCOM status, 46.7% of FHA patients had PCOM. After excluding these patients, the levels of AMH were significantly lower (P < 0.002) in FHA patients compared to controls. Within the FHA group, patients with PCOM had significantly higher ranks of AMH levels and BMI than those without PCOM. However, within the PCOM+ subgroup, the ranks of LH, FSH and A levels were still lower than in controls (P < 0.0001, <0.002 and <0.05, respectively). The positive correlation between AMH and LH was significant in the controls but not in the FHA group. However, in the FHA PCOM+, there was a strong positive correlation between BMI and LH. LIMITATIONS, REASONS FOR CAUTION This is a retrospective study; our controls did not represent the general population as they were recruited in an ART centre; we used a modified classification for PCOM using follicle count and/or AMH level with in-house thresholds to define the follicle excess; the AMH assay used is no longer commercially available. WIDER IMPLICATIONS OF THE FINDINGS Besides biasing the results of AMH assay in FHA patients, the presence of PCOM in FHA patients despite low gonadotropin and androgen levels raises the issue of epigenetically acquired amplification of androgen and/or FSH sensitivity within granulosa cells from polycystic ovaries. In terms of clinical practice, it seems important not to diagnose a low ovarian reserve in FHA patients too quickly on the basis of a decreased AMH level alone. On the contrary, a high AMH level in the context of a menstrual disorder and PCOM should not lead to a misdiagnosis of polycystic ovary syndrome (PCOS) if the basal LH is low. STUDY FUNDING/COMPETING INTEREST(S) None. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Sarah Makolle
- CHU Lille, Unité Fonctionnelle de Gynécologie Endocrinienne, Service de Gynécologie Médicale, Orthogénie et Sexologie, Hôpital Jeanne de Flandre, Lille, France
| | - Sophie Catteau-Jonard
- CHU Lille, Unité Fonctionnelle de Gynécologie Endocrinienne, Service de Gynécologie Médicale, Orthogénie et Sexologie, Hôpital Jeanne de Flandre, Lille, France.,Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean- Pierre Aubert Research Centre, Lille 59945, France
| | - Geoffroy Robin
- CHU Lille, Unité Fonctionnelle de Gynécologie Endocrinienne, Service de Gynécologie Médicale, Orthogénie et Sexologie, Hôpital Jeanne de Flandre, Lille, France.,Lille University, EA 4308 'Gametogenesis and Gamete Quality', Lille, France
| | - Didier Dewailly
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean- Pierre Aubert Research Centre, Lille 59945, France
| |
Collapse
|
147
|
Zhang FF, Zhang Q, Wang YL, Wang FF, Hardiman PJ, Qu F. Intergenerational Influences between Maternal Polycystic Ovary Syndrome and Offspring: An Updated Overview. J Pediatr 2021; 232:272-281. [PMID: 33482217 DOI: 10.1016/j.jpeds.2021.01.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Fang-Fang Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuan-Lin Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fang-Fang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Paul J Hardiman
- Institute for Women's Health, University College London, London, United Kingdom
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
148
|
Li J, Cui L, Jiang X, Zhao H, Zhao S, Shi Y, Wei D, You L, Ma J, Chen ZJ. Transmission of polycystic ovary syndrome susceptibility single-nucleotide polymorphisms and their association with phenotype changes in offspring. Hum Reprod 2021; 35:1711-1718. [PMID: 32619219 DOI: 10.1093/humrep/deaa125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/04/2020] [Indexed: 02/06/2023] Open
Abstract
STUDY QUESTION Does the inheritance of polycystic ovary syndrome (PCOS) susceptibility single-nucleotide polymorphism affect the phenotype of offspring? SUMMARY ANSWER Male offspring who inherit PCOS-related genetic variations from PCOS mothers were more susceptible to developing the metabolic abnormality in their later life. WHAT IS KNOWN ALREADY Genetic factors are considered the major etiology of PCOS. Previous studies have highlighted that offspring of women with PCOS had an increased risk of the same disease or PCOS-like symptoms. STUDY DESIGN, SIZE, DURATION The study involved 172 children born to women with PCOS and 529 children born to non-PCOS women. All offspring were conceived by assisted reproductive technologies. PARTICIPANTS/MATERIALS, SETTING, METHODS The offspring ranged from 1 to 8 years old. Metabolic phenotype analyses were performed in offspring aged from 2 to 8 (N = 619). Sanger sequencing, TaqMan and Sequenom MassARRAY were used to sequence the samples. MAIN RESULTS AND THE ROLE OF CHANCE In male offspring, the fasting insulin (FINS) (P = 0.037) homeostasis model assessment of insulin resistance (HOMA-IR) (P = 0.038) and the homeostasis model assessment of pancreatic beta-cell function (HOMA-β) (P = 0.038) levels were higher in offspring of PCOS mothers compared to controls. In female offspring, PCOS offspring had a significantly higher anti-Müllerian hormone levels (P = 0.001) compared to those from control mothers. In male offspring of PCOS mothers, subjects with a T allele at rs2349415 in the gene FSHR had higher FINS (P = 0.023), HOMA-IR (P = 0.030) and HOMA-β levels (P = 0.013) than those in the homozygous CC group. The same increased trend in FINS, HOMA-IR and HOMA-β levels could be found in the CC and TC group in rs2268361 located in gene FSHR compared to the TT group (P = 0.029, P = 0.030, P = 0.046, respectively). As for rs10818854 in the DENND1A gene, the AA and AG group had a higher FINS (P = 0.037) and HOMA-β (P = 0.008) levels than the homozygous CC group. LIMITATIONS, REASONS FOR CAUTION Firstly, the offspring may be too young to see any phenotype changes. Secondly, this study only analyzed the differences of genotype frequency using the dominant model instead of all three models due to the limited sample size of the homozygous model. The results, therefore, should be replicated and performed in a larger sample size population. Thirdly, environmental impacts cannot be ruled out. WIDER IMPLICATIONS OF THE FINDINGS The findings presented in this thesis add to our understanding the changes in offspring born to PCOS women and remind us to consider early intervention to avoid more severe effects. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the National Key Research and Development Program of China 2017YFC1001000 (to Z.-J.C.), the National Natural Science Foundation of China 81430029 (to Z.-J.C.), 81622021 and 31571548 (to H.Z.), the National Natural Science Foundation of Shandong Province JQ201816 (to H.Z.) and Shandong Provincial Key Research and Development Program 2017G006036 (to L.-L.C.) and 2018YFJH0504 (to Z.-J.C.). There are no conflicts of interest to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Jingyu Li
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Linlin Cui
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Xiao Jiang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Han Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Shigang Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Yuhua Shi
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Daimin Wei
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Li You
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Jinlong Ma
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, Shandong 250012, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200000, China.,Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China
| |
Collapse
|
149
|
Hoyos LR, Visser JA, McLuskey A, Chazenbalk GD, Grogan TR, Dumesic DA. Loss of anti-Müllerian hormone (AMH) immunoactivity due to a homozygous AMH gene variant rs10417628 in a woman with classical polycystic ovary syndrome (PCOS). Hum Reprod 2021; 35:2294-2302. [PMID: 32918081 DOI: 10.1093/humrep/deaa199] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Anti-Müllerian hormone (AMH) is produced by granulosa cells of pre-antral and small antral ovarian follicles. In polycystic ovary syndrome (PCOS), higher levels of serum AMH are usually encountered due to the ample presence of small antral follicles and a high AMH production per follicular unit which have led to the proposal of AMH as a serum diagnostic marker for PCOS or as a surrogate for polycystic ovarian morphology (PCOM). However, heterozygous coding mutations of the AMH gene with decreased in vitro bioactivity have been described in some women with PCOS. Such mutation carriers have a trend toward reduced serum AMH levels compared to noncarriers, although both types of women with PCOS have similar circulating gonadotropin and testosterone (T) levels. This report describes a normal-weight woman with PCOS by NIH criteria with severely reduced AMH levels (index woman with PCOS). Our objective was to examine the molecular basis for her reduced serum AMH levels and to compare her endocrine characteristics to similar-weight women with PCOS and detectable AMH levels. Twenty normoandrogenic ovulatory (control) and 13 age- and BMI-matched women with PCOS (19-35 years; 19-25 kg/m2) underwent transvaginal sonography and serum hormone measures including gonadotropins, sex hormone-binding globulin, total and free T, androstenedione, dehydroepiandrosterone sulfate, estrone, estradiol and AMH. The latter was measured by ELISA (Pico-AMH: Ansh Labs, Webster, TX, USA). Women with PCOS and detectable AMH had higher serum AMH (10.82 (6.74-13.40) ng/ml, median (interquartile range)), total and free T (total T: 55.5 (49.5-62.5) ng/dl; free T: 5.65 (4.75-6.6) pg/ml) levels and greater total antral follicle count (AFC) (46 (39-59) follicles) than controls (AMH: 4.03 (2.47-6.11) ng/ml; total T: 30 (24.5-34.5) ng/dl; free T: 2.2 (1.8-2.45) pg/ml; AFC 16 (14.5-21.5) follicles, P < 0.05, all values), along with a trend toward LH hypersecretion (P = 0.06). The index woman with PCOS had severely reduced serum AMH levels (∼0.1 ng/ml), although she also had a typical NIH-defined PCOS phenotype resembling that of the other women with PCOS and elevated AMH levels. All women with PCOS, including the index woman with PCOS, exhibited LH hypersecretion, hyperandrogenism, reduced serum estrogen/androgen ratios and PCOM. A homozygous Ala515Val variant (rs10417628) in the mature region of AMH was identified in the index woman with PCOS. Recombinant hAMH-515Val displayed normal processing and bioactivity, yet had severely reduced immunoactivity when measured by the commercial pico-AMH ELISA assay by Ansh Labs. In conclusion, homozygous AMH variant rs10417628 may severely impair serum AMH immunoactivity without affecting its bioactivity or PCOS phenotypic expression. Variants in AMH can interfere with serum AMH immunoactivity without affecting the phenotype in PCOS. This observation can be accompanied by discordance between AMH immunoactivity and bioactivity.
Collapse
Affiliation(s)
- Luis R Hoyos
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jenny A Visser
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Anke McLuskey
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gregorio D Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Tristan R Grogan
- Department of Medicine Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
150
|
Chen X, Kong L, Piltonen TT, Gissler M, Lavebratt C. Association of polycystic ovary syndrome or anovulatory infertility with offspring psychiatric and mild neurodevelopmental disorders: a Finnish population-based cohort study. Hum Reprod 2021; 35:2336-2347. [PMID: 32866965 PMCID: PMC7518708 DOI: 10.1093/humrep/deaa192] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/04/2020] [Indexed: 12/24/2022] Open
Abstract
STUDY QUESTION Is maternal polycystic ovary syndrome (PCOS) associated with increased risks for a broad spectrum of psychiatric and mild neurodevelopmental disorders in offspring? SUMMARY ANSWER Maternal PCOS and/or anovulatory infertility is independently, and jointly with maternal obesity, perinatal problems, cesarean delivery and gestational diabetes, associated with increased risks in offspring for almost all groups of psychiatric and mild neurodevelopmental disorders with onset in childhood or adolescence. WHAT IS KNOWN ALREADY Maternal PCOS was previously associated with autism spectrum disorder, attention-deficit/hyperactivity disorders and possibly developmental delay in offspring. Few studies have investigated the association between maternal PCOS and other psychiatric and neurodevelopmental disorders in offspring. STUDY DESIGN, SIZE, DURATION This was a population-based cohort study in Finland including all live births between 1996 and 2014 (n = 1 105 997). After excluding births to mothers with symptoms similar to PCOS, a total of 1 097 753 births by 590 939 mothers remained. Children were followed up until 31 December 2018, i.e. up to the age of 22 years. PARTICIPANTS/MATERIALS, SETTING, METHODS National registries were used to link data of the included births and their mothers. Data from 24 682 (2.2%) children born to mothers with PCOS were compared with 1 073 071 (97.8%) children born to mothers without PCOS. Cox proportional hazards modeling was used to evaluate the hazard ratio (HR) and 95% CI for the risk of neuropsychiatric disorders in relation to maternal PCOS. Stratified analyses were performed to test the independent role of PCOS and the joint effects of PCOS with maternal obesity, perinatal problems, cesarean delivery, gestational diabetes and use of fertility treatment. The analysis was adjusted for maternal age, country of birth, marriage status at birth, smoking, parity, psychiatric disorders, prescription of psychotropic N05/N06 during pregnancy and systemic inflammatory diseases when applicable. MAIN RESULTS AND THE ROLE OF CHANCE A total of 105 409 (9.8%) children were diagnosed with a neurodevelopmental or psychiatric disorder. Firstly, maternal PCOS was associated with any psychiatric diagnosis (HR 1.32; 95% CI 1.27-1.38) in offspring. Particularly, the risk was increased for sleeping disorders (HR 1.46; 95% CI 1.27-1.67), attention-deficit/hyperactivity disorders and conduct disorders (HR 1.42; 95% CI 1.33-1.52), tic disorders (HR 1.42; 95% CI 1.21-1.68), intellectual disabilities (HR 1.41; 95% CI 1.24-1.60), autism spectrum disorder (HR 1.40; 95% CI 1.26-1.57), specific developmental disorders (HR 1.37; 95% CI 1.30-1.43), eating disorders (HR 1.36; 95% CI 1.15-1.61), anxiety disorders (HR 1.33; 95% CI 1.26-1.41), mood disorders (HR 1.27; 95% CI 1.18-1.35) and other behavioral and emotional disorders (ICD-10 F98, HR 1.49; 95% CI 1.39-1.59). In short, there was no significant difference between sexes. The results were robust when restricting the analyses to the first-born children or births to mothers without psychiatric diagnosis or purchase of psychotropic medication. Secondly, stratified analysis according to maternal BMI showed that the risk of any neuropsychiatric disorder was increased in offspring to normal-weight mothers with PCOS (HR 1.20; 95% CI 1.09-1.32), and markedly higher in those to severely obese mothers with PCOS (HR 2.11; 95% CI 1.76-2.53) compared to offspring to normal-weight mothers without PCOS. When excluding perinatal problems, mothers with PCOS were still associated with increased risks of any neuropsychiatric disorders in offspring (HR 1.28; 95% CI 1.22-1.34) compared to mothers without PCOS. However, an additional increase was observed for PCOS in combination with perinatal problems (HR 1.99; 95% CI 1.84-2.16). Likewise, excluding cases with maternal gestational diabetes (HR 1.30; 95% CI 1.25-1.36), cesarean delivery (HR 1.29; 95% CI 1.23-1.35) or fertility treatment (HR 1.31; 95% CI 1.25-1.36) did not eliminate the associations. LIMITATIONS, REASONS FOR CAUTION The register-based prevalence of PCOS was lower than previously reported, suggesting that this study may capture the most severe cases. To combine anovulatory infertility with PCOS diagnosis as PCOS exposure might introduce diagnostic bias. It was not feasible to distinguish between subtypes of PCOS. Furthermore, familial factors might confound the association between maternal PCOS and neuropsychiatric disorders in offspring. Maternal BMI was available for birth cohort 2004-2014 only and there was no information on gestational weight gain. WIDER IMPLICATIONS OF THE FINDINGS This study provides further evidence that maternal PCOS and/or anovulatory infertility, independently and jointly with maternal obesity, perinatal problems, gestational diabetes and cesarean delivery, implies a broad range of adverse effects on offspring neurodevelopment. These findings may potentially help in counseling and managing pregnancies. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the joint research funding of Shandong University and Karolinska Institute (SDU-KI-2019-08 to X.C and C.L.), THL Finnish Institute for Health and Welfare: Drug and pregnancy project [M.G.], the Swedish Research Council [2014-10171 to C.L.], the regional agreement on medical training and clinical research (ALF) between Stockholm County Council and Karolinska Institute Stockholm County Council [SLL20170292 to C.L.], the Swedish Brain Foundation [FO2018-0141 and FO2019-0201 to C.L.]. X.C. was supported by the China Scholarship Council during her training in Karolinska Institute. L.K. was supported by the China Scholarship Council for his PhD study in Karolinska Institute. The authors have no competing interests to disclose. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Xinxia Chen
- School of Nursing, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden.,Centre for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Linghua Kong
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden.,Centre for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Mika Gissler
- Finnish Institute for Health and Welfare, Helsinki, Oulu, Finland.,Division of Family Medicine, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden.,Research Centre for Child Psychiatry, University of Turku, Turku, Finland
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden.,Centre for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|