101
|
Zargar MH, Shafia S, Masoodi SR, Mahajan Q, Khan N, Ahmad R. Variations in the inhibin gene in Kashmiri women with primary ovarian insufficiency. HUM FERTIL 2018; 23:111-116. [PMID: 30340444 DOI: 10.1080/14647273.2018.1525502] [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: 10/28/2022]
Abstract
Inhibin is a glycoprotein produced by granulosa cells and its main function is the negative feedback control of follicle stimulating hormone (FSH) which has an important role in folliculogenesis. Mutation in the INHα gene leading to decreased bioactive inhibin has been associated with primary ovarian insufficiency (POI). The aim of this study was to investigate the role of variations in the INHα gene in increasing the susceptibility to POI in Kashmiri women. INHα c.769G > A mutation was analysed in 100 POI cases and 100 controls using PCR-RFLP and agarose gel electrophoresis. The INHα c.769G > A mutation was found in 10% of POI cases with 8% having heterozygous mutation and 2% having a homozygous mutation. The frequency of mutation in healthy controls was zero. Statistically, a very significant association was found between INHα c.769G > A mutation and the occurrence of POI (p = 0.0015). Moreover, the mutation was also significantly associated with high levels of FSH in POI patients (p < 0.0001). Given the significant association of INHα c.769G > A mutation with the increased FSH levels and POI in Kashmiri population, we suggest this mutation can be used to identify POI variants for screening of women susceptible to POI before the disease onset and can further facilitate putative therapy for such patients.
Collapse
Affiliation(s)
- Mahrukh Hameed Zargar
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Syed Shafia
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Shariq Rashid Masoodi
- Department of Endocrinology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Qurteeba Mahajan
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Nabeela Khan
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Rehana Ahmad
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, India
| |
Collapse
|
102
|
Szymańska K, Kałafut J, Rivero-Müller A. The gonadotropin system, lessons from animal models and clinical cases. ACTA ACUST UNITED AC 2018; 70:561-587. [PMID: 30264954 DOI: 10.23736/s0026-4784.18.04307-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review article centers upon family of gonadotropin hormones which consists of two pituitary hormones - follicle-stimulating hormone (FSH) and luteinizing hormone (LH) as well as one non-pituitary hormone - human chorionic gonadotropin (hCG) secreted by placenta, and their receptors. Gonadotropins play an essential role in proper sexual development, puberty, gametogenesis, maintenance of pregnancy and male sexual differentiation during the fetal development. They belong to the family of glycoprotein hormones thus they constitute heterodimeric proteins built of common α subunit and hormone-specific β-subunit. Hitherto, several mutations in genes encoding both gonadotropins and their receptors have been identified in humans. Their occurrence resulted in a number of different phenotypes including delayed puberty, primary amenorrhea, hermaphroditism, infertility and hypogonadism. In order to understand the effects of mutations on the phenotype observed in affected patients, detailed molecular studies are required to map the relationship between the structure and function of gonadotropins and their receptors. Nonetheless, in vitro assays are often insufficient to understand physiology. Therefore, several animal models have been developed to unravel the physiological roles of gonadotropins and their receptors.
Collapse
|
103
|
Huhtaniemi I. MECHANISMS IN ENDOCRINOLOGY: Hormonal regulation of spermatogenesis: mutant mice challenging old paradigms. Eur J Endocrinol 2018; 179:R143-R150. [PMID: 29959220 DOI: 10.1530/eje-18-0396] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/28/2018] [Indexed: 11/08/2022]
Abstract
The two pituitary gonadotrophins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), and in particular LH-stimulated high intratesticular testosterone (ITT) concentration, are considered crucial for spermatogenesis. We have revisited these concepts in genetically modified mice, one being the LH receptor (R)-knockout mouse (LuRKO), the other a transgenic mouse expressing in Sertoli cells a highly constitutively active mutated Fshr (Fshr-CAM). It was found that full spermatogenesis was induced by exogenous testosterone treatment in LuRKO mice at doses that restored ITT concentration to a level corresponding to the normal circulating testosterone level in WT mice, ≈5 nmol/L, which is 1.4% of the normal high ITT concentration. When hypogonadal LuRKO and Fshr-CAM mice were crossed, the double-mutant mice with strong FSH signaling, but minimal testosterone production, showed near-normal spermatogenesis, even when their residual androgen action was blocked with the strong antiandrogen flutamide. In conclusion, our findings challenge two dogmas of the hormonal regulation of male fertility: (1) high ITT concentration is not necessary for spermatogenesis and (2) strong FSH stimulation can maintain spermatogenesis without testosterone. These findings have clinical relevance for the development of hormonal male contraception and for the treatment of idiopathic oligozoospermia.
Collapse
Affiliation(s)
- Ilpo Huhtaniemi
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Campus, London, UK
- Department of Physiology, Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
104
|
Barbonetti A, Calogero AE, Balercia G, Garolla A, Krausz C, La Vignera S, Lombardo F, Jannini EA, Maggi M, Lenzi A, Foresta C, Ferlin A. The use of follicle stimulating hormone (FSH) for the treatment of the infertile man: position statement from the Italian Society of Andrology and Sexual Medicine (SIAMS). J Endocrinol Invest 2018; 41:1107-1122. [PMID: 29392544 DOI: 10.1007/s40618-018-0843-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/27/2018] [Indexed: 12/18/2022]
Affiliation(s)
- A Barbonetti
- Casa di Cura San Raffaele Sulmona, Sulmona, AQ, Italy
| | - A E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - G Balercia
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Umberto I Hospital, Ancona, Polytechnic University of Marche, Ancona, Italy
| | - A Garolla
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padua, Italy
| | - C Krausz
- Department of Biomedical, Experimental and Clinical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - S La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - F Lombardo
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, University of Rome "La Sapienza", Rome, Italy
| | - E A Jannini
- Department of Systems Medicine, Chair of Endocrinology and Medical Sexology (ENDOSEX), University of Rome Tor Vergata, Rome, Italy
| | - M Maggi
- Department of Biomedical, Experimental and Clinical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - A Lenzi
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, University of Rome "La Sapienza", Rome, Italy
| | - C Foresta
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padua, Italy
| | - A Ferlin
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padua, Italy.
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.
| |
Collapse
|
105
|
Abudureyimu A, Cai Y, Huo S, Ren R, Zang R, Yang J, Ma Z, Cui Y. Expression and localization of follicle-stimulating hormone receptor in the yak uterus during different stages of the oestrous cycle. Reprod Domest Anim 2018; 53:1539-1545. [PMID: 30120840 DOI: 10.1111/rda.13313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/26/2018] [Indexed: 11/30/2022]
Abstract
Morphological changes of the uterus and alterations in its secretory activity under the influence of steroid hormones been well documented. The oestrous cycle is also associated with significant changes in plasma follicle-stimulating hormone (FSH), whose effects are mediated through its receptor (FSHR). Reports showed that in many mammals, FSHR was expressed in gonadal and extragonadal tissues including cervix, female reproductive tract, and pituitary gland. Follicle-stimulating hormone (FSH) signals through endothelial FSHR directly stimulate angiogenesis and involved in the timing of birth in human, and the presence of FSHR in the placenta is essential for normal pregnancy in mice. But little is known about FSHR expression in the yak uterus. The main objective of the present study was to determine the expression and localization of FSHR in the yak uterus during different phases of the oestrous cycle. Results showed that FSHR protein was localized in the surface and glandular epithelial cells, stroma cells, myometrial smooth muscle cells and blood vessel endothelial cells. The expression of FSHR protein peaked at oestrus, significantly decreased at dioestrus (p < 0.05) and increased again at proestrus. FSHR mRNA was highly expressed at both proestrus and oestrus, and decreased at metestrus with the lowest values at dioestrus (p < 0.05). In conclusion, FSHR expression in the yak uterus changed with the stage of the oestrous cycle suggesting that FSHR plays an essential role in regulating the endometrial and myometrial functions during the oestrus cycle in the yak.
Collapse
Affiliation(s)
- Ayimuguli Abudureyimu
- Life Science and Engineering College of Northwest University for Nationalities, Lanzhou, China.,College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yong Cai
- Experimental Center of Northwest University for Nationalities, Lanzhou, China
| | - Shengdong Huo
- Life Science and Engineering College of Northwest University for Nationalities, Lanzhou, China
| | - Rui Ren
- Life Science and Engineering College of Northwest University for Nationalities, Lanzhou, China
| | - Rongxin Zang
- Life Science and Engineering College of Northwest University for Nationalities, Lanzhou, China
| | - Jutian Yang
- Life Science and Engineering College of Northwest University for Nationalities, Lanzhou, China
| | - Zhongren Ma
- Life Science and Engineering College of Northwest University for Nationalities, Lanzhou, China
| | - Yan Cui
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
106
|
Nataraja S, Sriraman V, Palmer S. Allosteric Regulation of the Follicle-Stimulating Hormone Receptor. Endocrinology 2018; 159:2704-2716. [PMID: 29800292 DOI: 10.1210/en.2018-00317] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 05/16/2018] [Indexed: 01/08/2023]
Abstract
Follicle-stimulating hormone receptor (FSHR) belongs to the leucine-rich repeat family of the G protein-coupled receptor (LGR), which includes the glycoprotein hormone receptors luteinizing hormone receptor, thyrotropin receptor, and other LGRs 4, 5, 6, and 7. FSH is the key regulator of folliculogenesis in females and spermatogenesis in males. FSH elicits its physiological response through its cognate receptor on the cell surface. Binding of the hormone FSH to its receptor FSHR brings about conformational changes in the receptor that are transduced through the transmembrane domain to the intracellular region, where the downstream effector interaction takes place, leading to activation of the downstream signaling cascade. Identification of small molecules that could activate or antagonize FSHR provided interesting tools to study the signal transduction mechanism of the receptor. However, because of the nature of the ligand-receptor interaction of FSH-FSHR, which contains multiple sites in the extracellular binding domain, most of the small-molecule modulators of FSHR are unable to bind to the orthosteric site of the receptors. Rather they modulate receptor activation through allosteric sites in the transmembrane region. This review will discuss allosteric modulation of FSHR primarily through the discovery of small-molecule modulators, focusing on current data on the status of development and the utility of these as tools to better understand signaling mechanisms.
Collapse
|
107
|
Grazul-Bilska AT, Reyaz A, Valkov V, Dorsam ST, Redmer DA. Follicle stimulating hormone receptor protein is expressed in ovine uterus during the estrous cycle and utero-placenta during early pregnancy: An immunohistochemical study. Acta Histochem 2018; 120:420-428. [PMID: 29754696 DOI: 10.1016/j.acthis.2018.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/08/2018] [Indexed: 12/26/2022]
Abstract
Follicle stimulating hormone (FSH) is a well characterized gonadotropin that controls primarily development and functions of ovarian follicles in mammalian species. FSH binds to a specific G protein-coupled receptor (FSHR) belonging to the glycoprotein hormone receptor family that plays an essential role in reproduction. Although the primary location of FSHR is in the gonads (mainly in ovarian follicles), FSHR protein and/or mRNA have also been detected in extragonadal female reproductive tissues including embryo, placenta, endometrium, cervix, ovarian cancer tissues, and/or endometriotic lesions in several species. To determine the pattern of FSHR expression in the uterus and placenta, uterine tissues were collected at the early, mid- and/or late luteal phases of the estrous cycle from non-treated or FSH-treated ewes, and utero-placental tissues were collected during early pregnancy followed by immunohistochemistry and image generation. FSHR was immunolocalized to several uterine and utero-placental compartments including luminal epithelium, endometrial glands and surrounding stroma, myometrium, and endothelium and vascular smooth muscle cells in endometrium, myometrium and mesometrium. Intensity of staining and distribution of FSHR in selected compartments differed and seems to depend on the stage of the estrous cycle or pregnancy, and FSH-treatment. These novel data demonstrate differential expression of FSHR protein indicating that FSH plays a specific role in regulation of uterine and utero-placenta functions in sheep.
Collapse
|
108
|
|
109
|
Barseghyan H, Délot EC, Vilain E. New technologies to uncover the molecular basis of disorders of sex development. Mol Cell Endocrinol 2018; 468:60-69. [PMID: 29655603 PMCID: PMC7249677 DOI: 10.1016/j.mce.2018.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/06/2018] [Accepted: 04/06/2018] [Indexed: 02/04/2023]
Abstract
The elegant developmental biology experiments conducted in the 1940s by French physiologist Alfred Jost demonstrated that the sexual phenotype of a mammalian embryo depended whether the embryonic gonad develops into a testis or not. In humans, anomalies in the processes that regulate development of chromosomal, gonadal or anatomic sex result in a spectrum of conditions termed Disorders/Differences of Sex Development (DSD). Each of these conditions is rare, and understanding of their genetic etiology is still incomplete. Historically, DSD diagnoses have been difficult to establish due to the lack of standardization of anatomical and endocrine phenotyping procedures as well as genetic testing. Yet, a definitive diagnosis is critical for optimal management of the medical and psychosocial challenges associated with DSD conditions. The advent in the clinical realm of next-generation sequencing methods, with constantly decreasing price and turnaround time, has revolutionized the diagnostic process. Here we review the successes and limitations of the genetic methods currently available for DSD diagnosis, including Sanger sequencing, karyotyping, exome sequencing and chromosomal microarrays. While exome sequencing provides higher diagnostic rates, many patients still remain undiagnosed. Newer approaches, such as whole-genome sequencing and whole-genome mapping, along with gene expression studies, have the potential to identify novel DSD-causing genes and significantly increase total diagnostic yield, hopefully shortening the patient's journey to an accurate diagnosis and enhancing health-related quality-of-life outcomes for patients and families.
Collapse
Affiliation(s)
- Hayk Barseghyan
- Center for Genetic Medicine Research, Children's National Health System, Children's Research Institute, Washington, DC, 20010, USA.
| | - Emmanuèle C Délot
- Center for Genetic Medicine Research, Children's National Health System, Children's Research Institute, Washington, DC, 20010, USA.
| | - Eric Vilain
- Center for Genetic Medicine Research, Children's National Health System, Children's Research Institute, Washington, DC, 20010, USA.
| |
Collapse
|
110
|
Huhtaniemi I, Hovatta O, La Marca A, Livera G, Monniaux D, Persani L, Heddar A, Jarzabek K, Laisk-Podar T, Salumets A, Tapanainen JS, Veitia RA, Visser JA, Wieacker P, Wolczynski S, Misrahi M. Advances in the Molecular Pathophysiology, Genetics, and Treatment of Primary Ovarian Insufficiency. Trends Endocrinol Metab 2018; 29:400-419. [PMID: 29706485 DOI: 10.1016/j.tem.2018.03.010] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/07/2018] [Accepted: 03/12/2018] [Indexed: 12/22/2022]
Abstract
Primary ovarian insufficiency (POI) affects ∼1% of women before 40 years of age. The recent leap in genetic knowledge obtained by next generation sequencing (NGS) together with animal models has further elucidated its molecular pathogenesis, identifying novel genes/pathways. Mutations of >60 genes emphasize high genetic heterogeneity. Genome-wide association studies have revealed a shared genetic background between POI and reproductive aging. NGS will provide a genetic diagnosis leading to genetic/therapeutic counseling: first, defects in meiosis or DNA repair genes may predispose to tumors; and second, specific gene defects may predict the risk of rapid loss of a persistent ovarian reserve, an important determinant in fertility preservation. Indeed, a recent innovative treatment of POI by in vitro activation of dormant follicles proved to be successful.
Collapse
Affiliation(s)
- Ilpo Huhtaniemi
- Institute of Reproductive and Developmental Biology, Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Outi Hovatta
- Karolinska Institute, Stockholm, Sweden, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Antonio La Marca
- Mother-Infant Department, University of Modena and Reggio Emilia, Modena 41100, Italy
| | - Gabriel Livera
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation: UMR 967, INSERM; CEA/DRF/iRCM/SCSR; Univ. Paris Diderot, Sorbonne Paris Cité; Univ. Paris-Sud, Université Paris-Saclay, Fontenay aux Roses, F-92265, France
| | - Danielle Monniaux
- UMR85 PRC, Physiology of Reproduction and Behavior, INRA, CNRS, IFCE, University of Tours, 37380 Nouzilly, France
| | - Luca Persani
- Department of Clinical Sciences & Community Health, University of Milan, Milan 20122, Division of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano, Milan 20149, Italy
| | - Abdelkader Heddar
- Medical Faculty, Univ. Paris Sud and Paris Saclay, Bicetre Hospital 94275, Le Kremlin Bicêtre, France
| | - Katarzyna Jarzabek
- Department of Biology and Pathology of Human Reproduction, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland
| | - Triin Laisk-Podar
- Women's Clinic, Institute of Clinical Medicine, University of Tartu, L. Puusepa 8, Tartu, Estonia; Competence Centre on Health Technologies, 50410, Estonia
| | - Andres Salumets
- Women's Clinic, Institute of Clinical Medicine, University of Tartu, L. Puusepa 8, Tartu, Estonia; Competence Centre on Health Technologies, 50410, Estonia
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University, Hospital, Helsinki 00029, Finland; Department of Obstetrics and Gynecology, University Hospital of Oulu, University of Oulu, Medical Research Center Oulu and PEDEGO Research Unit, P.O BOX 23, FI-90029 OYS, Oulu, Finland
| | - Reiner A Veitia
- Molecular Oncology and Ovarian Pathologies Université Paris-Diderot/Paris 7, Institut Jacques Monod, 15 Rue Hélène Brion, Paris Cedex 13, France
| | - Jenny A Visser
- Dept. of Internal Medicine, Erasmus University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Peter Wieacker
- Institute of Human Genetics, University Hospital of Münster, Vesaliusweg 12-14 D48149 Münster, Germany
| | - Slawomir Wolczynski
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Sklodowskiej 24A, 15-276 Bialystok, Poland
| | - Micheline Misrahi
- Medical Faculty, Univ. Paris Sud and Paris Saclay, Bicetre Hospital 94275, Le Kremlin Bicêtre, France.
| |
Collapse
|
111
|
|
112
|
Liang QX, Wang ZB, Lin F, Zhang CH, Sun HM, Zhou L, Zhou Q, Schatten H, Odile FC, Brigitte B, Sun QY, Qian WP. Ablation of beta subunit of protein kinase CK2 in mouse oocytes causes follicle atresia and premature ovarian failure. Cell Death Dis 2018; 9:508. [PMID: 29725001 PMCID: PMC5938699 DOI: 10.1038/s41419-018-0505-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 03/12/2018] [Accepted: 03/14/2018] [Indexed: 12/31/2022]
Abstract
Premature ovarian failure (POF), a major cause of female infertility, is a complex disorder, but the molecular mechanisms underlying the disorder are only poorly understood. Here we report that protein kinase CK2 contributes to maintaining follicular survival through PI3K/AKT pathway and DNA damage response pathway. Targeted deletion of CK2β in mouse oocytes from the primordial follicle stage resulted in female infertility, which was attributed to POF incurring by massive follicle atresia. Downregulated PI3K/AKT signaling was found after CK2β deletion, indicated by reduced level of phosphorylated AKT (S473, T308, and S129) and altered AKT targets related to cell survival. Further studies discovered that CK2β-deficient oocytes showed enhanced γH2AX signals, indicative of accumulative unrepaired DSBs, which activated CHK2-dependant p53 and p63 signaling. The suppressed PI3K/AKT signaling and failed DNA damage response signaling probably contribute to large-scale oocyte loss and eventually POF. Our findings provide important new clues for elucidating the mechanisms underlying follicle atresia and POF.
Collapse
Affiliation(s)
- Qiu-Xia Liang
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, Guangdong, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.,University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Fei Lin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.,Center for Reproductive Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Chun-Hui Zhang
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, Guangdong, China
| | - Hong-Mei Sun
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, Guangdong, China
| | - Liang Zhou
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, Guangdong, China
| | - Qian Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.,University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Filhol-Cochet Odile
- INSERM U1036, Institute de Recherches en Technologies et Sciences pour le Vivant/Biologie du Cancer et de l'Infection, Commissariat à l'Énergie Atomique et aux Énerigies Alternatives Grenoble, Grenoble, France
| | | | - Qing-Yuan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China. .,University of Chinese Academy of Sciences, 100101, Beijing, China.
| | - Wei-Ping Qian
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, 518036, Shenzhen, Guangdong, China.
| |
Collapse
|
113
|
Santi D, Potì F, Simoni M, Casarini L. Pharmacogenetics of G-protein-coupled receptors variants: FSH receptor and infertility treatment. Best Pract Res Clin Endocrinol Metab 2018; 32:189-200. [PMID: 29678285 DOI: 10.1016/j.beem.2018.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Infertility treatment may represent a paradigmatic example of precision medicine. Follicle-stimulating hormone (FSH) has been proposed as a valuable therapeutic option both in males and in females, even if a standardized approach is far to be established. To date, several genetic mutations as well as polymorphisms have been demonstrated to significantly affect the pathophysiology of FSH-FSH receptor (FSHR) interaction, although the underlying molecular mechanisms remain unclear. This review aims to highlight possible aspects of FSH therapy that could benefit from a pharmacogenetic approach, providing an up-to-date overview of the variability of the response to FSH treatment in both sexes. Specific sections are dedicated to the clinical use of FSH in infertility and how FSHR polymorphisms may affect the therapeutic endpoints.
Collapse
Affiliation(s)
- Daniele Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy; Department of Medicine, Endocrinology, Metabolism and Geriatrics, Azienda Ospedaliero-Universitaria of Modena, Italy.
| | - Francesco Potì
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy; Department of Medicine, Endocrinology, Metabolism and Geriatrics, Azienda Ospedaliero-Universitaria of Modena, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy; Department of Medicine, Endocrinology, Metabolism and Geriatrics, Azienda Ospedaliero-Universitaria of Modena, Italy
| |
Collapse
|
114
|
Current approaches for the treatment of premature ovarian failure with stem cell therapy. Biomed Pharmacother 2018; 102:254-262. [PMID: 29567538 DOI: 10.1016/j.biopha.2018.03.056] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 03/10/2018] [Accepted: 03/11/2018] [Indexed: 02/08/2023] Open
Abstract
One of the common disorders found in women is premature ovarian failure (POF). Recently some studies have explained premature ovarian insufficiency (POI). The causes of it are unknown although various types of study have been done. The most common causes such as genetic and autoimmune conditions can have a role in POF and can lead to infertility. Some characterization of POF are hypo-oestrogenism (estrogen deficiency), increased gonadotropin level and most importantly amenorrhea. The main purpose of this review is to describe the cause and treatment of POF, especially stem cell therapy proposed in previous studies. Stem cells have self-renewal and regeneration potential, hence they can be very effective in the treatment of ovarian failure and consequently infertility. There are several kinds of stem cells such as, mesenchymal stem cells (MSCs), stem cells from extra-embryonic tissues, induced pluripotent stem cells (iPSCs), and ovarian stem cells that are used in POF stem cell therapy as observed in previous studies. This article reviews the latest studies on POF to summarize current understanding and future directions.
Collapse
|
115
|
Kanj RV, Ofei-Tenkorang NA, Altaye M, Gordon CM. Evaluation and Management of Primary Ovarian Insufficiency in Adolescents and Young Adults. J Pediatr Adolesc Gynecol 2018; 31:13-18. [PMID: 28782660 DOI: 10.1016/j.jpag.2017.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/23/2017] [Accepted: 07/31/2017] [Indexed: 01/24/2023]
Abstract
STUDY OBJECTIVE To identify clinical features associated with primary ovarian insufficiency (POI) and collect data on the evaluation and treatment received. DESIGN Retrospective chart review. Data were abstracted on etiology of POI, history, laboratory evaluation, imaging results, return for clinical care, and treatment plans. SETTING Urban children's hospital in Cincinnati, Ohio. PARTICIPANTS Fifty female patients, age 11-26 years, with initial presentation of POI between January 1, 2006 and December 31, 2015. MAIN OUTCOME MEASURES Etiology of POI, bone mineral density (BMD), laboratory evaluation, and services utilized at presentation. RESULTS Three hundred thirty-one charts were reviewed, 71 with confirmed diagnosis of POI, and 50 with sufficient data for inclusion. Among the 50, 21 (42%) had Turner syndrome, 18 (36%) remained idiopathic, and 11 (22%) had another condition (eg, autoimmune polyglandular syndrome, galactosemia, etc). Thirty-six (72%) were karyotyped; in 14 (28%), 21-hydroxylase antibodies were measured; 32 (64%) underwent dual-energy x-ray absorptiometry BMD measures of lumbar spine. Eight of 50 patients (16%) reported fracture. Of these, at presentation, 4 (50%) had low BMD, and 2 (25%) had slightly low BMD. On initial spinal dual-energy x-ray absorptiometry, 9 of 32 (28%) had low BMD (Z-score ≤ -2.0) and 7 of 32 (22%) were slightly low (-1.0 to -1.9). All started estrogen therapy within 2 years of presentation. In follow-up, only 2 patients (4%) saw a mental health consultant for emotional support. CONCLUSION POI is a model of estrogen deficiency with most cases due to Turner syndrome or idiopathic causes. At presentation, many had low BMD and few were seen for psychological support as part of multidisciplinary care.
Collapse
Affiliation(s)
- Rula V Kanj
- Division of Pediatric and Adolescent Gynecology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Nana Ama Ofei-Tenkorang
- Division of Adolescent and Transition Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Mekibib Altaye
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Catherine M Gordon
- Division of Adolescent and Transition Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| |
Collapse
|
116
|
Leutinizing hormone/choriogonadotropin receptor and follicle stimulating hormone receptor gene variants and risk of recurrent pregnancy loss: A case control study. Meta Gene 2018. [DOI: 10.1016/j.mgene.2017.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
117
|
Ren Y, Diao F, Katari S, Yatsenko S, Jiang H, Wood-Trageser MA, Rajkovic A. Functional study of a novel missense single-nucleotide variant of NUP107 in two daughters of Mexican origin with premature ovarian insufficiency. Mol Genet Genomic Med 2018; 6:276-281. [PMID: 29363275 PMCID: PMC5902394 DOI: 10.1002/mgg3.345] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/06/2017] [Accepted: 10/24/2017] [Indexed: 11/24/2022] Open
Abstract
Background Hypergonadotropic hypogonadism (HH) is a genetically heterogeneous disorder that usually presents with amenorrhea, atrophic ovaries, and low estrogen. Most cases of HH are idiopathic and nonsyndromic. Nucleoporin 107 (NUP107), a protein involved in transport between cytoplasm and nucleus with putative roles in meiosis/mitosis progression, was recently implicated as a cause of HH. We identified a NUP107 genetic variant in a nonconsanguineous family with two sisters affected with primary amenorrhea and HH, and generated a mouse model that carried the human variant. Methods We performed a high‐resolution X‐chromosome microarray and whole exome sequencing on parents and two sisters with HH to identify pathogenic variants. We generated a mouse model of candidate NUP107 variant using CRISPR/Cas9. Results Whole exome sequencing identified a novel and rare missense variant in the NUP107 gene (c.1063C>T, p.R355C) in both sisters with HH. In order to determine functional significance of this variant, we used CRISPR/Cas9 to introduce the human variant into the mouse genome. Mice with the homolog of the R355C variant, as well as the nine base pairs deletion in Nup107 had female subfertility. Conclusions Our findings indicate that NUP107 R355C variant falls in the category of variant of unknown significance as the cause of HH and infertility.
Collapse
Affiliation(s)
- Yu Ren
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Feiyang Diao
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Sunita Katari
- Division of Reproductive Endocrinology and Infertility, Magee-Womens Hospital of UPMC, Pittsburgh, PA, USA
| | - Svetlana Yatsenko
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Huaiyang Jiang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Aleksandar Rajkovic
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
118
|
Laissue P. The molecular complexity of primary ovarian insufficiency aetiology and the use of massively parallel sequencing. Mol Cell Endocrinol 2018; 460:170-180. [PMID: 28743519 DOI: 10.1016/j.mce.2017.07.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 07/21/2017] [Accepted: 07/22/2017] [Indexed: 11/28/2022]
Abstract
Primary ovarian insufficiency (POI) is a frequently occurring pathology, leading to infertility. Genetic anomalies have been described in POI and mutations in numerous genes have been definitively related to the pathogenesis of the disease. Some studies based on next generation sequencing (NGS) have been successfully undertaken as they have led to identify new mutations associated with POI aetiology. The purpose of this review is to present the most relevant molecules involved in diverse complex pathways, which may contribute towards POI. The main genes participating in bipotential gonad formation, sex determination, meiosis, folliculogenesis and ovulation are described to enable understanding how they may be considered putative candidates involved in POI. Considerations regarding NGS technical aspects such as design and data interpretation are mentioned. Successful NGS initiatives used for POI studying and future challenges are also discussed.
Collapse
Affiliation(s)
- Paul Laissue
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
119
|
Sayers N, Hanyaloglu AC. Intracellular Follicle-Stimulating Hormone Receptor Trafficking and Signaling. Front Endocrinol (Lausanne) 2018; 9:653. [PMID: 30450081 PMCID: PMC6225286 DOI: 10.3389/fendo.2018.00653] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 10/17/2018] [Indexed: 12/18/2022] Open
Abstract
Models of G protein-coupled receptor (GPCR) signaling have dramatically altered over the past two decades. Indeed, GPCRs such as the follicle-stimulating hormone receptor (FSHR) have contributed to these new emerging models. We now understand that receptor signaling is highly organized at a spatial level, whereby signaling not only occurs from the plasma membrane but distinct intracellular compartments. Recent studies in the role of membrane trafficking and spatial organization of GPCR signaling in regulating gonadotropin hormone receptor activity has identified novel intracellular compartments, which are tightly linked with receptor signaling and reciprocally regulated by the cellular trafficking machinery. Understanding the impact of these cell biological mechanisms to physiology and pathophysiology is emerging for certain GPCRs. However, for FSHR, the potential impact in both health and disease and the therapeutic possibilities of these newly identified systems is currently unknown, but offers the potential to reassess prior strategies, or unveil novel opportunities, in targeting this receptor.
Collapse
|
120
|
Ulloa-Aguirre A, Zariñán T, Jardón-Valadez E, Gutiérrez-Sagal R, Dias JA. Structure-Function Relationships of the Follicle-Stimulating Hormone Receptor. Front Endocrinol (Lausanne) 2018; 9:707. [PMID: 30555414 PMCID: PMC6281744 DOI: 10.3389/fendo.2018.00707] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/09/2018] [Indexed: 12/16/2022] Open
Abstract
The follicle-stimulating hormone receptor (FSHR) plays a crucial role in reproduction. This structurally complex receptor is a member of the G-protein coupled receptor (GPCR) superfamily of membrane receptors. As with the other structurally similar glycoprotein hormone receptors (the thyroid-stimulating hormone and luteinizing hormone-chorionic gonadotropin hormone receptors), the FSHR is characterized by an extensive extracellular domain, where binding to FSH occurs, linked to the signal specificity subdomain or hinge region. This region is involved in ligand-stimulated receptor activation whereas the seven transmembrane domain is associated with receptor activation and transmission of the activation process to the intracellular loops comprised of amino acid sequences, which predicate coupling to effectors, interaction with adapter proteins, and triggering of downstream intracellular signaling. In this review, we describe the most important structural features of the FSHR intimately involved in regulation of FSHR function, including trafficking, dimerization, and oligomerization, ligand binding, agonist-stimulated activation, and signal transduction.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- *Correspondence: Alfredo Ulloa-Aguirre
| | - Teresa Zariñán
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Eduardo Jardón-Valadez
- Departamento de Ciencias Ambientales, Universidad Autónoma Metropolitana Unidad Lerma, Lerma, Mexico
| | - Rubén Gutiérrez-Sagal
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - James A. Dias
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, United States
| |
Collapse
|
121
|
Szymańska K, Kałafut J, Przybyszewska A, Paziewska B, Adamczuk G, Kiełbus M, Rivero-Müller A. FSHR Trans-Activation and Oligomerization. Front Endocrinol (Lausanne) 2018; 9:760. [PMID: 30619090 PMCID: PMC6301190 DOI: 10.3389/fendo.2018.00760] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/30/2018] [Indexed: 12/12/2022] Open
Abstract
Follicle stimulating hormone (FSH) plays a key role in human reproduction through, among others, induction of spermatogenesis in men and production of estrogen in women. The function FSH is performed upon binding to its cognate receptor-follicle-stimulating hormone receptor (FSHR) expressed on the surface of target cells (granulosa and Sertoli cells). FSHR belongs to the family of G protein-coupled receptors (GPCRs), a family of receptors distinguished by the presence of various signaling pathway activation as well as formation of cross-talking aggregates. Until recently, it was claimed that the FSHR occurred naturally as a monomer, however, the crystal structure as well as experimental evidence have shown that FSHR both self-associates and forms heterodimers with the luteinizing hormone/chorionic gonadotropin receptor-LHCGR. The tremendous gain of knowledge is also visible on the subject of receptor activation. It was once thought that activation occurs only as a result of ligand binding to a particular receptor, however there is mounting evidence of trans-activation as well as biased signaling between GPCRs. Herein, we describe the mechanisms of aforementioned phenomena as well as briefly describe important experiments that contributed to their better understanding.
Collapse
Affiliation(s)
- Kamila Szymańska
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Joanna Kałafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Alicja Przybyszewska
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Beata Paziewska
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Grzegorz Adamczuk
- Independent Medical Biology Unit, Medical University of Lublin, Lublin, Poland
| | - Michał Kiełbus
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Adolfo Rivero-Müller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- *Correspondence: Adolfo Rivero-Müller ;
| |
Collapse
|
122
|
Follicle-Stimulating Hormone Receptor: Advances and Remaining Challenges. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:1-58. [DOI: 10.1016/bs.ircmb.2018.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
123
|
Sekar N, Kulkarni R, Ozalkar S, Prabhu YD, Renu K, Ramgir SS, Abilash V. Association of Exon 10A and 10B inactivating mutation of follicle stimulating hormone receptor gene (FSHR) and Polycystic Ovarian Syndrome in Vellore cohort. ACTA ACUST UNITED AC 2017. [DOI: 10.1088/1757-899x/263/2/022036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
124
|
Torrealday S, Kodaman P, Pal L. Premature Ovarian Insufficiency - an update on recent advances in understanding and management. F1000Res 2017; 6:2069. [PMID: 29225794 PMCID: PMC5710309 DOI: 10.12688/f1000research.11948.1] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/23/2017] [Indexed: 12/11/2022] Open
Abstract
Premature ovarian insufficiency is a complex and relatively poorly understood entity with a myriad of etiologies and multisystem sequelae that stem from premature deprivation of ovarian sex hormones. Timely diagnosis with a clear understanding of the various comorbidities that can arise from estrogen deficiency is vital to appropriately counsel and treat these patients. Prompt initiation of hormone therapy is critical to control the unsolicited menopausal symptoms that many women experience and to prevent long-term health complications. Despite ongoing efforts at improving our understanding of the mechanisms involved, any advancement in the field in recent decades has been modest at best and researchers remain thwarted by the complexity and heterogeneity of the underpinnings of this entity. In contrast, the practice of clinical medicine has made meaningful strides in providing assurance to the women with premature ovarian insufficiency that their quality of life as well as long-term health can be optimized through timely intervention. Ongoing research is clearly needed to allow pre-emptive identification of the at-risk population and to identify mechanisms that if addressed in a timely manner, can prolong ovarian function and physiology.
Collapse
Affiliation(s)
- Saioa Torrealday
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics & Gynecology, Womack Army Medical Center, Fort Bragg, North Carolina, USA
| | - Pinar Kodaman
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lubna Pal
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
125
|
Guzel Y, Aba YA, Yakin K, Oktem O. Menstrual cycle characteristics of young females with occult primary ovarian insufficiency at initial diagnosis and one-year follow-up with serum amh level and antral follicle count. PLoS One 2017; 12:e0188334. [PMID: 29176793 PMCID: PMC5703527 DOI: 10.1371/journal.pone.0188334] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/04/2017] [Indexed: 11/29/2022] Open
Abstract
Occult primary ovarian insufficiency (also known as incipient ovarian failure or diminished ovarian reserve) is defined as serum AMH level ≤1.1ng/mL in women under age 30. Limited data is available regarding the prevalence of occult POI, the preceding menstrual characteristics and its natural course in otherwise healthy young females. We aimed in this prospective observational study to determine the prevalence of occult POI in young females (< age 30) screened with serum AMH measurement; and analyze the patterns of change in their menstruation at initial assessment and one-year follow-up in relation to the changes in ovarian reserve quantitatively assessed with AMH and AFC. 963 young female college students under age 30 voluntarily participated in this study. 43 of them (4.4%) were diagnosed with occult POI as their AMH levels were ≤ 1.1ng/mL. Thirty-eight (83.4%) of them have regular cycles and denied any menstrual irregularity in the last 12 months. This rate was not statistically different from 7.3% of those with AMH>1.1ng/mL who reported at least one abnormal menstrual cycle in the last year (p = 0.36). Cycle length was significantly shorter in females with AMH ≤ 1.1ng/mL compared to those with AMH>1.1ng/mL (25.1±3.2 vs. 31.2±2.8 respectively, p<0.001). Karyotype, FMR-1 mutation analyses and auto-antibody screening returned normal in all. At one-year follow-up AMH, AFC and mean cycle length were further reduced compared to their values at initial assessment. Now, a greater proportion of the participants with occult POI were menstruating regularly at every 21 days compared to the initial evaluation one year ago (39.5% vs. 13.9% respectively, p = 0.013). Twenty-five underwent oocyte cryopreservation. These findings underscore the importance of screening young females with AMH for possible occult POI. It also emphasizes that young females with critically diminished ovarian reserve may continue to menstruate regularly without any characteristic menstrual abnormality other than shortening of cycle length.
Collapse
Affiliation(s)
- Yilmaz Guzel
- Istanbul Aydin University, Department of Obstetrics and Gynecology, Istanbul, Turkey
| | - Yilda Arzu Aba
- Istanbul Aydin University, School of Health Sciences, Istanbul, Turkey
| | - Kayhan Yakin
- American Hospital Women’s Health Center Assisted Reproduction Unit, Istanbul, Turkey
- Koc University School of Medicine, Department of Obstetrics and Gynecology, the Division Reproductive Endocrinology and Infertility, Istanbul, Turkey
| | - Ozgur Oktem
- American Hospital Women’s Health Center Assisted Reproduction Unit, Istanbul, Turkey
- Koc University School of Medicine, Department of Obstetrics and Gynecology, the Division Reproductive Endocrinology and Infertility, Istanbul, Turkey
- * E-mail:
| |
Collapse
|
126
|
A novel homozygous mutation in the FSHR gene is causative for primary ovarian insufficiency. Fertil Steril 2017; 108:1050-1055.e2. [PMID: 29157895 DOI: 10.1016/j.fertnstert.2017.09.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/21/2017] [Accepted: 09/07/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To identify the potential FSHR mutation in a Chinese woman with primary ovarian insufficiency (POI). DESIGN Genetic and functional studies. SETTING University-based reproductive medicine center. PATIENT(S) A POI patient, her family members, and another 192 control women with regular menstruation. INTERVENTION(S) Ovarian biopsy was performed in the patient. Sanger sequencing was carried out for the patient, her sister, and parents. The novel variant identified was further confirmed with the use of control subjects. MAIN OUTCOME MEASURE(S) Sanger sequencing and genotype analysis to identify the potential variant of the FSHR gene; hematoxylin and eosin staining of the ovarian section to observe the follicular development; Western blotting and immunofluorescence to detect FSH receptor (FSHR) expression; and cyclic adenosine monophosphate (cAMP) assay to monitor FSH-induced signaling. RESULT(S) Histologic examination of the ovaries in the patient revealed follicular development up to the early antral stage. Mutational screening and genotype analysis of the FSHR gene identified a novel homozygous mutation c.175C>T (p.R59X) in exon 2, which was inherited in the autosomal recessive mode from her heterozygous parents but was absent in her sister and the 192 control women. Functional studies demonstrated that in vitro the nonsense mutation caused the loss of full-length FSHR expression and that p.R59X mutant showed no response to FSH stimulation in the cAMP level. CONCLUSION(S) The mutation p.R59X in FSHR is causative for POI by means of arresting folliculogenesis.
Collapse
|
127
|
Ghezelayagh Z, Totonchi M, Zarei-Moradi S, Asadpour O, Maroufizadeh S, Eftekhari-Yazdi P, Gourabi H, Mohseni-Meybodi A. The Impact of Genetic Variation and Gene Expression Level of The Follicle-Stimulating Hormone Receptor on Ovarian Reserve. CELL JOURNAL 2017; 19:620-626. [PMID: 29105397 PMCID: PMC5672101 DOI: 10.22074/cellj.2018.4183] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 09/15/2016] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Ovarian reserve is defined as the capacity of the ovary to provide fertile oocytes. Diminished ovarian reserve (DOR) is a disorder in which ovaries are prone to go through early menopause. Where this loss of function occurs before the age of 40, it results in the premature ovarian failure (POF) disease. Throughout folliculogenesis, the follicle-stimulating hormone receptor (FSHR) starts a signaling cascade in the granulosa cells where its inactivation leads to the arrest of follicle maturation and therefore adversely affects ovarian reserve. The aim of this study was to investigate the association of genetic variation (polymorphisms and inactivating mutations) of FSHR with POF and DOR. MATERIALS AND METHODS This case-control study comprised 84 POF, 52 DOR and 80 fertile Iranian women. To determine the presence of the 566C>T mutation and the -29G>A polymorphism in FSHR, PCR-RFLP method was used. SSCP-sequencing was used to identify any allelic variants in exon 10. The expression of human FSHR at the transcript level was also compared between DOR and fertile controls by real time-polymerase chain reaction (PCR). RESULTS The 566C>T polymorphism was normal in all the cases. All genotypes of -29G>A and 919G>A (exon 10) polymorphisms were observed. Statistically significant differences were seen in the genotypic distribution of both polymorphisms when comparing the control group with the DOR patient group. A decrease was observed in FSHR expression of DOR patients compared with the control group but was not significant. CONCLUSIONS We conclude that the -29G>A and 919G>A polymorphisms in FSHR may be associated with DOR. Although these polymorphisms had significant differences at the genic level, no significant variation was found at the transcript level.
Collapse
Affiliation(s)
- Zeinab Ghezelayagh
- University of Science and Culture, Faculty of Basic Sciences and Advanced Technologies in Biology, ACECR, Tehran, Iran.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mehdi Totonchi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Shabnam Zarei-Moradi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Ommolbanin Asadpour
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Saman Maroufizadeh
- Department of Epidemiology and Reproductive Health, Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Poopak Eftekhari-Yazdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Hamid Gourabi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Anahita Mohseni-Meybodi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
128
|
Christofolini DM, Cordts EB, Santos-Pinheiro F, Kayaki EA, Dornas MCF, Santos MDC, Bianco B, Barbosa CP. How polymorphic markers contribute to genetic diseases in different populations? The study of inhibin A for premature ovarian insufficiency. EINSTEIN-SAO PAULO 2017; 15:269-272. [PMID: 29091146 PMCID: PMC5823038 DOI: 10.1590/s1679-45082017ao4052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/20/2017] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE To verify the incidence of the G679A mutation in exon 2 of the gene inhibin alpha (INHA), in women with secondary amenorrhea and diagnosis of premature ovarian insufficiency, and in controls. METHODS A 5mL sample of peripheral blood was collected from all study participants in an EDTA tube and was used for DNA extraction. For the patient group, 5mL of blood were also collected in a tube containing heparin for karyotype, and 5mL were collected in a dry tube for follicle stimulant hormone dosage. All patient and control samples were initially submitted to analysis of the G679A variant in exon 2 of the INHA gene by PCR-RFLP technique. Samples from patients with premature ovarian insufficiency after PCR-RFLP were submitted to Sanger sequencing of the encoding exons 2 and 3. Sequencing was performed on ABI 3500 GeneticAnalyzer equipment and the results were evaluated by SeqA and Variant Reporter software. RESULTS Samples of 70 women with premature ovarian insufficiency and 97 fertile controls were evaluated. The G769A variant was found in only one patient in the Premature Ovarian Insufficiency Group and in no control, and it appears to be rare in Brazilian patients with premature ovarian insufficiency. This polymorphism was previously associated to premature ovarian insufficiency in several populations worldwide. CONCLUSION There is genetic heterogeneity regarding the INHA gene in different populations, and among the causes of premature ovarian insufficiency.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bianca Bianco
- Faculdade de Medicina do ABC, Santo André, SP, Brazil
| | | |
Collapse
|
129
|
Papadimitriou K, Kountourakis P, Kottorou AE, Antonacopoulou AG, Rolfo C, Peeters M, Kalofonos HP. Follicle-Stimulating Hormone Receptor (FSHR): A Promising Tool in Oncology? Mol Diagn Ther 2017; 20:523-530. [PMID: 27392476 DOI: 10.1007/s40291-016-0218-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The cellular pathway of follicle-stimulating hormone (FSH) and its receptor (FSHR) is typically involved in reproduction in mammals. In humans, the FSHR is normally found in cells of the testis and the ovary, while it is scarcely expressed in other normal tissues. The expression of FSH/FSHR is studied in prostate, thyroid, and ovarian cancer tissues. Recently, the expression of FSHR was uniformly documented in malignant vascular endothelial cells from different tumor types, while in normal or inflammatory tissues its expression was scarce, suggesting a potential role of a pan-receptor in cancer. Subsequent studies have attempted to verify this unique specificity of this molecule and further define its features in malignant microenvironments but have had conflicting results, mostly because of differing techniques and immaturity of antibodies. Still, the lack of FSHR expression in most non-cancerous cells, in contrast to its specific correlation with the malignant tissue microenvironment, implies a potential role as both a diagnostic and a therapeutic tool. FSHR might also have a very specific role in malignancies, such as angiogenic and/or growth factor malignancies, but this is yet to be validated. Moreover, the expression of FSHR in endothelial malignant cells could have a predictive impact on disease progression, especially in relation to therapies targeting the tumor vasculature. In this review we look deep into the physiology of the FSH/FSHR pathway and evaluate the potential of FSHR as a predictive and prognostic tool in oncology.
Collapse
Affiliation(s)
| | | | | | | | - Christian Rolfo
- Department of Medical Oncology, University Hospital of Antwerp, 10 Wilrijksraat, 2650, Edegem, Belgium
| | - Marc Peeters
- Department of Medical Oncology, University Hospital of Antwerp, 10 Wilrijksraat, 2650, Edegem, Belgium
| | | |
Collapse
|
130
|
Trofimova T, Lizneva D, Suturina L, Walker W, Chen YH, Azziz R, Layman LC. Genetic basis of eugonadal and hypogonadal female reproductive disorders. Best Pract Res Clin Obstet Gynaecol 2017; 44:3-14. [DOI: 10.1016/j.bpobgyn.2017.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/20/2017] [Accepted: 05/02/2017] [Indexed: 12/21/2022]
|
131
|
Wang B, Li L, Zhu Y, Zhang W, Wang X, Chen B, Li T, Pan H, Wang J, Kee K, Cao Y. Sequence variants of KHDRBS1 as high penetrance susceptibility risks for primary ovarian insufficiency by mis-regulating mRNA alternative splicing. Hum Reprod 2017; 32:2138-2146. [DOI: 10.1093/humrep/dex263] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/03/2017] [Indexed: 11/12/2022] Open
|
132
|
Clinical efficacy of recombinant versus highly purified follicle-stimulating hormone according to follicle-stimulating hormone receptor genotype. Pharmacogenet Genomics 2017; 26:288-93. [PMID: 26959715 DOI: 10.1097/fpc.0000000000000215] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Conflicting data have been reported on the comparative doses of recombinant follicle-stimulating hormone (rFSH) and urinary highly purified follicle-stimulating hormone (HP-FSH) required for ovarian stimulation. Nothing is known about the clinical efficacy of rFSH or HP-FSH depending on the N680S follicle-stimulating hormone receptor (FSHR) polymorphism. Our aim was to investigate whether the N680S polymorphism of the FSHR gene affects ovarian response with different forms of FSH. MATERIALS AND METHODS This retrospective cohort study includes 382 cycles performed at Instituto Bernabeu from 191 oocyte donors. All donors carried out two cycles: one with rFSH and the other one with HP-FSH (group 1, n=63), both with HP-FSH (group 2, n=100) or both with rFSH (group 3, n=28). The results were compared by pairs from each patient. The main outcomes were oocyte yield, metaphase II matured oocytes (MII), days of stimulation, and gonadotropin dosage. RESULTS No significant differences were found when we compared the cycles for donors in group 1. However, according to the FSHR polymorphism, statistical differences were shown. For the SS genotype, more oocytes (16.9 vs. 18.4) and MII (12.8 vs. 15.5) were yielded in the HP-FSH cycle. For the NS genotype, more oocyte (20.1 vs. 16.9) and MII (17.4 vs. 14.2) were yielded in the rFSH cycle. For the NN genotype, no differences were found. No differences were found when we compared the cycles in groups 2 and 3 irrespective of the FSHR polymorphism. CONCLUSION For the first time, we have shown in a population of egg donors that the N680S FSHR gene polymorphism affects the efficacy of HP-FSH or rFSH. The FSHR genotype is an important factor to determine the dosage and the nature of the gonadotropin selected for ovarian stimulation.
Collapse
|
133
|
Xie M, Li M, Zhou J, Ding X, Shao Y, Jing J, Liu Y, Yao B. Brain-derived neurotrophic factor promotes human granulosa-like tumor cell steroidogenesis and proliferation by activating the FSH receptor-mediated signaling pathway. Sci Rep 2017; 7:180. [PMID: 28282971 PMCID: PMC5428030 DOI: 10.1038/s41598-017-00203-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/14/2017] [Indexed: 01/08/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and FSH receptor (FSHR) are expressed in ovarian granulosa cells, and play important roles in regulating follicle growth and oocyte maturation. Studies have linked the BDNF-associated signaling pathway to FSHR mRNA expression in the regulation of follicle development, but the mechanisms remain unknown. In the current study, we found that BDNF stimulated the secretion of estradiol and progesterone, and increased the proliferation of KGN cells (human granulosa-like tumor cell line). BDNF treatment also increased phosphorylated and ubiquitinated FSHR, and activated cAMP/PKA/CREB signaling pathway. Moreover, inhibition of BDNF expression by siRNA markedly reduced the estradiol secretion and down-regulated FSHR, aromatase and phosphorylated CREB; meanwhile, FSH treatment partly alleviated the effects of BDNF siRNA on KGN cells. These findings suggested that BDNF modulates graunlosa cell functions and the action probably mediated by FSHR-coupled signaling pathway, to affect aromatase-mediated steroidogenesis. These results provide an alternative target to optimize ovarian granulosa cell function.
Collapse
Affiliation(s)
- Min Xie
- Center of Reproductive Medicine, Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Meiling Li
- Center of Reproductive Medicine, Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Ji Zhou
- Center of Reproductive Medicine, Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Xiaomeng Ding
- Center of Reproductive Medicine, Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Yidan Shao
- Center of Reproductive Medicine, Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Jun Jing
- Center of Reproductive Medicine, Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Yuxiu Liu
- Department of Medical Statistics, Jinling Hospital, Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Bing Yao
- Center of Reproductive Medicine, Jinling Hospital, Clinical School of Medical College, Nanjing University, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
134
|
Kawashima I, Kawamura K. Disorganization of the germ cell pool leads to primary ovarian insufficiency. Reproduction 2017; 153:R205-R213. [PMID: 28289071 DOI: 10.1530/rep-17-0015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 02/10/2017] [Accepted: 03/13/2017] [Indexed: 01/26/2023]
Abstract
The mammalian ovary is an organ that controls female germ cell development, storing them and releasing mature oocytes for transporting to the oviduct. During the fetal stage, female germ cells change from a proliferative state to meiosis before forming follicles with the potential for the growth of surrounding somatic cells. Understanding of molecular and physiological bases of germ cell development in the fetal ovary contributed not only to the elucidation of genetic disorders in primary ovarian insufficiency (POI), but also to the advancement of novel treatments for patients with POI. Accumulating evidence indicates that mutations in NOBOX, DAZL and FIGLAgenes are associated with POI. In addition, cell biology studies revealed the important roles of these genes as essential translational factors for germ cell development. Recent insights into the role of the PI3K (phosphatidylinositol 3-kinase)-Akt signaling pathway in primordial follicle activation allowed the development of a new infertility treatment, IVA (in vitro activation), leading to successful pregnancy/delivery in POI patients. Furthermore, elucidation of genetic dynamics underlying female germ cell development could allow regeneration of oocytes from ES (embryonic stem)/iPS (induced pluripotent stem) cells in mammals. The purpose of this review is to summarize basic findings related to female germ cell development and potential clinical implications, especially focusing on POI etiologies. We also summarize evolving new POI therapies based on IVA as well as oocyte regeneration.
Collapse
Affiliation(s)
- Ikko Kawashima
- Department of Advanced Reproductive MedicineSt. Marianna University School of Medicine, Kawasaki City, Kanagawa, Japan
| | - Kazuhiro Kawamura
- Department of Advanced Reproductive MedicineSt. Marianna University School of Medicine, Kawasaki City, Kanagawa, Japan
| |
Collapse
|
135
|
Liu Y, Lin H. Genetic analysis of the reproductive axis in fish using genome-editing nucleases. Sci Bull (Beijing) 2017; 62:302-308. [PMID: 36659358 DOI: 10.1016/j.scib.2017.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 10/24/2016] [Accepted: 11/06/2016] [Indexed: 01/21/2023]
Abstract
Reproduction in fish is controlled by the brain-pituitary-gonad reproductive axis. Although genes of the reproductive axis are conserved from fish to humans, their in vivo functions are less clear in fish. Mutant lines of the reproductive axis have been systematically investigated in zebrafish and medaka using recently developed genome-editing nucleases. Here, we review recent progress in the genetic analysis of the reproductive axis in fish as well as the opportunities and challenges of applying genome-editing nucleases in fisheries.
Collapse
Affiliation(s)
- Yun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals, and the Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China.
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals, and the Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
136
|
Banerjee AA, Achrekar SK, Joseph S, Pathak BR, Mahale SD. Functional characterization of two naturally occurring mutations V 221G and T 449N in the follicle stimulating hormone receptor. Mol Cell Endocrinol 2017; 440:69-79. [PMID: 27889471 DOI: 10.1016/j.mce.2016.11.020] [Citation(s) in RCA: 8] [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: 09/15/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 12/28/2022]
Abstract
Naturally occurring mutations in follicle stimulating hormone receptor (FSHR) affect the receptor function. Here, we characterized two such previously reported mutations, V221G and T449N, in the extracellular domain and transmembrane helix 3, of FSHR, respectively. Functional studies with the V221G mutant demonstrated an impairment in FSH binding and signaling. Validation of X-ray crystallography data indicating the contribution of FSHR specific residues in the vicinity of V221 to contribute to FSH-FSHR interaction was carried out. In vitro mutational studies showed that these residues are determinants of both FSH binding and FSH induced signaling. Analysis of the T449N mutation revealed that it results in an increase in FSH binding and high cAMP response at lower doses of FSH. A marginal hCG induced and no TSH induced cAMP production was also observed. These findings corroborated with the clinical manifestations of primary amenorrhea (V221G) and spontaneous ovarian hyperstimulation syndrome (T449N) in women harbouring these mutations.
Collapse
Affiliation(s)
- Antara A Banerjee
- Division of Structural Biology, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Swati K Achrekar
- ICMR Biomedical Informatics Centre, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Shaini Joseph
- ICMR Biomedical Informatics Centre, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Bhakti R Pathak
- Division of Structural Biology, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Smita D Mahale
- Division of Structural Biology, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India; ICMR Biomedical Informatics Centre, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India.
| |
Collapse
|
137
|
Núñez Miguel R, Sanders J, Furmaniak J, Rees Smith B. Glycosylation pattern analysis of glycoprotein hormones and their receptors. J Mol Endocrinol 2017; 58:25-41. [PMID: 27875255 DOI: 10.1530/jme-16-0169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/13/2016] [Indexed: 11/08/2022]
Abstract
We have studied glycosylation patterns in glycoprotein hormones (GPHs) and glycoprotein hormone receptor (GPHR) extracellular domains (ECD) from different species to identify areas not glycosylated that could be involved in intermolecular or intramolecular interactions. Comparative models of the structure of the TSHR ECD in complex with TSH and in complex with TSHR autoantibodies (M22, stimulating and K1-70, blocking) were obtained based on the crystal structures of the FSH-FSHR ECD, M22-TSHR leucine-rich repeat domain (LRD) and K1-70-TSHR LRD complexes. The glycosylation sites of the GPHRs and GPHs from all species studied were mapped on the model of the human TSH TSHR ECD complex. The areas on the surfaces of GPHs that are known to interact with their receptors are not glycosylated and two areas free from glycosylation, not involved in currently known interactions, have been identified. The concave faces of GPHRs leucine-rich repeats 3-7 are free from glycosylation, consistent with known interactions with the hormones. In addition, four other non-glycosylated areas have been identified, two located on the receptors' convex surfaces, one in the long loop of the hinge regions and one at the C-terminus of the extracellular domains. Experimental evidence suggests that the non-glycosylated areas identified on the hormones and receptors are likely to be involved in forming intramolecular or intermolecular interactions.
Collapse
|
138
|
|
139
|
Rossetti R, Ferrari I, Bonomi M, Persani L. Genetics of primary ovarian insufficiency. Clin Genet 2016; 91:183-198. [PMID: 27861765 DOI: 10.1111/cge.12921] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 12/15/2022]
Abstract
Primary ovarian insufficiency (POI) is characterized by a loss of ovarian function before the age of 40 and account for one major cause of female infertility. POI relevance is continuously growing because of the increasing number of women desiring conception beyond 30 years of age, when POI prevalence is >1%. POI is highly heterogeneous and can present with ovarian dysgenesis and primary amenorrhea, or with secondary amenorrhea, and it can be associated with other congenital or acquired abnormalities. In most cases POI remains classified as idiopathic. However, the age of menopause is an inheritable trait and POI has a strong genetic component. This is confirmed by the existence of several candidate genes, experimental and natural models. The variable expressivity of POI defect may indicate that, this disease may frequently be considered as a multifactorial or oligogenic defect. The most common genetic contributors to POI are the X chromosome-linked defects. Here, we review the principal X-linked and autosomal genes involved in syndromic and non-syndromic forms of POI with the expectation that this list will soon be upgraded, thus allowing the possibility to predict the risk of an early age at menopause in families with POI.
Collapse
Affiliation(s)
- R Rossetti
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - I Ferrari
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - M Bonomi
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - L Persani
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
140
|
Varimo T, Miettinen PJ, Känsäkoski J, Raivio T, Hero M. Congenital hypogonadotropic hypogonadism, functional hypogonadotropism or constitutional delay of growth and puberty? An analysis of a large patient series from a single tertiary center. Hum Reprod 2016; 32:147-153. [PMID: 27927844 DOI: 10.1093/humrep/dew294] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/19/2016] [Accepted: 10/28/2016] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION What diagnoses underlie delayed puberty (DP) and predict its outcome? SUMMARY ANSWER A multitude of different diagnoses underlie DP, and in boys a history of cryptorchidism, small testicular size and slow growth velocity (GV) predict its clinical course. WHAT IS KNOWN ALREADY DP is caused by a variety of underlying etiologies. Hormonal markers can be used in the differential diagnosis of DP but none of them have shown complete diagnostic accuracy. STUDY DESIGN, SIZE, DURATION Medical records of 589 patients evaluated for DP in a single tertiary care center between 2004 and 2014 were retrospectively reviewed. PARTICIPANTS/MATERIALS, SETTING, METHODS Clinical and biochemical data of 174 boys and 70 girls who fulfilled the criteria of DP were included in the analyses. We characterized the frequencies of underlying conditions and evaluated the predictive efficacy of selected clinical and hormonal markers. MAIN RESULTS AND THE ROLE OF CHANCE Thirty etiologies that underlie DP were identified. No markers of clinical value could be identified in the girls, whereas a history of cryptorchidism in the boys was associated with an increase in the risk of permanent hypogonadism (odds ratio 17.2 (95% CI; 3.4-85.4, P < 0.001)). The conditions that cause functional hypogonadotropic hypogonadism were more frequent in boys with a GV below 3 cm/yr than in those growing faster (19% vs 4%, P < 0.05). In this series, the most effective markers to discriminate the prepubertal boys with constitutional delay of growth and puberty (CDGP) from those with congenital hypogonadotropic hypogonadism (CHH) were testicular volume (cut-off 1.1 ml with a sensitivity of 100% and a specificity of 91%), GnRH-induced maximal LH (cut-off 4.3 IU/L; 100%, 75%) and basal inhibin B (INHB) level (cut-off 61 ng/L; 90%, 83%). LIMITATIONS, REASONS FOR CAUTION The main limitation of the study is the retrospective design. WIDER IMPLICATIONS OF THE FINDINGS Prior cryptorchidism and slow GV are two important clinical cues that may help clinicians to predict the clinical course of DP in boys, whereas markers of similar value could not be identified in girls. In prepubertal boys, testicular size appeared as effective as INHB and GnRH-induced LH levels in the differential diagnosis between CHH and CDGP. STUDY FUNDING/COMPETING INTERESTS This study was supported by the Academy of Finland (268356), Foundation for Pediatric Research (7495), Sigrid Juselius Foundation (2613) and the Finnish Medical Foundation (011115). The authors have no competing interests to report. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Tero Varimo
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Päivi J Miettinen
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Molecular Neurology, and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| | - Johanna Känsäkoski
- Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Taneli Raivio
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland .,Faculty of Medicine, Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Matti Hero
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
141
|
Tucker EJ, Grover SR, Bachelot A, Touraine P, Sinclair AH. Premature Ovarian Insufficiency: New Perspectives on Genetic Cause and Phenotypic Spectrum. Endocr Rev 2016; 37:609-635. [PMID: 27690531 DOI: 10.1210/er.2016-1047] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Premature ovarian insufficiency (POI) is one form of female infertility, defined by loss of ovarian activity before the age of 40 and characterized by amenorrhea (primary or secondary) with raised gonadotropins and low estradiol. POI affects up to one in 100 females, including one in 1000 before the age of 30. Substantial evidence suggests a genetic basis for POI; however, the majority of cases remain unexplained, indicating that genes likely to be associated with this condition are yet to be discovered. This review discusses the current knowledge of the genetic basis of POI. We highlight genes typically known to cause syndromic POI that can be responsible for isolated POI. The role of mouse models in understanding POI pathogenesis is discussed, and a thorough list of candidate POI genes is provided. Identifying a genetic basis for POI has multiple advantages, such as enabling the identification of presymptomatic family members who can be offered counseling and cryopreservation of eggs before depletion, enabling personalized treatment based on the cause of an individual's condition, and providing better understanding of disease mechanisms that ultimately aid the development of improved treatments.
Collapse
Affiliation(s)
- Elena J Tucker
- Murdoch Children's Research Institute (E.J.T., S.R.G., A.H.S.), Royal Children's Hospital, Melbourne, VIC 3052 Australia; Department of Paediatrics (E.J.T., S.R.G., A.H.S.), University of Melbourne, Melbourne, VIC 3010, Australia; Department of Paediatric and Adolescent Gynaecology (S.R.G.), Royal Children's Hospital, Melbourne, VIC 3052, Australia; Assistance Publique Hôpitaux de Paris, (A.B., P.T.), IE3M, Université Pierre et Marie Curie, Paris 6 University, Department of Endocrinology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et des Pathologies Gynécologiques Rares, Pitié-Salpêtrière Hospital, Université Pierre et Marie Curie, 75013 Paris, France; Institut National de la Santé et de la Recherche Médicale (A.B., P.T.), 75654 Paris, France
| | - Sonia R Grover
- Murdoch Children's Research Institute (E.J.T., S.R.G., A.H.S.), Royal Children's Hospital, Melbourne, VIC 3052 Australia; Department of Paediatrics (E.J.T., S.R.G., A.H.S.), University of Melbourne, Melbourne, VIC 3010, Australia; Department of Paediatric and Adolescent Gynaecology (S.R.G.), Royal Children's Hospital, Melbourne, VIC 3052, Australia; Assistance Publique Hôpitaux de Paris, (A.B., P.T.), IE3M, Université Pierre et Marie Curie, Paris 6 University, Department of Endocrinology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et des Pathologies Gynécologiques Rares, Pitié-Salpêtrière Hospital, Université Pierre et Marie Curie, 75013 Paris, France; Institut National de la Santé et de la Recherche Médicale (A.B., P.T.), 75654 Paris, France
| | - Anne Bachelot
- Murdoch Children's Research Institute (E.J.T., S.R.G., A.H.S.), Royal Children's Hospital, Melbourne, VIC 3052 Australia; Department of Paediatrics (E.J.T., S.R.G., A.H.S.), University of Melbourne, Melbourne, VIC 3010, Australia; Department of Paediatric and Adolescent Gynaecology (S.R.G.), Royal Children's Hospital, Melbourne, VIC 3052, Australia; Assistance Publique Hôpitaux de Paris, (A.B., P.T.), IE3M, Université Pierre et Marie Curie, Paris 6 University, Department of Endocrinology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et des Pathologies Gynécologiques Rares, Pitié-Salpêtrière Hospital, Université Pierre et Marie Curie, 75013 Paris, France; Institut National de la Santé et de la Recherche Médicale (A.B., P.T.), 75654 Paris, France
| | - Philippe Touraine
- Murdoch Children's Research Institute (E.J.T., S.R.G., A.H.S.), Royal Children's Hospital, Melbourne, VIC 3052 Australia; Department of Paediatrics (E.J.T., S.R.G., A.H.S.), University of Melbourne, Melbourne, VIC 3010, Australia; Department of Paediatric and Adolescent Gynaecology (S.R.G.), Royal Children's Hospital, Melbourne, VIC 3052, Australia; Assistance Publique Hôpitaux de Paris, (A.B., P.T.), IE3M, Université Pierre et Marie Curie, Paris 6 University, Department of Endocrinology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et des Pathologies Gynécologiques Rares, Pitié-Salpêtrière Hospital, Université Pierre et Marie Curie, 75013 Paris, France; Institut National de la Santé et de la Recherche Médicale (A.B., P.T.), 75654 Paris, France
| | - Andrew H Sinclair
- Murdoch Children's Research Institute (E.J.T., S.R.G., A.H.S.), Royal Children's Hospital, Melbourne, VIC 3052 Australia; Department of Paediatrics (E.J.T., S.R.G., A.H.S.), University of Melbourne, Melbourne, VIC 3010, Australia; Department of Paediatric and Adolescent Gynaecology (S.R.G.), Royal Children's Hospital, Melbourne, VIC 3052, Australia; Assistance Publique Hôpitaux de Paris, (A.B., P.T.), IE3M, Université Pierre et Marie Curie, Paris 6 University, Department of Endocrinology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et des Pathologies Gynécologiques Rares, Pitié-Salpêtrière Hospital, Université Pierre et Marie Curie, 75013 Paris, France; Institut National de la Santé et de la Recherche Médicale (A.B., P.T.), 75654 Paris, France
| |
Collapse
|
142
|
Effect of Follicle Stimulating Hormone Receptor Gene Polymorphisms in Cervical Cancer Risk. Pathol Oncol Res 2016; 23:565-572. [DOI: 10.1007/s12253-016-0152-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 11/09/2016] [Indexed: 02/03/2023]
|
143
|
Allegra A, Marino A, Raimondo S, Maiorana A, Gullo S, Scaglione P, Volpes A, Alessandro R. The carriers of the A/G-G/G allelic combination of the c.2039 A>G and c.-29 G>A FSH receptor polymorphisms retrieve the highest number of oocytes in IVF/ICSI cycles. J Assist Reprod Genet 2016; 34:263-273. [PMID: 27817039 DOI: 10.1007/s10815-016-0835-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/23/2016] [Indexed: 01/27/2023] Open
Abstract
PURPOSE The objective of this study was the elucidation of the possible role of the single-nucleotide polymorphisms (SNP) at position -29 and 2039 of the FSH receptor gene (FSHR) as independent predictive markers of ovarian response. Indeed, the tailoring of reproductive treatments is crucial for both maximizing the success of IVF patients and obtaining a reduction in hypo- or hyper-response rates. METHODS This prospective, observational study analyzed the association of -29 and 2039 FSHR polymorphisms with the number of retrieved oocytes in 140 patients attending an IVF/ICSI cycle for severe male factors (≤5,000,000 spermatozoa/mL) or tubal factors at the ANDROS Day Surgery Clinic, Palermo, Italy. RESULTS The results of this study demonstrate that the genetic combination of A/G for polymorphism c.2039 A>G with G/G for polymorphism c.-29 G>A is significantly associated with the highest number of collected oocytes (p = 0.03). This association was significant even after controlling for the effect of other clinical variables. CONCLUSIONS The A/G-G/G allelic variant, identified as an independent variable, if confirmed in a larger number of patients, could be considered as a new genetic biomarker, which could increase the efficacy of prediction models for ovarian stimulation.
Collapse
Affiliation(s)
- Adolfo Allegra
- Reproductive Medicine Unit, ANDROS Day Surgery Clinic, Via Ausonia 43/45, 90144, Palermo, Italy.
| | - Angelo Marino
- Reproductive Medicine Unit, ANDROS Day Surgery Clinic, Via Ausonia 43/45, 90144, Palermo, Italy
| | - Stefania Raimondo
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| | - Antonio Maiorana
- Unità Operativa Complessa di Ginecologia ed Ostetricia, A.R.N.A.S. Ospedale Civico, Palermo, Italy
| | - Salvatore Gullo
- Medical Statistics Unit, ANDROS Day Surgery Clinic, Palermo, Italy
| | - Piero Scaglione
- Reproductive Medicine Unit, ANDROS Day Surgery Clinic, Via Ausonia 43/45, 90144, Palermo, Italy
| | - Aldo Volpes
- Reproductive Medicine Unit, ANDROS Day Surgery Clinic, Via Ausonia 43/45, 90144, Palermo, Italy
| | - Riccardo Alessandro
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università di Palermo, Palermo, Italy
| |
Collapse
|
144
|
Mouse Models for the Study of Synthesis, Secretion, and Action of Pituitary Gonadotropins. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 143:49-84. [PMID: 27697204 DOI: 10.1016/bs.pmbts.2016.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Gonadotropins play fundamental roles in reproduction. More than 30years ago, Cga transgenic mice were generated, and more than 20years ago, the phenotypes of Cga null mice were reported. Since then, numerous mouse strains have been generated and characterized to address several questions in reproductive biology involving gonadotropin synthesis, secretion, and action. More recently, extragonadal expression, and in some cases, functions of gonadotropins in nongonadal tissues have been identified. Several genomic and proteomic approaches including novel mouse genome editing tools are available now. It is anticipated that these and other emerging technologies will be useful to build an integrated network of gonadotropin signaling pathways in various tissues. Undoubtedly, research on gonadotropins will continue to provide new knowledge and allow us transcend from benchside to the bedside.
Collapse
|
145
|
Dewailly D, Robin G, Peigne M, Decanter C, Pigny P, Catteau-Jonard S. Interactions between androgens, FSH, anti-Müllerian hormone and estradiol during folliculogenesis in the human normal and polycystic ovary. Hum Reprod Update 2016; 22:709-724. [PMID: 27566840 DOI: 10.1093/humupd/dmw027] [Citation(s) in RCA: 312] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 07/12/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Androgens, FSH, anti-Müllerian hormone (AMH) and estradiol (E2) are essential in human ovarian folliculogenesis. However, the interactions between these four players is not fully understood. OBJECTIVES AND RATIONALE The purpose of this review is to highlight the chronological sequence of the appearance and function of androgens, FSH, AMH and E2 and to discuss controversies in the relationship between FSH and AMH. A better understanding of this interaction could supplement our current knowledge about the pathophysiology of the polycystic ovary syndrome (PCOS). SEARCH METHODS A literature review was performed using the following search terms: androgens, FSH, FSH receptor, anti-Mullerian hormone, AMHRII, estradiol, follicle, ovary, PCOS, aromatase, granulosa cell, oocyte. The time period searched was 1980-2015 and the databases interrogated were PubMed and Web of Science. OUTCOMES During the pre-antral ('gonadotropin-independent') follicle growth, FSH is already active and promotes follicle growth in synergy with theca cell-derived androgens. Conversely, AMH is inhibitory by counteracting FSH. We challenge the hypothesis that AMH is regulated by androgens and propose rather an indirect effect through an androgen-dependent amplification of FSH action on granulosa cells (GCs) from small growing follicles. This hypothesis implies that FSH stimulates AMH expression. During the antral ('gonadotropin-dependent') follicle growth, E2 production results from FSH-dependent activation of aromatase. Conversely, AMH is inhibitory but the decline of its expression, amplified by E2, allows full expression of aromatase, characteristic of the large antral follicles. We propose a theoretical scheme made up of two triangles that follow each other chronologically. In PCOS, pre-antral follicle growth is excessive (triangle 1) because of intrinsic androgen excess that renders GCs hypersensitive to FSH, with consequently excessive AMH expression. Antral follicle growth and differentiation are disturbed (triangle 2) because of the abnormally persisting inhibition of FSH effects by AMH that blocks aromatase. Beside anovulation, this scenario may also serve to explain the higher receptiveness to gonadotropin therapy and the increased risk of ovarian hyperstimulation syndrome (OHSS) in patients with PCOS. WIDER IMPLICATIONS Within GCs, the balance between FSH and AMH effects is pivotal in the shift from androgen- to oestrogen-driven follicles. Our two triangles hypothesis, based on updated data from the literature, offers a pedagogic template for the understanding of folliculogenesis in the normal and polycystic ovary. It opens new avenues for the treatment of anovulation due to PCOS.
Collapse
Affiliation(s)
- Didier Dewailly
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France .,Faculté de Médecine, Université Lille Nord de France, 59000 Lille, France
| | - Geoffroy Robin
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France
| | - Maëliss Peigne
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France
| | - Christine Decanter
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France
| | - Pascal Pigny
- Faculté de Médecine, Université Lille Nord de France, 59000 Lille, France.,CHU Lille, Laboratoire de Biochimie & Hormonologie, Centre de Biologie Pathologie, F-59037 Lille, France
| | - Sophie Catteau-Jonard
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France.,Faculté de Médecine, Université Lille Nord de France, 59000 Lille, France
| |
Collapse
|
146
|
Busch AS, Hagen CP, Almstrup K, Main KM, Juul A. Genetic variations altering FSH action affect circulating hormone levels as well as follicle growth in healthy peripubertal girls. Hum Reprod 2016; 31:897-904. [PMID: 26905078 DOI: 10.1093/humrep/dew022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 01/22/2016] [Indexed: 02/07/2023] Open
Abstract
STUDY QUESTION Do variants of the genes encoding follicle stimulating hormone (FSH) beta subunit (B) and FSH receptor (R) impact circulating reproductive hormone levels and ovarian follicle maturation in healthy peripubertal girls? SUMMARY ANSWER FSHB and FSHR genetic variants exert, alone or their combination, distinct effects on reproductive hormone levels as well as ovarian follicle maturation in healthy peripubertal girls. WHAT IS KNOWN ALREADY FSHB and FSHR genetic variants impact reproductive hormone levels as well as associated pathologies in women. While FSHR c. 2039A>G is known to alter gonadotrophin levels in women, FSHR c.-29G>A has not yet been shown to exert effect and there are conflicting results concerning FSHB c.-211G>T. STUDY DESIGN, SIZE, DURATION This population-based study included 633 girls recruited as part of two cohorts, the COPENHAGEN Puberty Study (2006-2014, a cross-sectional and ongoing longitudinal study) and the Copenhagen Mother-Child Cohort (1997-2002, including transabdominal ultrasound (TAUS) of the ovaries in a subset of 91 peripubertal girls). PARTICIPANTS/MATERIALS, SETTING, METHODS Clinical examinations, including pubertal breast stage (Tanner's classification B1-B5) were performed. Circulating levels of FSH, luteinizing hormone (LH), estradiol, anti-Mullerian hormone (AMH) and inhibin-B were assessed by immunoassays. In a subset of the girls (n = 91), ovarian volume and the number/size of antral follicles were assessed by TAUS. Genotypes were determined by competitive PCR. MAIN RESULTS AND THE ROLE OF CHANCE FSHR c.2039A>G minor alleles were positively associated with serum FSH (β = 0.08, P = 0.004), LH (β = 0.06, P = 0.012) and estradiol (β = 0.06, P = 0.017) (adjusted for Tanner stages). In a combined model, FSHR c.-29G>A and FSHR c.2039A>G alleles were positively associated with FSH levels in early-pubertal girls (B2 + B3, n = 327, r = 0.1, P = 0.02) and in young adolescents (B4 + B5, n = 149, r = 0.2, P = 0.01). Serum AMH and inhibin B levels were not significantly influenced by the single nucleotide polymorphisms (SNPs). Single SNPs were not associated with follicles counts, however, a cumulative minor allele count (FSHB c.-211 G>T and FSHR c.-29G>A) was negatively associated with the number of large follicles (≥5 mm) (n = 91, P = 0.04) (adjusted for Tanner stages). LIMITATIONS, REASONS FOR CAUTION Since we studied girls and young adolescents during pubertal transition, our study may not be fully comparable with previous studies on FSHB and FSHR variants in adult women. The group of young adolescents (Tanner B4 + B5) reflects the endocrine situation in adult women best, however, the group is not large enough to contribute substantially to the conflicting results concerning the influence of FSHB c.-211G>T in adult women. Furthermore, we have no information about the exact day of the menstrual cycle in the subgroup of girls with menarche. WIDER IMPLICATIONS OF THE FINDINGS The sex-specific interaction of FSHB and FSHR genetic variants and physiological as well as pathological conditions is being increasingly elucidated. The variant triplet set might serve as diagnostic and pharmacogenetic marker. For the first time, we show an additional effect of FSHR c.-29G>A on serum FSH levels in healthy girls. Moreover, morphological data suggest impaired FSH-induced maturation of ovarian follicles in minor allele carriers of FSHB c.-211G>T and FSHR c.-29G>A. This may explain previous findings of delayed pubertal onset in these girls. STUDY FUNDING/COMPETING INTERESTS Funding was provided by the Danish Agency for Science, Technology and Innovation (09-067180), Danish Ministry of the Environment, CeHoS (MST-621-00065), Capital Region of Denmark (December 2011), Ministry of Higher Education and Science (DFF-1331-00113) and EDMaRC (Danish Ministry of Health). A.S.B. was funded from December 2015 by ReproUnion (EU Interreg Öresund-Kattegat-Skagerrak). The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Alexander S Busch
- Department of Growth and Reproduction and EDMaRC, Rigshospitalet, Section 5064, University of Copenhagen, Blegdamsvej 9, DK-2100, Copenhagen O, Denmark
| | - Casper P Hagen
- Department of Growth and Reproduction and EDMaRC, Rigshospitalet, Section 5064, University of Copenhagen, Blegdamsvej 9, DK-2100, Copenhagen O, Denmark
| | - Kristian Almstrup
- Department of Growth and Reproduction and EDMaRC, Rigshospitalet, Section 5064, University of Copenhagen, Blegdamsvej 9, DK-2100, Copenhagen O, Denmark
| | - Katharina M Main
- Department of Growth and Reproduction and EDMaRC, Rigshospitalet, Section 5064, University of Copenhagen, Blegdamsvej 9, DK-2100, Copenhagen O, Denmark
| | - Anders Juul
- Department of Growth and Reproduction and EDMaRC, Rigshospitalet, Section 5064, University of Copenhagen, Blegdamsvej 9, DK-2100, Copenhagen O, Denmark
| |
Collapse
|
147
|
Bramble MS, Goldstein EH, Lipson A, Ngun T, Eskin A, Gosschalk JE, Roach L, Vashist N, Barseghyan H, Lee E, Arboleda VA, Vaiman D, Yuksel Z, Fellous M, Vilain E. A novel follicle-stimulating hormone receptor mutation causing primary ovarian failure: a fertility application of whole exome sequencing. Hum Reprod 2016; 31:905-14. [PMID: 26911863 DOI: 10.1093/humrep/dew025] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 01/28/2016] [Indexed: 12/11/2022] Open
Abstract
STUDY QUESTION Can whole exome sequencing (WES) and in vitro validation studies be used to find the causative genetic etiology in a patient with primary ovarian failure and infertility? SUMMARY ANSWER A novel follicle-stimulating hormone receptor (FSHR) mutation was found by WES and shown, via in vitro flow cytometry studies, to affect membrane trafficking. WHAT IS KNOWN ALREADY WES may diagnose up to 25-35% of patients with suspected disorders of sex development (DSD). FSHR mutations are an extremely rare cause of 46, XX gonadal dysgenesis with primary amenorrhea due to hypergonadotropic ovarian failure. STUDY DESIGN, SIZE, DURATION A WES study was followed by flow cytometry studies of mutant protein function. PARTICIPANTS/MATERIALS, SETTING, METHODS The study subjects were two Turkish sisters with hypergonadotropic primary amenorrhea, their parents and two unaffected sisters. The affected siblings and both parents were sequenced (trio-WES). Transient transfection of HEK 293T cells was performed with a vector containing wild-type FSHR as well as the novel FSHR variant that was discovered by WES. Cellular localization of FSHR protein as well as FSH-stimulated cyclic AMP (cAMP) production was evaluated using flow cytometry. MAIN RESULTS AND THE ROLE OF CHANCE Both affected sisters were homozygous for a previously unreported missense mutation (c.1222G>T, p.Asp408Tyr) in the second transmembrane domain of FSHR. Modeling predicted disrupted secondary structure. Flow cytometry demonstrated an average of 48% reduction in cell-surface signal detection (P < 0.01). The mean fluorescent signal for cAMP (second messenger of FSHR), stimulated by FSH, was reduced by 50% in the mutant-transfected cells (P < 0.01). LIMITATIONS, REASONS FOR CAUTION This is an in vitro validation. All novel purported genetic variants can be clinically reported only as 'variants of uncertain significance' until more patients with a similar phenotype are discovered with the same variant. WIDER IMPLICATIONS OF THE FINDINGS We report the first WES-discovered FSHR mutation, validated by quantitative flow cytometry. WES is a valuable tool for diagnosis of rare genetic diseases, and flow cytometry allows for quantitative characterization of purported variants. WES-assisted diagnosis allows for treatments aimed at the underlying molecular etiology of disease. Future studies should focus on pharmacological and assisted reproductive treatments aimed at the disrupted FSHR, so that patients with FSH resistance can be treated by personalized medicine. STUDY FUNDING/COMPETING INTERESTS E.V. is partially funded by the DSD Translational Research Network (NICHD 1R01HD068138). M.S.B. is funded by the Neuroendocrinology, Sex Differences and Reproduction training grant (NICHD 5T32HD007228). The authors have no competing interests to disclose.
Collapse
Affiliation(s)
- Matthew S Bramble
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| | - Ellen H Goldstein
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of California Los Angeles, 10833 Le Conte Avenue, Room 24-130 CHS, Los Angeles, CA 90095, USA
| | - Allen Lipson
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| | - Tuck Ngun
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| | - Ascia Eskin
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| | - Jason E Gosschalk
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| | - Lara Roach
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| | - Neerja Vashist
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| | - Hayk Barseghyan
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| | - Eric Lee
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| | - Valerie A Arboleda
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| | - Daniel Vaiman
- Department of Development, Reproduction, and Cancer, Institut Cochin, U1016 Inserm, University Sorbonne Paris, CNRS UMR8104, Paris, France
| | - Zafer Yuksel
- Department of Medical Genetics, Women and Children Hospital, Halkkent Mh. Fatih Sultan Mehmet Boulevard No. 23, Mersin 33240, Turkey
| | - Marc Fellous
- Department of Development, Reproduction, and Cancer, Institut Cochin, U1016 Inserm, University Sorbonne Paris, CNRS UMR8104, Paris, France
| | - Eric Vilain
- Department of Human Genetics, David Geffen School of Medicine at the University of California Los Angeles, 695 Charles E Young Drive South, Los Angeles, CA 90095, USA
| |
Collapse
|
148
|
Abstract
In the female gonad, distinct signalling pathways activate ovarian differentiation while repressing the formation of testes. Human disorders of sex development (DSDs), such as 46,XX DSDs, can arise when this signalling is aberrant. Here we review the current understanding of the genetic mechanisms that control gonadal development, with particular emphasis on those that drive or inhibit ovarian differentiation. We discuss how disruption to these molecular pathways can lead to 46,XX disorders of ovarian development. Finally, we look at recently characterized novel genes and pathways that contribute and speculate how advances in technology will aid in further characterization of normal and disrupted human ovarian development.
Collapse
|
149
|
Abstract
As age at pubertal onset declines and age at first pregnancy increases, the mechanisms that regulate female reproductive lifespan become increasingly relevant to population health. The timing of menarche and menopause can have profound effects not only on fertility but also on the risk of diseases such as type 2 diabetes mellitus, cardiovascular disease and breast cancer. Genetic studies have identified dozens of highly penetrant rare mutations associated with reproductive disorders, and also ∼175 common genetic variants associated with the timing of puberty or menopause. These findings, alongside other functional studies, have highlighted a diverse range of mechanisms involved in reproductive ageing, implicating core biological processes such as cell cycle regulation and energy homeostasis. The aim of this article is to review the contribution of such genetic findings to our understanding of the molecular regulation of reproductive timing, as well as the biological basis of the epidemiological links between reproductive ageing and disease risk.
Collapse
Affiliation(s)
- John R.B. Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0QQ
| | - Anna Murray
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon & Exeter Hospital, Barrack Road, Exeter, EX2 5DW
| | - Felix R Day
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0QQ
| | - Ken K Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Box 285 Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0QQ
- Department of Paediatrics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0QQ
| |
Collapse
|
150
|
Pelosi E, Forabosco A, Schlessinger D. Genetics of the ovarian reserve. Front Genet 2015; 6:308. [PMID: 26528328 PMCID: PMC4606124 DOI: 10.3389/fgene.2015.00308] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/24/2015] [Indexed: 11/13/2022] Open
Abstract
Primordial follicles or non-growing follicles (NGFs) are the functional unit of reproduction, each comprising a single germ cell surrounded by supporting somatic cells. NGFs constitute the ovarian reserve (OR), prerequisite for germ cell ovulation and the continuation of the species. The dynamics of the reserve is determined by the number of NGFs formed and their complex subsequent fates. During the reproductive lifespan, the OR progressively diminishes due to follicle atresia as well as recruitment, maturation, and ovulation. The depletion of the OR is the major determining driver of menopause, which ensues when the number of primordial follicles falls below a threshold of ∼1,000. Therefore, genes and processes involved in follicle dynamics are particularly important to understand the process of menopause, both in the typical reproductive lifespan and in conditions like primary ovarian insufficiency, defined as menopause before age 40. Genes and their variants that affect the timing of menopause thereby provide candidates for diagnosis of and intervention in problems of reproductive lifespan. We review the current knowledge of processes and genes involved in the development of the OR and in the dynamics of ovarian follicles.
Collapse
Affiliation(s)
- Emanuele Pelosi
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | | | - David Schlessinger
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|