1
|
Li XF, Hu J, Liu L, Tang Y, Zhou JL, Gu YF, Li Y, Tan C, Tu C, Zhang QJ, Hu L, Meng LL, Lu GX, Lin G, Du J, Tan YQ, He WB, Gong F. Women with FSHR-inactivating mutations causing resistant ovarian syndrome benefit from oocyte in-vitro maturation. Reprod Biomed Online 2025; 50:104733. [PMID: 40222241 DOI: 10.1016/j.rbmo.2024.104733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 04/15/2025]
Abstract
RESEARCH QUESTION Is in-vitro maturation (IVM) of oocytes effective for treating women with resistant ovarian syndrome (ROS) carrying biallelic FSHR-inactivating mutations? DESIGN Three patients with ROS were recruited for this study. Candidate pathogenic mutations were identified using whole-exome sequencing (WES). A cAMP production assay was performed to evaluate the effects of these mutations. IVM of oocytes was performed to achieve pregnancy. RESULTS After undergoing conventional IVF in the first cycle, WES was performed for the three patients with ROS. They were found to be carrying biallelic FHSR mutations, including four novel missense mutations (c.263C>T, c.1106T>C, c.211G>T and c.1349T>A) and one previously reported missense mutation (c.44G>A). A cAMP production assay was performed to evaluate the effects of these mutations, which revealed that they led to almost-complete inactivation of the mutated receptors upon FSH stimulation. IVM of the oocytes was performed for two patients and blastocysts were obtained successfully; these patients delivered healthy babies following frozen embryo transfer. CONCLUSIONS The five missense mutations in FSHR were deleterious and associated with ROS in the three patients. To the best of our knowledge, this is the first report of patients with biallelic FSHR-inactivating mutations who gave birth to genetically related children after IVM treatment. The findings show that IVM is an effective treatment for patients with ROS with FSHR-inactivating mutations.
Collapse
Affiliation(s)
- Xiao-Feng Li
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China; National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Jing Hu
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China; National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Ling Liu
- National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China
| | - Yi Tang
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China; National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Jing-Lin Zhou
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China
| | - Yi-Fan Gu
- National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Yong Li
- National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Chen Tan
- National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China
| | - Chaofeng Tu
- National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Qian-Jun Zhang
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China; National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Liang Hu
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China; National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Lan-Lan Meng
- National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Guang-Xiu Lu
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China; National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Ge Lin
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China; National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Juan Du
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China; National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Yue-Qiu Tan
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China; National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China
| | - Wen-Bin He
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China; National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China.
| | - Fei Gong
- Hunan Guangxiu Hospital Affiliated with Hunan Normal University, Hunan Normal University Health Science Centre, Changsha, PR China; National Engineering and Research Centre of Human Stem Cells and Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, PR China; Reproductive and Genetic Hospital of CITIC-Xiangya and Clinical Research Centre for Reproduction and Genetics in Hunan Province, Changsha, Hunan, PR China.
| |
Collapse
|
2
|
Yi H, Chen L, Zhang J, Wen Y, Zheng X, Chen X. Infertility management in resistant ovary syndrome: a review. Front Endocrinol (Lausanne) 2025; 16:1560981. [PMID: 40171200 PMCID: PMC11958170 DOI: 10.3389/fendo.2025.1560981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/21/2025] [Indexed: 04/03/2025] Open
Abstract
Resistant ovary syndrome is a rare endocrinological disorder characterized by elevated serum gonadotropins and normal ovarian reserves. The leading causes of this condition include FSHR mutations, and autoimmune disorders. Due to follicle maturation defects and anovulation, these patients have menstrual disturbances and infertility. Endocrinological disorders can be treated by regular hormone replacement therapy. However, there is no consensus regarding the infertility issues and current treatment remains experimental with controlled ovarian stimulation (COS) and in vitro maturation. Herein, we conducted a review of the current literature, which concludes that: 1) patients with FSHR mutations had very poor COS outcomes; 2) follicle-stimulating hormone and luteinizing hormone levels poorly predict COS outcomes; 3) both recombinant and urinary gonadotropins may be effective in COS; 4) the dosage of exogenous gonadotropins is not the key to successful COS; 5) in vitro maturation is a feasible option for patients carrying FSHR mutations or unsuccessful COS cycles.
Collapse
Affiliation(s)
- Hong Yi
- Department of Reproductive Health, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
| | - Lin Chen
- Department of Obstetrics and Gynecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jinglei Zhang
- Reproductive Medicine Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yangxing Wen
- Reproductive Medicine Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xi Zheng
- The First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Xiaoyan Chen
- Department of Obstetrics and Gynecology, Maternal-Fetal Medicine Institute, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
| |
Collapse
|
3
|
Schöneberg T. Modulating vertebrate physiology by genomic fine-tuning of GPCR functions. Physiol Rev 2025; 105:383-439. [PMID: 39052017 DOI: 10.1152/physrev.00017.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/08/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024] Open
Abstract
G protein-coupled receptors (GPCRs) play a crucial role as membrane receptors, facilitating the communication of eukaryotic species with their environment and regulating cellular and organ interactions. Consequently, GPCRs hold immense potential in contributing to adaptation to ecological niches and responding to environmental shifts. Comparative analyses of vertebrate genomes reveal patterns of GPCR gene loss, expansion, and signatures of selection. Integrating these genomic data with insights from functional analyses of gene variants enables the interpretation of genotype-phenotype correlations. This review underscores the involvement of GPCRs in adaptive processes, presenting numerous examples of how alterations in GPCR functionality influence vertebrate physiology or, conversely, how environmental changes impact GPCR functions. The findings demonstrate that modifications in GPCR function contribute to adapting to aquatic, arid, and nocturnal habitats, influencing camouflage strategies, and specializing in particular dietary preferences. Furthermore, the adaptability of GPCR functions provides an effective mechanism in facilitating past, recent, or ongoing adaptations in animal domestication and human evolution and should be considered in therapeutic strategies and drug development.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
- School of Medicine, University of Global Health Equity, Kigali, Rwanda
| |
Collapse
|
4
|
Carter D, Better M, Abbasi S, Zulfiqar F, Shapiro R, Ensign LM. Nanomedicine for Maternal and Fetal Health. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2303682. [PMID: 37817368 PMCID: PMC11004090 DOI: 10.1002/smll.202303682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/25/2023] [Indexed: 10/12/2023]
Abstract
Conception, pregnancy, and childbirth are complex processes that affect both mother and fetus. Thus, it is perhaps not surprising that in the United States alone, roughly 11% of women struggle with infertility and 16% of pregnancies involve some sort of complication. This presents a clear need to develop safe and effective treatment options, though the development of therapeutics for use in women's health and particularly in pregnancy is relatively limited. Physiological and biological changes during the menstrual cycle and pregnancy impact biodistribution, pharmacokinetics, and efficacy, further complicating the process of administration and delivery of therapeutics. In addition to the complex pharmacodynamics, there is also the challenge of overcoming physiological barriers that impact various routes of local and systemic administration, including the blood-follicle barrier and the placenta. Nanomedicine presents a unique opportunity to target and sustain drug delivery to the reproductive tract and other relevant organs in the mother and fetus, as well as improve the safety profile and minimize side effects. Nanomedicine-based approaches have the potential to improve the management and treatment of infertility, obstetric complications, and fetal conditions.
Collapse
Affiliation(s)
- Davell Carter
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Marina Better
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Saed Abbasi
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fareeha Zulfiqar
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rachel Shapiro
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Laura M. Ensign
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Departments of Gynecology and Obstetrics, Biomedical Engineering, Oncology, and Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
Zou X, Liang X, Dai W, Zhu T, Wang C, Zhou Y, Qian Y, Yan Z, Gao C, Gao L, Cui Y, Liu J, Meng Y. Peroxiredoxin 4 deficiency induces accelerated ovarian aging through destroyed proteostasis in granulosa cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167334. [PMID: 38971505 DOI: 10.1016/j.bbadis.2024.167334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/08/2024]
Abstract
Ovarian aging, a complex and challenging concern within the realm of reproductive medicine, is associated with reduced fertility, menopausal symptoms and long-term health risks. Our previous investigation revealed a correlation between Peroxiredoxin 4 (PRDX4) and human ovarian aging. The purpose of this research was to substantiate the protective role of PRDX4 against ovarian aging and elucidate the underlying molecular mechanism in mice. In this study, a Prdx4-/- mouse model was established and it was observed that the deficiency of PRDX4 led to only an accelerated decline of ovarian function in comparison to wild-type (WT) mice. The impaired ovarian function observed in this study can be attributed to an imbalance in protein homeostasis, an exacerbation of endoplasmic reticulum stress (ER stress), and ultimately an increase in apoptosis of granulosa cells. Furthermore, our research reveals a noteworthy decline in the expression of Follicle-stimulating hormone receptor (FSHR) in aging Prdx4-/- mice, especially the functional trimer, due to impaired disulfide bond formation. Contrarily, the overexpression of PRDX4 facilitated the maintenance of protein homeostasis, mitigated ER stress, and consequently elevated E2 levels in a simulated KGN cell aging model. Additionally, the overexpression of PRDX4 restored the expression of the correct spatial conformation of FSHR, the functional trimer. In summary, our research reveals the significant contribution of PRDX4 in delaying ovarian aging, presenting a novel and promising therapeutic target for ovarian aging from the perspective of endoplasmic reticulum protein homeostasis.
Collapse
Affiliation(s)
- Xiaofei Zou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiuru Liang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wangjuan Dai
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ting Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chaoyi Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yutian Zhou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yi Qian
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhengjie Yan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chao Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yugui Cui
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jiayin Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yan Meng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
6
|
Federici S, Rossetti R, Moleri S, Munari EV, Frixou M, Bonomi M, Persani L. Primary ovarian insufficiency: update on clinical and genetic findings. Front Endocrinol (Lausanne) 2024; 15:1464803. [PMID: 39391877 PMCID: PMC11466302 DOI: 10.3389/fendo.2024.1464803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/02/2024] [Indexed: 10/12/2024] Open
Abstract
Primary ovarian insufficiency (POI) is a disorder of insufficient ovarian follicle function before the age of 40 years with an estimated prevalence of 3.7% worldwide. Its relevance is emerging due to the increasing number of women desiring conception late or beyond the third decade of their lives. POI clinical presentation is extremely heterogeneous with a possible exordium as primary amenorrhea due to ovarian dysgenesis or with a secondary amenorrhea due to different congenital or acquired abnormalities. POI significantly impacts non only on the fertility prospect of the affected women but also on their general, psychological, sexual quality of life, and, furthermore, on their long-term bone, cardiovascular, and cognitive health. In several cases the underlying cause of POI remains unknown and, thus, these forms are still classified as idiopathic. However, we now know the age of menopause is an inheritable trait and POI has a strong genetic background. This is confirmed by the existence of several candidate genes, experimental and natural models. The most common genetic contributors to POI are the X chromosome-linked defects. Moreover, the variable expressivity of POI defect suggests it can be considered as a multifactorial or oligogenic defect. Here, we present an updated review on clinical findings and on the principal X-linked and autosomal genes involved in syndromic and non-syndromic forms of POI. We also provide current information on the management of the premature hypoestrogenic state as well as on fertility preservation in subjects at risk of POI.
Collapse
Affiliation(s)
- Silvia Federici
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Raffaella Rossetti
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Silvia Moleri
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Elisabetta V. Munari
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Maria Frixou
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Marco Bonomi
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Luca Persani
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| |
Collapse
|
7
|
Touraine P, Chabbert-Buffet N, Plu-Bureau G, Duranteau L, Sinclair AH, Tucker EJ. Premature ovarian insufficiency. Nat Rev Dis Primers 2024; 10:63. [PMID: 39266563 DOI: 10.1038/s41572-024-00547-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 09/14/2024]
Abstract
Premature ovarian insufficiency (POI) is a cause of infertility and endocrine dysfunction in women, defined by loss of normal, predictable ovarian activity before the age of 40 years. POI is clinically characterized by amenorrhoea (primary or secondary) with raised circulating levels of follicle-stimulating hormone. This condition can occur due to medical interventions such as ovarian surgery or cytotoxic cancer therapy, metabolic and lysosomal storage diseases, infections, chromosomal anomalies and autoimmune diseases. At least 1 in 100 women is affected by POI, including 1 in 1,000 before the age of 30 years. Substantial evidence suggests a genetic basis to POI. However, the cause of idiopathic POI remains unknown in most patients, indicating that gene variants associated with this condition remain to be discovered. Over the past 10 years, tremendous progress has been made in our knowledge of genes involved in POI. Genetic approaches in diagnosis are important as they enable patients with familial POI to be identified, with the opportunity for oocyte preservation. Moreover, genetic approaches could provide a better understanding of disease mechanisms, which will ultimately aid the development of improved treatments.
Collapse
Affiliation(s)
- Philippe Touraine
- Department of Endocrinology and Reproductive Medicine, AP-HP Pitié Salpêtrière Hospital, Sorbonne Université Médecine, Paris, France.
- Inserm U1151 INEM, Necker Hospital, Paris, France.
| | - Nathalie Chabbert-Buffet
- Department of Obstetrics, Gynecology and Reproductive Medicine, Tenon Hospital, AP-HP Sorbonne Université, Paris, France
- INSERM UMR S 938, CDR St Antoine, Paris, France
| | - Genevieve Plu-Bureau
- Department of Medical Gynecology, AP-HP Port Royal-Cochin Hospital, Université Paris Cité, Paris, France
- U1151 EPOPEE Team, Paris, France
| | - Lise Duranteau
- Department of Medical Gynecology, Bicêtre Hospital, AP-HP Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Andrew H Sinclair
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Elena J Tucker
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
8
|
Radomsky T, Anderson RC, Millar RP, Newton CL. Restoring function to inactivating G protein-coupled receptor variants in the hypothalamic-pituitary-gonadal axis 1. J Neuroendocrinol 2024; 36:e13418. [PMID: 38852954 DOI: 10.1111/jne.13418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 03/30/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024]
Abstract
G protein-coupled receptors (GPCRs) are central to the functioning of the hypothalamic-pituitary-gonadal axis (HPG axis) and include the rhodopsin-like GPCR family members, neurokinin 3 receptor, kappa-opioid receptor, kisspeptin 1 receptor, gonadotropin-releasing hormone receptor, and the gonadotropin receptors, luteinizing hormone/choriogonadotropin receptor and follicle-stimulating hormone receptor. Unsurprisingly, inactivating variants of these receptors have been implicated in a spectrum of reproductive phenotypes, including failure to undergo puberty, and infertility. Clinical induction of puberty in patients harbouring such variants is possible, but restoration of fertility is not always a realisable outcome, particularly for those patients suffering from primary hypogonadism. Thus, novel pharmaceuticals and/or a fundamental change in approach to treating these patients are required. The increasing wealth of data describing the effects of coding-region genetic variants on GPCR function has highlighted that the majority appear to be dysfunctional as a result of misfolding of the encoded receptor protein, which, in turn, results in impaired receptor trafficking through the secretory pathway to the cell surface. As such, these intracellularly retained receptors may be amenable to 'rescue' using a pharmacological chaperone (PC)-based approach. PCs are small, cell permeant molecules hypothesised to interact with misfolded intracellularly retained proteins, stabilising their folding and promoting their trafficking through the secretory pathway. In support of the use of this approach as a viable therapeutic option, it has been observed that many rescued variant GPCRs retain at least a degree of functionality when 'rescued' to the cell surface. In this review, we examine the GPCR PC research landscape, focussing on the rescue of inactivating variant GPCRs with important roles in the HPG axis, and describe what is known regarding the mechanisms by which PCs restore trafficking and function. We also discuss some of the merits and obstacles associated with taking this approach forward into a clinical setting.
Collapse
Affiliation(s)
- Tarryn Radomsky
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ross C Anderson
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Robert P Millar
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Claire L Newton
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
9
|
Current evaluation of amenorrhea: a committee opinion. Fertil Steril 2024; 122:52-61. [PMID: 38456861 DOI: 10.1016/j.fertnstert.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 03/09/2024]
Abstract
The purpose of this American Society for Reproductive Medicine Practice Committee Opinion is to provide clinicians with principles and strategies for the diagnostic evaluation of patients presenting with primary or secondary amenorrhea. This revised document replaces the Practice Committee Document titled "Current evaluation of amenorrhea," last published in 2008 (Fertil Steril 2008;90:S219-25).
Collapse
|
10
|
Xekouki P, Konstantinidou A, Tatsi C, Sertedaki A, Settas N, Loutradis D, Chrousos GP, Kanaka-Gantenbein C, Dacou-Voutetakis C, Voutetakis A. HNF1A gene mutations and premature ovarian failure (POF): evidence from a clinical paradigm combining MODY 3 and POF. Hormones (Athens) 2024; 23:345-350. [PMID: 38311659 PMCID: PMC11219395 DOI: 10.1007/s42000-024-00529-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/17/2024] [Indexed: 02/06/2024]
Abstract
Premature ovarian failure (POF) defines the occurrence of ovarian failure prior to the age of 40. It occurs in one out of 100 women but is very rare before age 20 (1:10,000). Maturity-onset diabetes of the young (MODY), caused by mutations in the HNF1A gene, is also a rare disorder; all types of MODY account for 1-2% of adult diabetic cases. These two rare nosologic entities coexisted in an adolescent girl evaluated for delayed puberty. Although this combination could represent a chance association, an interrelation might exist. We examined HNF1A expression in human fetal and adult ovaries by immunohistochemistry using a polyclonal HNF1A antibody. HNF1A protein was expressed in both the fetal and adult human ovaries. Based on these findings, we hypothesize that HNF1A participates in ovarian organogenesis and/or function and that mutations in the HNF1A gene might represent another molecular defect causing POF, possibly in combination with other genetic factors. The study underlines the importance of rare clinical paradigms in leading the way to elucidation of the pathogenetic mechanisms of rare diseases.
Collapse
Affiliation(s)
- P Xekouki
- Endocrine and Diabetes Clinic, University General Hospital of Heraklion, Medical School, University of Crete, 71500, Heraklion, Crete, Greece.
| | - A Konstantinidou
- 1st Department of Pathology, Unit of Perinatal Pathology, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - C Tatsi
- Division of Endocrinology, Diabetes and Metabolism, "Aghia Sophia" Children's Hospital ENDO-ERN Center for Rare Paediatric Endocrine Diseases, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - A Sertedaki
- Division of Endocrinology, Diabetes and Metabolism, "Aghia Sophia" Children's Hospital ENDO-ERN Center for Rare Paediatric Endocrine Diseases, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - N Settas
- Division of Endocrinology, Diabetes and Metabolism, "Aghia Sophia" Children's Hospital ENDO-ERN Center for Rare Paediatric Endocrine Diseases, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - D Loutradis
- 1st Department of Obstetrics and Gynecology, Alexandra Hospital, Athens University Medical School, Lourou 4-2, 115 28, Athens, Greece
| | - G P Chrousos
- Division of Endocrinology, Diabetes and Metabolism, "Aghia Sophia" Children's Hospital ENDO-ERN Center for Rare Paediatric Endocrine Diseases, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - C Kanaka-Gantenbein
- Division of Endocrinology, Diabetes and Metabolism, "Aghia Sophia" Children's Hospital ENDO-ERN Center for Rare Paediatric Endocrine Diseases, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - C Dacou-Voutetakis
- Division of Endocrinology, Diabetes and Metabolism, "Aghia Sophia" Children's Hospital ENDO-ERN Center for Rare Paediatric Endocrine Diseases, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - A Voutetakis
- Department of Pediatrics, University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
11
|
Bakhshalizadeh S, Afkhami F, Bell KM, Robevska G, van den Bergen J, Cronin S, Jaillard S, Ayers KL, Kumar P, Siebold C, Xiao Z, Tate EW, Danaei S, Farzadi L, Shahbazi S, Sinclair AH, Tucker EJ. Diverse genetic causes of amenorrhea in an ethnically homogeneous cohort and an evolving approach to diagnosis. Mol Cell Endocrinol 2024; 587:112212. [PMID: 38521400 DOI: 10.1016/j.mce.2024.112212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/12/2024] [Accepted: 03/16/2024] [Indexed: 03/25/2024]
Abstract
RESEARCH QUESTION Premature ovarian insufficiency (POI) is characterised by amenorrhea associated with elevated follicle stimulating hormone (FSH) under the age of 40 years and affects 1-3.7% women. Genetic factors explain 20-30% of POI cases, but most causes remain unknown despite genomic advancements. DESIGN We used whole exome sequencing (WES) in four Iranian families, validated variants via Sanger sequencing, and conducted the Acyl-cLIP assay to measure HHAT enzyme activity. RESULTS Despite ethnic homogeneity, WES revealed diverse genetic causes, including a novel homozygous nonsense variant in SYCP2L, impacting synaptonemal complex (SC) assembly, in the first family. Interestingly, the second family had two independent causes for amenorrhea - the mother had POI due to a novel homozygous loss-of-function variant in FANCM (required for chromosomal stability) and her daughter had primary amenorrhea due to a novel homozygous GNRHR (required for gonadotropic signalling) frameshift variant. WES analysis also provided cytogenetic insights. WES revealed one individual was in fact 46, XY and had a novel homozygous missense variant of uncertain significance in HHAT, potentially responsible for complete sex reversal although functional assays did not support impaired HHAT activity. In the remaining individual, WES indicated likely mosaic Turners with the majority of X chromosome variants having an allelic balance of ∼85% or ∼15%. Microarray validated the individual had 90% 45,XO. CONCLUSIONS This study demonstrates the diverse causes of amenorrhea in a small, isolated ethnic cohort highlighting how a genetic cause in one individual may not clarify familial cases. We propose that, in time, genomic sequencing may become a single universal test required for the diagnosis of infertility conditions such as POI.
Collapse
Affiliation(s)
- Shabnam Bakhshalizadeh
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Fateme Afkhami
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Katrina M Bell
- Department of Bioinformatics, Murdoch Children's Research Institute, Melbourne, Australia
| | | | | | - Sara Cronin
- Cyto-Molecular Diagnostic Research Laboratory, Victorian Clinical Genetics Services and Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052, Victoria, Australia
| | - Sylvie Jaillard
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France; CHU Rennes, Service de Cytogénétique et Biologie Cellulaire, F-35033, Rennes, France
| | - Katie L Ayers
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Pramod Kumar
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Zhangping Xiao
- Department of Chemistry, Imperial College London, 82 Wood Lane, London, W12 0BZ, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, 82 Wood Lane, London, W12 0BZ, UK
| | - Shahla Danaei
- Department of Obstetrics and Gynecology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Laya Farzadi
- Department of Obstetrics and Gynecology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Shahbazi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Andrew H Sinclair
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Elena J Tucker
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
12
|
Duan J, He XH, Li SJ, Xu HE. Cryo-electron microscopy for GPCR research and drug discovery in endocrinology and metabolism. Nat Rev Endocrinol 2024; 20:349-365. [PMID: 38424377 DOI: 10.1038/s41574-024-00957-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/29/2024] [Indexed: 03/02/2024]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors, with many GPCRs having crucial roles in endocrinology and metabolism. Cryogenic electron microscopy (cryo-EM) has revolutionized the field of structural biology, particularly regarding GPCRs, over the past decade. Since the first pair of GPCR structures resolved by cryo-EM were published in 2017, the number of GPCR structures resolved by cryo-EM has surpassed the number resolved by X-ray crystallography by 30%, reaching >650, and the number has doubled every ~0.63 years for the past 6 years. At this pace, it is predicted that the structure of 90% of all human GPCRs will be completed within the next 5-7 years. This Review highlights the general structural features and principles that guide GPCR ligand recognition, receptor activation, G protein coupling, arrestin recruitment and regulation by GPCR kinases. The Review also highlights the diversity of GPCR allosteric binding sites and how allosteric ligands could dictate biased signalling that is selective for a G protein pathway or an arrestin pathway. Finally, the authors use the examples of glycoprotein hormone receptors and glucagon-like peptide 1 receptor to illustrate the effect of cryo-EM on understanding GPCR biology in endocrinology and metabolism, as well as on GPCR-related endocrine diseases and drug discovery.
Collapse
Affiliation(s)
- Jia Duan
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Xin-Heng He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shu-Jie Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Department of Traditional Chinese Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
13
|
Hu S, Jing Y, Fu Y, Ye X. Association of FSHR gene polymorphisms with poor ovarian response in patients undergoing IVF: A systematic review and meta-analysis. Gene 2024; 909:148314. [PMID: 38412944 DOI: 10.1016/j.gene.2024.148314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/17/2024] [Accepted: 02/21/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND The results of studies on the association between polymorphisms in the FSHR gene and the risk of POR undergoing IVF have been inconsistent with each other, so we conducted a meta-analysis of all the available studies to explore the association between polymorphisms in the FSHR gene and the risk of POR. METHODS Literature that met the inclusion criteria was collected by searching six electronic databases and basic data from included studies were extracted. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to assess the strength of association between follicle-stimulating hormone receptor (FSHR) gene polymorphism and poor ovarian response (POR) risk. Begg's and Egger's tests were used to determine whether there was publication bias, and sensitivity analysis and TSA analysis were used to verify the stability and reliability of the results. RESULTS We included 24 articles, 22 of which explored rs6166, including 2,206 cases and 3,897 controls. 6 articles explored rs6165, including 444 cases and 875 controls. Under additive, heterozygote, and dominant models, rs6166 was significantly associated with POR (S vs. N: OR = 1.29, 95 % CI = 1.05-1.59, P = 0.017; NS vs. NN: OR = 1.33, 95 % CI = 1.02-1.74, P = 0.038; NS + SS vs. NN: OR = 1.38, 95 % CI = 1.04-1.84, P = 0.025). In ethnicity-based subgroup analyses, the additive, homozygote, heterozygote, and dominant models increased Asian POR risk. Among the five genetic models, rs6165 was significantly associated with POR (T vs. C: OR = 1.64, 95 % CI = 1.25-2.16, P = 0.000; TT vs. CC: OR = 2.76, 95 % CI = 1.43-5.32, P = 0.003; CT vs. CC: OR = 1.58, 95 % CI = 1.19-2.10, P = 0.001; TT vs. CC + CT: OR = 2.32, 95 % CI = 1.67-3.23, P = 0.000; CT + TT vs. CC: OR = 1.80, 95 % CI = 1.22-2.65, P = 0.003). In ethnicity-based subgroup analyses, all five genetic models increased the risk of POR in Caucasians. CONCLUSION According to the current meta-analysis, the rs6166 S allele was significantly associated with an increased risk of POR, especially in Asian populations. The rs6165 T allele was significantly associated with an increased risk of POR, especially in Caucasian populations.
Collapse
Affiliation(s)
- Siya Hu
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yunnan Jing
- Department of Acupuncture and Moxibustion, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yiman Fu
- Department of Obstetrics and Gynecology, Chongqing Jiangbei District Hospital of Traditional Chinese Medicine, Chongqing 400020, China
| | - Xiuying Ye
- Department of Obstetrics and Gynecology, Chongqing Jiangbei District Hospital of Traditional Chinese Medicine, Chongqing 400020, China.
| |
Collapse
|
14
|
Persico T, Tranquillo ML, Seracchioli R, Zuccarello D, Sorrentino U. PGT-M for Premature Ovarian Failure Related to CGG Repeat Expansion of the FMR1 Gene. Genes (Basel) 2023; 15:6. [PMID: 38275588 PMCID: PMC10815814 DOI: 10.3390/genes15010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Primary ovarian failure (POF) is caused by follicle exhaustion and is associated with menstrual irregularities and elevated gonadotropin levels, which lead to infertility before the age of 40 years. The etiology of POI is mostly unknown, but a heterogeneous genetic and familial background can be identified in a subset of cases. Abnormalities in the fragile X mental retardation 1 gene (FMR1) are among the most prevalent monogenic causes of POI. These abnormalities are caused by the expansion of an unstable CGG repeat in the 5' untranslated region of FMR1. Expansions over 200 repeats cause fragile X syndrome (FXS), whereas expansions between 55 and 200 CGG repeats, which are defined as a fragile X premutation, have been associated with premature ovarian failure type 1 (POF1) in heterozygous females. Preimplantation genetic testing for monogenic diseases (PGT-M) can be proposed when the female carries a premutation or a full mutation. In this narrative review, we aim to recapitulate the clinical and molecular features of POF1 and their implications in the context of PGT-M.
Collapse
Affiliation(s)
- Tiziana Persico
- Medically Assisted Procreation Center, Maternal and Child Department, Beauregard Hospital, Valle D’Aosta Local Public Health, 11100 Aoste, Italy
| | - Maria Lucrezia Tranquillo
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (M.L.T.); (R.S.)
| | - Renato Seracchioli
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy; (M.L.T.); (R.S.)
- Division of Gynaecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero, University of Bologna, 40138 Bologna, Italy
| | - Daniela Zuccarello
- Clinical Genetics and Epidemiology Unit, University of Padova, 35128 Padova, Italy; (D.Z.); (U.S.)
| | - Ugo Sorrentino
- Clinical Genetics and Epidemiology Unit, University of Padova, 35128 Padova, Italy; (D.Z.); (U.S.)
| |
Collapse
|
15
|
Verrilli L. Primary Ovarian Insufficiency and Ovarian Aging. Obstet Gynecol Clin North Am 2023; 50:653-661. [PMID: 37914485 DOI: 10.1016/j.ogc.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Primary ovarian insufficiency (POI) is a complex condition of aberrant ovarian aging. POI etiologies are varied, and most cases have no identifiable underlying cause. Caring for women with POI requires an approach that understands the importance of ovarian function in a variety of target organs and tissues.
Collapse
Affiliation(s)
- Lauren Verrilli
- Department of Obstetrics and Gynecology, University of Utah School of Medicine, 50 North Medical Drive, Salt Lake City, UT, USA.
| |
Collapse
|
16
|
Martirosyan YO, Silachev DN, Nazarenko TA, Birukova AM, Vishnyakova PA, Sukhikh GT. Stem-Cell-Derived Extracellular Vesicles: Unlocking New Possibilities for Treating Diminished Ovarian Reserve and Premature Ovarian Insufficiency. Life (Basel) 2023; 13:2247. [PMID: 38137848 PMCID: PMC10744991 DOI: 10.3390/life13122247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/18/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
Despite advancements in assisted reproductive technology (ART), achieving successful pregnancy rates remains challenging. Diminished ovarian reserve and premature ovarian insufficiency hinder IVF success-about 20% of in vitro fertilization (IVF) patients face a poor prognosis due to a low response, leading to higher cancellations and reduced birth rates. In an attempt to address the issue of premature ovarian insufficiency (POI), we conducted systematic PubMed and Web of Science research, using keywords "stem cells", "extracellular vesicles", "premature ovarian insufficiency", "diminished ovarian reserve" and "exosomes". Amid the complex ovarian dynamics and challenges like POI, stem cell therapy and particularly the use of extracellular vesicles (EVs), a great potential is shown. EVs trigger paracrine mechanisms via microRNAs and bioactive molecules, suppressing apoptosis, stimulating angiogenesis and activating latent regenerative potential. Key microRNAs influence estrogen secretion, proliferation and apoptosis resistance. Extracellular vesicles present a lot of possibilities for treating infertility, and understanding their molecular mechanisms is crucial for maximizing EVs' therapeutic potential in addressing ovarian disorders and promoting reproductive health.
Collapse
Affiliation(s)
- Yana O. Martirosyan
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (T.A.N.); (A.M.B.); (P.A.V.); (G.T.S.)
| | - Denis N. Silachev
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (T.A.N.); (A.M.B.); (P.A.V.); (G.T.S.)
- Department of Functional Biochemistry of Biopolymers, A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
| | - Tatiana A. Nazarenko
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (T.A.N.); (A.M.B.); (P.A.V.); (G.T.S.)
| | - Almina M. Birukova
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (T.A.N.); (A.M.B.); (P.A.V.); (G.T.S.)
| | - Polina A. Vishnyakova
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (T.A.N.); (A.M.B.); (P.A.V.); (G.T.S.)
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Gennadiy T. Sukhikh
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (T.A.N.); (A.M.B.); (P.A.V.); (G.T.S.)
| |
Collapse
|
17
|
Moses E, Franek R, Harel I. A scalable and tunable platform for functional interrogation of peptide hormones in fish. eLife 2023; 12:e85960. [PMID: 37872843 PMCID: PMC10597582 DOI: 10.7554/elife.85960] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 09/16/2023] [Indexed: 10/25/2023] Open
Abstract
Pituitary hormones play a central role in shaping vertebrate life history events, including growth, reproduction, metabolism, and aging. The regulation of these traits often requires precise control of hormone levels across diverse timescales. However, fine tuning circulating hormones in-vivo has traditionally been experimentally challenging. Here, using the naturally short-lived turquoise killifish (N. furzeri), we describe a high-throughput platform that combines loss- and gain-of-function of peptide hormones. Mutation of three primary pituitary hormones, growth hormone (gh1), follicle stimulating hormone (fshb), and thyroid stimulating hormone (tshb), alters somatic growth and reproduction. Thus, suggesting that while the killifish undergoes extremely rapid growth and maturity, it still relies on vertebrate-conserved genetic networks. As the next stage, we developed a gain-of-function vector system in which a hormone is tagged using a self-cleavable fluorescent reporter, and ectopically expressed in-vivo through intramuscular electroporation. Following a single electroporation, phenotypes, such as reproduction, are stably rescued for several months. Notably, we demonstrate the versatility of this approach by using multiplexing, dose-dependent, and doxycycline-inducible systems to achieve tunable and reversible expression. In summary, this method is relatively high-throughput, and facilitates large-scale interrogation of life-history strategies in fish. Ultimately, this approach could be adapted for modifying aquaculture species and exploring pro-longevity interventions.
Collapse
Affiliation(s)
- Eitan Moses
- Department of Genetics, the Silberman Institute, The Hebrew University of JerusalemJerusalemIsrael
| | - Roman Franek
- Department of Genetics, the Silberman Institute, The Hebrew University of JerusalemJerusalemIsrael
- University of South Bohemia in Ceske Budejovice, South Bohemian Research Center of Aquaculture and Biodiversity of HydrocenosesVodnanyCzech Republic
| | - Itamar Harel
- Department of Genetics, the Silberman Institute, The Hebrew University of JerusalemJerusalemIsrael
| |
Collapse
|
18
|
Kaur M, Singh S, Kaur A. Polymorphisms in FSHR modulating susceptibility to polycystic ovary syndrome: an updated meta-analysis. J Ovarian Res 2023; 16:183. [PMID: 37653412 PMCID: PMC10472705 DOI: 10.1186/s13048-023-01238-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/13/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Two polymorphisms, rs6165 and rs6166 located in the intracellular domain of FSHR has been reported to affect folliculogenesis, steroidogenesis and oocyte maturation. Several studies have highlighted the role of FSHR polymorphisms in PCOS but the findings are conflicting. A meta-analysis was carried out to decipher the emerging perspectives. METHODOLOGY A comprehensive literature search was made using PubMed, PCOSkb, and Google Scholar. New Ottawa Scale has been utilized to evaluate the quality of each article. To evaluate the strength of association under different genetic models of rs6165 and rs6166 polymorphisms, odds ratio with a 95% confidence interval (CI) was calculated. RESULTS A total of 20 articles were selected for the present study. In pooled analysis and after the stratification by ethnicity, polymorphism rs6165 remains unrelated to the onset of PCOS. Besides, rs6166 exhibits significant protection in the Indian population under recessive, additive, and allele models (OR = 0.7, CI: 0.54-0.9, p = 0.006, OR = 0.65, CI: 0.48-0.89, p = 0.006, OR = 0.82, CI: 0.7-0.95, p = 0.01, respectively) and low to moderate risk in the Caucasian population under allele model (OR = 1.17, CI: 1.04-1.32, p = 0.01). CONCLUSION This meta-analysis suggests that GG genotype of rs6166 provides protection against PCOS, in a population-specific manner.
Collapse
Affiliation(s)
- Mandeep Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, 143005, India
| | - Sukhjashanpreet Singh
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, 143005, India
| | - Anupam Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| |
Collapse
|
19
|
Yang X, Yang L. Current understanding of the genomic abnormities in premature ovarian failure: chance for early diagnosis and management. Front Med (Lausanne) 2023; 10:1194865. [PMID: 37332766 PMCID: PMC10274511 DOI: 10.3389/fmed.2023.1194865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Premature ovarian failure (POF) is an insidious cause of female infertility and a devastating condition for women. POF also has a strong familial and heterogeneous genetic background. Management of POF is complicated by the variable etiology and presentation, which are generally characterized by abnormal hormone levels, gene instability and ovarian dysgenesis. To date, abnormal regulation associated with POF has been found in a small number of genes, including autosomal and sex chromosomal genes in folliculogenesis, granulosa cells, and oocytes. Due to the complex genomic contributions, ascertaining the exact causative mechanisms has been challenging in POF, and many pathogenic genomic characteristics have yet to be elucidated. However, emerging research has provided new insights into genomic variation in POF as well as novel etiological factors, pathogenic mechanisms and therapeutic intervention approaches. Meanwhile, scattered studies of transcriptional regulation revealed that ovarian cell function also depends on specific biomarker gene expression, which can influence protein activities, thus causing POF. In this review, we summarized the latest research and issues related to the genomic basis for POF and focused on insights gained from their biological effects and pathogenic mechanisms in POF. The present integrated studies of genomic variants, gene expression and related protein abnormalities were structured to establish the role of etiological genes associated with POF. In addition, we describe the design of some ongoing clinical trials that may suggest safe, feasible and effective approaches to improve the diagnosis and therapy of POF, such as Filgrastim, goserelin, resveratrol, natural plant antitoxin, Kuntai capsule et al. Understanding the candidate genomic characteristics in POF is beneficial for the early diagnosis of POF and provides appropriate methods for prevention and drug treatment. Additional efforts to clarify the POF genetic background are necessary and are beneficial for researchers and clinicians regarding genetic counseling and clinical practice. Taken together, recent genomic explorations have shown great potential to elucidate POF management in women and are stepping from the bench to the bedside.
Collapse
Affiliation(s)
- Xu Yang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lin Yang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Ko EJ, Shin JE, Lee JY, Ryu CS, Hwang JY, Kim YR, Ahn EH, Kim JH, Kim NK. Association of Polymorphisms in FSHR, INHA, ESR1, and BMP15 with Recurrent Implantation Failure. Biomedicines 2023; 11:biomedicines11051374. [PMID: 37239044 DOI: 10.3390/biomedicines11051374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/29/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Recurrent implantation failure (RIF) refers to two or more unsuccessful in vitro fertilization embryo transfers in the same individual. Embryonic characteristics, immunological factors, and coagulation factors are known to be the causes of RIF. Genetic factors have also been reported to be involved in the occurrence of RIF, and some single nucleotide polymorphisms (SNPs) may contribute to RIF. We examined SNPs in FSHR, INHA, ESR1, and BMP15, which have been associated with primary ovarian failure. A cohort of 133 RIF patients and 317 healthy controls consisting of all Korean women was included. Genotyping was performed by Taq-Man genotyping assays to determine the frequency of the following polymorphisms: FSHR rs6165, INHA rs11893842 and rs35118453, ESR1 rs9340799 and rs2234693, and BMP15 rs17003221 and rs3810682. The differences in these SNPs were compared between the patient and control groups. Our results demonstrate a decreased prevalence of RIF in subjects with the FSHR rs6165 A>G polymorphism [AA vs. AG adjusted odds ratio (AOR) = 0.432; confidence interval (CI) = 0.206-0.908; p = 0.027, AA+AG vs. GG AOR = 0.434; CI = 0.213-0.885; p = 0.022]. Based on a genotype combination analysis, the GG/AA (FSHR rs6165/ESR1 rs9340799: OR = 0.250; CI = 0.072-0.874; p = 0.030) and GG-CC (FSHR rs6165/BMP15 rs3810682: OR = 0.466; CI = 0.220-0.987; p = 0.046) alleles were also associated with a decreased RIF risk. Additionally, the FSHR rs6165GG and BMP15 rs17003221TT+TC genotype combination was associated with a decreased RIF risk (OR = 0.430; CI = 0.210-0.877; p = 0.020) and increased FSH levels, as assessed by an analysis of variance. The FSHR rs6165 polymorphism and genotype combinations are significantly associated with RIF development in Korean women.
Collapse
Affiliation(s)
- Eun-Ju Ko
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea
| | - Ji-Eun Shin
- Department of Obstetrics and Gynecology, Fertility Center of CHA Bundang Medical Center, CHA University, Seongnam 13520, Republic of Korea
| | - Jung-Yong Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea
| | - Chang-Soo Ryu
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea
| | - Ji-Young Hwang
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul 06125, Republic of Korea
| | - Young-Ran Kim
- Department of Obstetrics and Gynecology, Fertility Center of CHA Bundang Medical Center, CHA University, Seongnam 13520, Republic of Korea
| | - Eun-Hee Ahn
- Department of Obstetrics and Gynecology, Fertility Center of CHA Bundang Medical Center, CHA University, Seongnam 13520, Republic of Korea
| | - Ji-Hyang Kim
- Department of Obstetrics and Gynecology, Fertility Center of CHA Bundang Medical Center, CHA University, Seongnam 13520, Republic of Korea
| | - Nam-Keun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Republic of Korea
| |
Collapse
|
21
|
Zeng Y, Li L, Li Q, Hu J, Zhang N, Wu L, Yan Z, Qu R, Dong J, Liu R, Choy KW, Wang L, Sang Q, Guan Y, Chen B. Genetic screening in patients with ovarian dysfunction. Clin Genet 2023; 103:352-357. [PMID: 36373164 DOI: 10.1111/cge.14267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/15/2022]
Abstract
Ovarian dysfunction, including premature ovarian insufficiency and decreased ovarian reserve, affects the ovarian reserve and is one of the leading causes of female infertility. More and more cases of ovarian dysfunction are associated with genetic factors. Here, we identified eight potential variants in five genes (MSH4, HFM1, SYCE1, FSHR, and C14orf39) from six independent families by exome sequencing. The splice-site variants in SYCE1 and MSH4 affected canonical splicing isoforms, leading to missing protein domains or premature termination. Our findings expand the mutational spectrum of ovarian dysfunction and provide potential biomarkers for future genetic counseling and for more personalized treatments. Exome sequencing was shown to be a useful tool to better dissect the genetic basis for ovarian dysfunction and yielded a genetic diagnosis in about 5.0% (6/124) of cases in a cohort of 124 patients with ovarian dysfunction.
Collapse
Affiliation(s)
- Yang Zeng
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Lin Li
- Key Laboratory of Human Reproduction and Genetics, Department of Reproductive Medicine, Nanchang Reproductive Hospital, Nanchang, Jiangxi, China
| | - Qingchun Li
- Department of Reproductive Medicine, Binzhou Medical University Hospital, Binzhou, China
| | - Jijun Hu
- Department of Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nana Zhang
- Department of Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ling Wu
- The Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Yan
- The Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ronggui Qu
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Jie Dong
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Ruyi Liu
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Kwong Wai Choy
- Department of Obsterics and Gynecology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
- Zhuhai Fudan Innovation Institute, Zhuhai, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
- Zhuhai Fudan Innovation Institute, Zhuhai, China
| | - Yichun Guan
- Department of Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| |
Collapse
|
22
|
Suzuki E, Miyado M, Kuroki Y, Fukami M. Genetic variants of G-protein coupled receptors associated with pubertal disorders. Reprod Med Biol 2023; 22:e12515. [PMID: 37122876 PMCID: PMC10134480 DOI: 10.1002/rmb2.12515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/02/2023] [Accepted: 04/11/2023] [Indexed: 05/02/2023] Open
Abstract
Background The human hypothalamic-pituitary-gonadal (HPG) axis is the regulatory center for pubertal development. This axis involves six G-protein coupled receptors (GPCRs) encoded by KISS1R, TACR3, PROKR2, GNRHR, LHCGR, and FSHR. Methods Previous studies have identified several rare variants of the six GPCR genes in patients with pubertal disorders. In vitro assays and animal studies have provided information on the function of wild-type and variant GPCRs. Main Findings Of the six GPCRs, those encoded by KISS1R and TACR3 are likely to reside at the top of the HPG axis. Several loss-of-function variants in the six genes were shown to cause late/absent puberty. In particular, variants in KISS1R, TACR3, PROKR2, and GNRHR lead to hypogonadotropic hypogonadism in autosomal dominant, recessive, and oligogenic manners. Furthermore, a few gain-of-function variants of KISS1R, PROKR2, and LHCGR have been implicated in precocious puberty. The human HPG axis may contain additional GPCRs. Conclusion The six GPCRs in the HPG axis govern pubertal development through fine-tuning of hormone secretion. Rare sequence variants in these genes jointly account for a certain percentage of genetic causes of pubertal disorders. Still, much remains to be clarified about the molecular network involving the six GPCRs.
Collapse
Affiliation(s)
- Erina Suzuki
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Mami Miyado
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
- Department of Food and NutritionBeppu UniversityOitaJapan
| | - Yoko Kuroki
- Department of Genome Medicine, National Center for Child Health and DevelopmentTokyoJapan
- Division of Collaborative Research, National Center for Child Health and DevelopmentTokyoJapan
- Division of Diversity ResearchNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Maki Fukami
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
- Division of Diversity ResearchNational Research Institute for Child Health and DevelopmentTokyoJapan
| |
Collapse
|
23
|
Fetal germ cell development in humans, a link with infertility. Semin Cell Dev Biol 2022; 131:58-65. [PMID: 35431137 DOI: 10.1016/j.semcdb.2022.03.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 12/14/2022]
Abstract
Gametes are cells that have the unique ability to give rise to new individuals as well as transmit (epi)genetic information across generations. Generation of functionally competent gametes, oocytes and sperm cells, depends to some extent on several fundamental processes that occur during fetal development. Direct studies on human fetal germ cells remain hindered by ethical considerations and inaccessibility to human fetal material. Therefore, the majority of our current knowledge of germ cell development still comes from an invaluable body of research performed using different mammalian species. During the last decade, our understanding of human fetal germ cells has increased due to the successful use of human pluripotent stem cells to model aspects of human early gametogenesis and advancements on single-cell omics. Together, this has contributed to determine the cell types and associated molecular signatures in the developing human gonads. In this review, we will put in perspective the knowledge obtained from several mammalian models (mouse, monkey, pig). Moreover, we will discuss the main events during human fetal (female) early gametogenesis and how the dysregulation of this highly complex and lengthy process can link to infertility later in life.
Collapse
|
24
|
Mu Z, Shen S, Lei L. Resistant ovary syndrome: Pathogenesis and management strategies. Front Med (Lausanne) 2022; 9:1030004. [PMCID: PMC9626816 DOI: 10.3389/fmed.2022.1030004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
Abstract
Resistant ovary syndrome (ROS) is a rare and difficult gynecological endocrine disorder that poses a serious risk to women’s reproductive health. The clinical features are normal sex characteristics, regular female karyotype, and usual ovarian reserve, but elevated endogenous gonadotropin levels and low estrogen levels with primary or secondary amenorrhea. Although there have been many case reports of the disease over the past 50 years, the pathogenesis of the disease is still poorly understood, and there are still no effective clinical management strategies. In this review, we have collected all the current reports on ROS and summarized the pathogenesis and treatment strategies for this disease, intending to provide some clinical references for the management and treatment of this group of patients and provide the foothold for future studies.
Collapse
|
25
|
Saengkaew T, Howard SR. Genetics of pubertal delay. Clin Endocrinol (Oxf) 2022; 97:473-482. [PMID: 34617615 PMCID: PMC9543006 DOI: 10.1111/cen.14606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022]
Abstract
The timing of pubertal development is strongly influenced by the genetic background, and clinical presentations of delayed puberty are often found within families with clear patterns of inheritance. The discovery of the underlying genetic regulators of such conditions, in recent years through next generation sequencing, has advanced the understanding of the pathogenesis of disorders of pubertal timing and the potential for genetic testing to assist diagnosis for patients with these conditions. This review covers the significant advances in the understanding of the biological mechanisms of delayed puberty that have occurred in the last two decades.
Collapse
Affiliation(s)
- Tansit Saengkaew
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Endocrinology Unit, Department of Paediatrics, Faculty of MedicinePrince of Songkla UniversitySongkhlaThailand
| | - Sasha R. Howard
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
26
|
McGlacken-Byrne SM, Achermann JC, Conway GS. Management of a Girl With Delayed Puberty and Elevated Gonadotropins. J Endocr Soc 2022; 6:bvac108. [PMID: 35935072 PMCID: PMC9351373 DOI: 10.1210/jendso/bvac108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Indexed: 11/19/2022] Open
Abstract
A girl presenting with delayed puberty and elevated gonadotropins may have a range of conditions such as Turner syndrome (TS), primary ovarian insufficiency (POI), and 46,XY disorders of sexual development (DSD). An organized and measured approach to investigation can help reach a timely diagnosis. Management of young people often requires specialist multidisciplinary input to address the endocrine and nonendocrine features of these complex conditions, as well as the psychological challenges posed by their diagnosis. Next-generation sequencing within the research setting has revealed several genetic causes of POI and 46,XY DSD, which may further facilitate an individualized approach to care of these young people in the future. Pubertal induction is required in many and the timing of this may need to be balanced with other issues specific to the condition (eg, allowing time for information-sharing in 46,XY DSD, optimizing growth in TS). Shared decision-making and sign-posting to relevant support groups from the outset can help empower young people and their families to manage these conditions. We describe 3 clinical vignettes of girls presenting with delayed puberty and hypergonadotropic amenorrhea and discuss their clinical management in the context of current literature and guidelines.
Collapse
Affiliation(s)
- Sinéad M McGlacken-Byrne
- Institute for Women’s Health, University College London, London WC1E 6AU, UK
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Department of Paediatric Endocrinology, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - John C Achermann
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Gerard S Conway
- Institute for Women’s Health, University College London, London WC1E 6AU, UK
| |
Collapse
|
27
|
Liquiritin ameliorates metabolic and endocrine alterations in a mouse model of polycystic ovary syndrome. REPRODUCTIVE AND DEVELOPMENTAL MEDICINE 2022. [DOI: 10.1097/rd9.0000000000000025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
28
|
Silvén H, Savukoski SM, Pesonen P, Pukkala E, Gissler M, Suvanto E, Niinimäki M. Incidence and familial risk of premature ovarian insufficiency in the Finnish female population. Hum Reprod 2022; 37:1030-1036. [PMID: 35134918 PMCID: PMC9071220 DOI: 10.1093/humrep/deac014] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/21/2021] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION What is the incidence of premature ovarian insufficiency (POI), has the incidence of POI changed over time, and what is the risk of POI among relatives of POI women? SUMMARY ANSWER The incidence of POI increased among females aged 15-19 years from 2007 onwards and decreased in older age groups, and among relatives of women with POI the risk of POI is significantly increased. WHAT IS KNOWN ALREADY So far, there has been no good quality, nationwide studies of the incidence of POI. Early menopause has been associated with the elevated risk of early menopause among relatives, but the knowledge of the familial risk of POI is scarce. Lower socioeconomic status has been associated with lower age at natural menopause. STUDY DESIGN, SIZE, DURATION Population-based study with 5011 women diagnosed with POI in 1988-2017. The data were collected from national registries and covers POI subjects in entire Finland. PARTICIPANTS/MATERIALS, SETTING, METHODS Women with hormone replacement therapy reimbursement for POI were identified from Social Insurance Institution (SII). We calculated POI incidence in different age groups and studied the changes in the incidence rate over time in 5-year segments. Four population-based controls were selected from the Digital and Population Data Services Agency (DVV) for each POI woman. Family members of the POI cases and controls were identified from the DVV and linked to SII reimbursement data to identify POI diagnoses among them. The familial risk of POI was estimated with a logistical regression model. MAIN RESULTS AND THE ROLE OF CHANCE The incidence was highest in the 35-39 age group, ranging from 73.8/100 000 women-years in 1993-1997 to 39.9/100 000 women-years in 2013-2017. From 2007, the incidence among 15- to 19-year-olds rose from 7.0 to 10.0/100 000 women-years in 2015-2017. Cumulative incidence of POI for women under 40 years in 1988-2017 was 478/100 000 women. The relative risk of POI among relatives of women with POI was 4.6 (95% CI 3.3-6.5) compared to relatives of women without POI. POI women tended to have slightly lower socioeconomic status and level of education compared to controls. LIMITATIONS, REASONS FOR CAUTION For some women with POI, diagnosis or reimbursement may be lacking. However, we presume that these women represent a minority due to the nature of the disease and the economic benefits of reimbursement. Some changes in the incidence of POI can reflect changes in clinical practice and changing treatments and reimbursement criteria. WIDER IMPLICATIONS OF THE FINDINGS The risk of developing POI is significantly higher in women who have first-degree relatives diagnosed with POI. Raising awareness of the increased risk might lead to earlier diagnosis and initiation of hormonal replacement therapy, possibly preventing adverse effects of low oestrogen levels, such as osteoporosis. STUDY FUNDING/COMPETING INTEREST(S) This work was financially supported by the Oulu University Hospital. H.S. received a grant from Finnish Menopause Society. S.M.S. received a grant from the Finnish Menopause Society, the Finnish Medical Foundation and the Juho Vainio Foundation. The authors do not have any competing interests to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- H Silvén
- Department of Obstetrics and Gynecology, Oulu University Hospital, Oulu, Finland
- PEDEGO Research Unit, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - S M Savukoski
- Department of Obstetrics and Gynecology, Oulu University Hospital, Oulu, Finland
- PEDEGO Research Unit, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - P Pesonen
- Northern Finland Birth Cohorts, Arctic Biobank, Infrastructure for Population Studies, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - E Pukkala
- Faculty of Social Sciences, Tampere University, Tampere, Finland
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland
| | - M Gissler
- Information Services Department, THL Finnish Institute for Health and Welfare, Helsinki, Finland
- Academic Primary Health Care Centre, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, 171 76, Karolinska Institute, Stockholm, Sweden
| | - E Suvanto
- Department of Obstetrics and Gynecology, Oulu University Hospital, Oulu, Finland
- PEDEGO Research Unit, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - M Niinimäki
- Department of Obstetrics and Gynecology, Oulu University Hospital, Oulu, Finland
- PEDEGO Research Unit, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
29
|
Li H, Chang T, Mu H, Xiang W. Case report: Birth achieved after effective ovarian stimulation combined with dexamethasone in a patient with resistant ovary syndrome. J Ovarian Res 2022; 15:42. [PMID: 35392972 PMCID: PMC8988423 DOI: 10.1186/s13048-022-00976-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 03/30/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Resistant ovary syndrome (ROS) is a rare endocrine disorder and there have been few reports of live births by affected patients. As gonadotropin resistance leads immature oocytes, some researchers reported few live births with in vitro maturation (IVM) of oocytes, but IVM is not always successful in ROS patients. Here, we report an original case of ROS, associated with Ig-FSHR in the serum, who achieved a live birth following ovarian stimulation combined with dexamethasone treatment. CASE PRESENTATION The 30-year-old woman presented with secondary amenorrhea and infertility. Her serum FSH levels were found to be higher than normal, but in discordance with a normal anti-Müllerian hormone (AMH) level and antral follicle count. Genetic investigation found no mutations potentially affecting FSHR. With reference of previous ROS studies, the patient's serum was analyzed for antibodies directed against FSHR and dot blot analysis showed strong reactivity with FSHR. Then, dexamethasone was proposed to the patient, and she successfully became pregnant, finally delivering a healthy girl by caesarean section. CONCLUSION To our best knowledge, this is the first report of the successful treatment of ROS using ovarian stimulation combined with dexamethasone. In some cases of ROS, high doses of exogenous gonadotropins in combination with immunosuppressive therapy could be an effective approach.
Collapse
Affiliation(s)
- Huiying Li
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tianli Chang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongbei Mu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenpei Xiang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China. .,Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
30
|
Bai RQ, He WB, Peng Q, Shen SH, Yu QQ, Du J, Tan YQ, Wang YH, Liu BJ. A novel FAM83H variant causes familial amelogenesis imperfecta with incomplete penetrance. Mol Genet Genomic Med 2022; 10:e1902. [PMID: 35212465 PMCID: PMC9000937 DOI: 10.1002/mgg3.1902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/14/2022] [Accepted: 01/31/2022] [Indexed: 11/26/2022] Open
Abstract
Background Amelogenesis imperfecta (AI) is known to be a monogenic genetic disease caused by a variety of genes demonstrating a wide spectrum of penetrance. FAM83H is reported to be involved in AI: however, whether FAM83H causes AI with incomplete penetrance is unclear. Methods Whole‐exome sequencing was performed on two patients with AI, and putative disease‐related variants were validated by Sanger sequencing. Bioinformatic and in vitro functional analyses were performed to functionally characterize the identified disease‐causing variants. Results We identified a novel heterozygous nonsense variant of FAM83H (NM_198488: c.1975G > T, p.Glu659Ter); in vitro functional analysis showed that this mutant produced mislocalized proteins and was deleterious. Surprisingly, the clinical manifestations of each of the six individuals carrying this variant were different, with one carrier appearing to be completely asymptomatic for AI. Conclusion Our findings expand the variant spectrum for FAM83H and the phenotypic spectrum for FAM83H‐associated AI and suggest that FAM83H‐mediated AI exhibits incomplete penetrance.
Collapse
Affiliation(s)
- Rui-Qi Bai
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, People's Republic of China
| | - Wen-Bin He
- National Engineering and Research Center of Human Stem Cells, Changsha Hunan, People's Republic of China.,Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, China.,Hunan International Scientific and Technological Cooperation base of Development and carcinogenesis, Changsha, China
| | - Qian Peng
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, People's Republic of China
| | - Su-Hui Shen
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, People's Republic of China
| | - Qian-Qian Yu
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, People's Republic of China
| | - Juan Du
- National Engineering and Research Center of Human Stem Cells, Changsha Hunan, People's Republic of China.,Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, China.,Hunan International Scientific and Technological Cooperation base of Development and carcinogenesis, Changsha, China
| | - Yue-Qiu Tan
- National Engineering and Research Center of Human Stem Cells, Changsha Hunan, People's Republic of China.,Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, China.,Hunan International Scientific and Technological Cooperation base of Development and carcinogenesis, Changsha, China
| | - Yue-Hong Wang
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, People's Republic of China
| | - Bin-Jie Liu
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
31
|
Issa NM, Elhady GM. Cytogenetic abnormalities in a sample of females with premature ovarian failure. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2022. [DOI: 10.1186/s43043-022-00098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Premature ovarian failure (POF) is a complex heterogeneous disorder characterized by the triad of amenorrhea, hypergonadotropinism, and hypoestrogenism in women before the expected age of menopause. In most POF patients, the etiology is idiopathic. X chromosome abnormalities are known to be responsible for many POF cases but the effect of sex chromosome low level mosaicism on ovarian function still remains unclear. The aim of this study was to investigate the prevalence and type of cytogenetic abnormalities as well as low-level sex chromosome mosaicism in Egyptian females with POF.
Results
The present study recruited thirty women with POF and thirty women with normal reproductive history as a control group. Conventional cytogenetic analysis was carried out on POF patients in order to detect cytogenetic abnormalities. FISH on interphase and metaphase nuclei from patients with normal karyotype as well as from thirty control women with normal reproductive history was performed using X, Y, and 18 centromeric probes to evaluate low-level sex chromosome mosaicism. Conventional cytogenetic analysis of peripheral blood lymphocytes demonstrated chromosomal aberrations in 7 cases. FISH revealed that the rate of X chromosome mosaicism was significantly higher in POF patients than in the control group.
Conclusion
We concluded that X chromosome abnormalities including low level mosaicism may be underlying the pathology of POF as increased mosaicism may lead to accelerated oocyte aging and premature follicular atresia.
Collapse
|
32
|
Conforti A, Tüttelmann F, Alviggi C, Behre HM, Fischer R, Hu L, Polyzos NP, Chuderland D, Rama Raju GA, D’Hooghe T, Simoni M, Sunkara SK, Longobardi S. Effect of Genetic Variants of Gonadotropins and Their Receptors on Ovarian Stimulation Outcomes: A Delphi Consensus. Front Endocrinol (Lausanne) 2022; 12:797365. [PMID: 35178027 PMCID: PMC8844496 DOI: 10.3389/fendo.2021.797365] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/15/2021] [Indexed: 01/20/2023] Open
Abstract
Background A Delphi consensus was conducted to evaluate the influence of single nucleotide polymorphisms (SNPs) in genes encoding gonadotropin and gonadotropin receptors on clinical ovarian stimulation outcomes following assisted reproductive technology (ART) treatment. Methods Nine experts plus two Scientific Coordinators discussed and amended statements plus supporting references proposed by the Scientific Coordinators. The statements were distributed via an online survey to 36 experts, who voted on their level of agreement or disagreement with each statement. Consensus was reached if the proportion of participants agreeing or disagreeing with a statement was >66%. Results Eleven statements were developed, of which two statements were merged. Overall, eight statements achieved consensus and two statements did not achieve consensus. The statements reaching consensus are summarized here. (1) SNP in the follicle stimulating hormone receptor (FSHR), rs6166 (c.2039A>G, p.Asn680Ser) (N=5 statements): Ser/Ser carriers have higher basal FSH levels than Asn/Asn carriers. Ser/Ser carriers require higher amounts of gonadotropin during ovarian stimulation than Asn/Asn carriers. Ser/Ser carriers produce fewer oocytes during ovarian stimulation than Asn/Asn or Asn/Ser carriers. There is mixed evidence supporting an association between this variant and ovarian hyperstimulation syndrome. (2) SNP of FSHR, rs6165 (c.919G>A, p.Thr307Ala) (N=1 statement): Few studies suggest Thr/Thr carriers require a shorter duration of gonadotropin stimulation than Thr/Ala or Ala/Ala carriers. (3) SNP of FSHR, rs1394205 (-29G>A) (N=1 statement): Limited data in specific ethnic groups suggest that A/A allele carriers may require higher amounts of gonadotropin during ovarian stimulation and produce fewer oocytes than G/G carriers. (4) SNP of FSH β-chain (FSHB), rs10835638 (-211G>T) (N=1 statement): There is contradictory evidence supporting an association between this variant and basal FSH levels or oocyte number. (5) SNPs of luteinizing hormone β-chain (LHB) and LH/choriogonadotropin receptor (LHCGR) genes (N=1 statement): these may influence ovarian stimulation outcomes and could represent potential future targets for pharmacogenomic research in ART, although data are still very limited. Conclusions This Delphi consensus provides clinical perspectives from a diverse international group of experts. The consensus supports a link between some variants in gonadotropin/gonadotropin receptor genes and ovarian stimulation outcomes; however, further research is needed to clarify these findings.
Collapse
Affiliation(s)
- Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University Federico II, Naples, Italy
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Carlo Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, University Federico II, Naples, Italy
| | - Hermann M. Behre
- Center for Reproductive Medicine and Andrology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Robert Fischer
- Department of Gynecological Endocrinology and Reproductive Medicine, Fertility Center Hamburg, Hamburg, Germany
| | - Liang Hu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Nikolaos P. Polyzos
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain
| | - Dana Chuderland
- Global Medical Affairs Fertility, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Thomas D’Hooghe
- Global Medical Affairs Fertility, Merck Healthcare KGaA, Darmstadt, Germany
- Research Group Reproductive Medicine, Department of Development and Regeneration, Organ Systems, Group Biomedical Sciences, KU Leuven (University of Leuven), Leuven, Belgium
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sesh K. Sunkara
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Salvatore Longobardi
- Global Clinical Development, Research and Development, Merck KGaA, Darmstadt, Germany
| |
Collapse
|
33
|
Chen X, Chen L, Wang Y, Shu C, Zhou Y, Wu R, Jin B, Yang L, Sun J, Qi M, Shu J. Identification and characterization of novel compound heterozygous variants in FSHR causing primary ovarian insufficiency with resistant ovary syndrome. Front Endocrinol (Lausanne) 2022; 13:1013894. [PMID: 36704038 PMCID: PMC9871476 DOI: 10.3389/fendo.2022.1013894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Primary ovarian insufficiency (POI) is among the foremost causes of women infertility due to premature partial or total loss of ovarian function. Resistant ovary syndrome (ROS) is a subtype of POI manifested as normal ovarian reserve but insensitive to gonadotropin stimulation. Inactivating variants of follicle-stimulating hormone receptor (FSHR), a class A G-protein coupled receptor, have been associated with POI and are inherited via an autosomal recessive pattern. In this study, we investigated the genetic causes of a primary infertility patient manifested as POI with ROS, and elucidated the structural and functional impact of variants of uncertain significance. Next-generation sequencing (NGS) combined with Sanger sequencing revealed novel compound heterozygous FSHR variants: c.1384G>C/p.Ala462Pro and c.1862C>T/p.Ala621Val, inherited from her father and mother, respectively. The two altered amino acid sequences, localized in the third and seventh transmembrane helix of FSHR, were predicted as deleterious by in silico prediction. In vitro experiments revealed that the p.Ala462Pro variant resulted in barely detectable levels of intracellular signaling both in cAMP-dependent CRE-reporter activity and ERK activation and displayed a severely reduced plasma membrane receptor expression. In contrast, the p.Ala621Val variant resulted in partial loss of receptor activation without disruption of cell surface expression. In conclusion, two unreported inactivating FSHR variants potentially responsible for POI with ROS were first identified. This study expands the current phenotypic and genotypic spectrum of POI.
Collapse
Affiliation(s)
- Xiaopan Chen
- Reproductive Medicine Center, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Department of Genetic and Genomic Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Jing Shu, ; Xiaopan Chen,
| | - Linjie Chen
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Yang Wang
- Department of Genetic and Genomic Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- The Second Clinical Medical School of Wenzhou Medical University, Wenzhou, China
| | - Chongyi Shu
- Reproductive Medicine Center, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yier Zhou
- Reproductive Medicine Center, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Ruifang Wu
- Reproductive Medicine Center, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Bihui Jin
- Reproductive Medicine Center, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Leixiang Yang
- Department of Genetic and Genomic Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Junhui Sun
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ming Qi
- Department of Cell Biology and Medical Genetics, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jing Shu
- Reproductive Medicine Center, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- The Second Clinical Medical School of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Jing Shu, ; Xiaopan Chen,
| |
Collapse
|
34
|
Ulloa-Aguirre A, Zariñán T, Gutiérrez-Sagal R, Tao YX. Targeting trafficking as a therapeutic avenue for misfolded GPCRs leading to endocrine diseases. Front Endocrinol (Lausanne) 2022; 13:934685. [PMID: 36093106 PMCID: PMC9452723 DOI: 10.3389/fendo.2022.934685] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/13/2022] [Indexed: 02/05/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are plasma membrane proteins associated with an array of functions. Mutations in these receptors lead to a number of genetic diseases, including diseases involving the endocrine system. A particular subset of loss-of-function mutant GPCRs are misfolded receptors unable to traffic to their site of function (i.e. the cell surface plasma membrane). Endocrine disorders in humans caused by GPCR misfolding include, among others, hypo- and hyper-gonadotropic hypogonadism, morbid obesity, familial hypocalciuric hypercalcemia and neonatal severe hyperparathyroidism, X-linked nephrogenic diabetes insipidus, congenital hypothyroidism, and familial glucocorticoid resistance. Several in vitro and in vivo experimental approaches have been employed to restore function of some misfolded GPCRs linked to endocrine disfunction. The most promising approach is by employing pharmacological chaperones or pharmacoperones, which assist abnormally and incompletely folded proteins to refold correctly and adopt a more stable configuration to pass the scrutiny of the cell's quality control system, thereby correcting misrouting. This review covers the most important aspects that regulate folding and traffic of newly synthesized proteins, as well as the experimental approaches targeted to overcome protein misfolding, with special focus on GPCRs involved in endocrine diseases.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación (RAI), National University of Mexico and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
- *Correspondence: Alfredo Ulloa-Aguirre,
| | - Teresa Zariñán
- Red de Apoyo a la Investigación (RAI), National University of Mexico and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
| | - Rubén Gutiérrez-Sagal
- Red de Apoyo a la Investigación (RAI), National University of Mexico and Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, Mexico
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology & Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| |
Collapse
|
35
|
Abstract
Primary ovarian insufficiency (POI) is determined by exhaustion of follicles in the ovaries, which leads to infertility before the age of 40 years. It is characterized by a strong familial and heterogeneous genetic background. Therefore, we will mainly discuss the genetic basis of POI in this review. We identified 107 genes related to POI etiology in mammals described by several independent groups. Thirty-four of these genes (AARS2, AIRE, ANTXR1, ATM, BMPR1B, CLPP, CYP17A1, CYP19A1, DCAF17, EIF2B, ERAL1, FANCA, FANCC, FMR1, FOXL2, GALT, GNAS, HARS2, HSD17B4, LARS2, LMNA, MGME1, NBN, PMM2, POLG, PREPL, RCBTB1, RECQL2/3/4, STAR, TWNK, and XRCC4/9) have been linked to syndromic POI and are mainly implicated in metabolism function and meiosis/DNA repair. In addition, the majority of genes associated with nonsyndromic POI, widely expanded by high-throughput techniques over the last decade, have been implicated in ovarian development and meiosis/DNA repair pathways (ATG7, ATG9, ANKRD31, BMP8B, BMP15, BMPR1A, BMPR1B, BMPR2, BNC1, BRCA2, CPEB1, C14ORF39, DAZL, DIAPH2, DMC1, ERCC6, FANCL, FANCM, FIGLA, FSHR, GATA4, GDF9, GJA4, HELQ, HSF2BP, HFM1, INSL3, LHCGR, LHX8, MCM8, MCM9, MEIOB, MSH4, MSH5, NANOS3, NOBOX, NOTCH2, NR5A1, NUP107, PGRMC1, POLR3H, PRDM1, PRDM9, PSMC3IP, SOHLH1, SOHLH2, SPIDR, STAG3, SYCE1, TP63, UBR2, WDR62, and XRCC2), whereas a few are related to metabolic functions (EIF4ENIF1, KHDRBS1, MRPS22, POLR2C). Some genes, such as STRA8, FOXO3A, KIT, KITL, WNT4, and FANCE, have been shown to cause ovarian insufficiency in rodents, but mutations in these genes have yet to be elucidated in women affected by POI. Lastly, some genes have been rarely implicated in its etiology (AMH, AMHR2, ERRC2, ESR1, INHA, LMN4, POF1B, POU5F1, REC8, SMC1B). Considering the heterogeneous genetic and familial background of this disorder, we hope that an overview of literature data would reinforce that genetic screening of those patients is worthwhile and helpful for better genetic counseling and patient management.
Collapse
Affiliation(s)
- Monica Malheiros França
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Section of Endocrinology Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL, USA.
| | - Berenice Bilharinho Mendonca
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
36
|
Rivero-Müller A, Huhtaniemi I. Genetic variants of gonadotrophins and their receptors: Impact on the diagnosis and management of the infertile patient. Best Pract Res Clin Endocrinol Metab 2022; 36:101596. [PMID: 34802912 DOI: 10.1016/j.beem.2021.101596] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This narrative review is concerned with genetic variants of the genes encoding gonadotrophin subunits and their receptors, as well as their implications into the diagnosis and treatment of infertility. We first review briefly the basics of molecular biology and biochemistry of gonadotrophin and gonadotrophin receptor structure and function, then describe the phenotypic effects of polymorphisms and mutations of these genes, followed by diagnostic aspects. We will then summarise the information that inactivating gonadotrophin receptor mutations have provided about the controversial topic of extragonadal gonadotrophin action. Finally, we will close with the current and future therapeutic approaches on patients with gonadotrophin and their receptor mutations.
Collapse
Affiliation(s)
- Adolfo Rivero-Müller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, 20-093, Poland
| | - Ilpo Huhtaniemi
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK.
| |
Collapse
|
37
|
Lundin K, Sepponen K, Väyrynen P, Liu X, Yohannes DA, Survila M, Ghimire B, Känsäkoski J, Katayama S, Partanen J, Vuoristo S, Paloviita P, Rahman N, Raivio T, Luiro K, Huhtaniemi I, Varjosalo M, Tuuri T, Tapanainen JS. OUP accepted manuscript. Mol Hum Reprod 2022; 28:6574364. [PMID: 35471239 PMCID: PMC9308958 DOI: 10.1093/molehr/gaac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 03/11/2022] [Indexed: 11/14/2022] Open
Affiliation(s)
- K Lundin
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - K Sepponen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - P Väyrynen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - X Liu
- Molecular Systems Biology Research Group, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
- Proteomics Unit, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
| | - D A Yohannes
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Translational Immunology & Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - M Survila
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - B Ghimire
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - J Känsäkoski
- Department of Physiology, University of Helsinki, Helsinki, Finland
| | - S Katayama
- Folkhälsan Research Center, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J Partanen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - S Vuoristo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - P Paloviita
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - N Rahman
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - T Raivio
- Department of Physiology, University of Helsinki, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- New Children's Hospital, Pediatric Research Center, Helsinki University Hospital, HUH, Helsinki, Finland
| | - K Luiro
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - I Huhtaniemi
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Metabolism, Endocrinology and Reproduction, Faculty of Medicine, Hammersmith Campus, Imperial College London, London, UK
| | - M Varjosalo
- Molecular Systems Biology Research Group, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
- Proteomics Unit, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
| | - T Tuuri
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - J S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, University Hospital of Oulu, University of Oulu, Medical Research Center Oulu and PEDEGO Research Unit, Oulu, Finland
- Corresponding author. Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, PO Box 140, 00029 Helsinki, Finland. Tel: +358-94711; E-mail:
| |
Collapse
|
38
|
Hanyroup S, Anderson RC, Nataraja S, Yu HN, Millar RP, Newton CL. Rescue of Cell Surface Expression and Signaling of Mutant Follicle-Stimulating Hormone Receptors. Endocrinology 2021; 162:6311857. [PMID: 34192304 DOI: 10.1210/endocr/bqab134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Indexed: 11/19/2022]
Abstract
Mutations in G protein-coupled receptors (GPCRs) underlie numerous diseases. Many cause receptor misfolding and failure to reach the cell surface. Pharmacological chaperones are cell-permeant small molecules that engage nascent mutant GPCRs in the endoplasmic reticulum, stabilizing folding and "rescuing" cell surface expression. We previously demonstrated rescue of cell surface expression of luteinizing hormone receptor mutants by an allosteric agonist. Here we demonstrate that a similar approach can be employed to rescue mutant follicle-stimulating hormone receptors (FSHRs) with poor cell surface expression using a small-molecule FSHR agonist, CAN1404. Seventeen FSHR mutations described in patients with reproductive dysfunction were expressed in HEK 293T cells, and cell surface expression was determined by enzyme-linked immunosorbent assay of epitope-tagged FSHRs before/after treatment with CAN1404. Cell surface expression was severely reduced to ≤18% of wild-type (WT) for 11, modestly reduced to 66% to 84% of WT for 4, and not reduced for 2. Of the 11 with severely reduced cell surface expression, restoration to ≥57% of WT levels was achieved for 6 by treatment with 1 µM CAN1404 for 24 h, and a corresponding increase in FSH-induced signaling was observed for 4 of these, indicating restored functionality. Therefore, CAN1404 acts as a pharmacological chaperone and can rescue cell surface expression and function of certain mutant FSHRs with severely reduced cell surface expression. These findings aid in advancing the understanding of the effects of genetic mutations on GPCR function and provide a proof of therapeutic principle for FSHR pharmacological chaperones.
Collapse
Affiliation(s)
- Sharika Hanyroup
- Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ross C Anderson
- Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | | | | | - Robert P Millar
- Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Integrative Biomedical Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- School of Medicine, Medical and Biological Sciences Building, University of St Andrews, St Andrews, UK
| | - Claire L Newton
- Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
39
|
The Roles of Luteinizing Hormone, Follicle-Stimulating Hormone and Testosterone in Spermatogenesis and Folliculogenesis Revisited. Int J Mol Sci 2021; 22:ijms222312735. [PMID: 34884539 PMCID: PMC8658012 DOI: 10.3390/ijms222312735] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/17/2022] Open
Abstract
Spermatogenesis and folliculogenesis involve cell–cell interactions and gene expression orchestrated by luteinizing hormone (LH) and follicle-stimulating hormone (FSH). FSH regulates the proliferation and maturation of germ cells independently and in combination with LH. In humans, the requirement for high intratesticular testosterone (T) concentration in spermatogenesis remains both a dogma and an enigma, as it greatly exceeds the requirement for androgen receptor (AR) activation. Several data have challenged this dogma. Here we report our findings on a man with mutant LH beta subunit (LHβ) that markedly reduced T production to 1–2% of normal., but despite this minimal LH stimulation, T production by scarce mature Leydig cells was sufficient to initiate and maintain complete spermatogenesis. Also, in the LH receptor (LHR) knockout (LuRKO) mice, low-dose T supplementation was able to maintain spermatogenesis. In addition, in antiandrogen-treated LuRKO mice, devoid of T action, the transgenic expression of a constitutively activating follicle stimulating hormone receptor (FSHR) mutant was able to rescue spermatogenesis and fertility. Based on rodent models, it is believed that gonadotropin-dependent follicular growth begins at the antral stage, but models of FSHR inactivation in women contradict this claim. The complete loss of FSHR function results in the complete early blockage of folliculogenesis at the primary stage, with a high density of follicles of the prepubertal type. These results should prompt the reassessment of the role of gonadotropins in spermatogenesis, folliculogenesis and therapeutic applications in human hypogonadism and infertility.
Collapse
|
40
|
Utilization of Whole Exome Sequencing in Non-Syndromic Premature Ovarian Failure: Ficolin-3 Gene Mutation in an Iranian Family. IRANIAN BIOMEDICAL JOURNAL 2021; 25:441-6. [PMID: 34641644 DOI: 10.52547/ibj.25.6.441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background Premature ovarian failure is a heterogeneous disorder, leading to early menopause. Several genes have been identified as the cause of non-syndromic premature ovarian failure (POF). Our aim was to explore the genetic defects in Iranian patients with POF. Methods We studied a family with three females exhibiting non-syndromic POF. WES was performed for one of the affected individuals after ruling out the presence of CGG repeat expansion at fragile X mental retardation 1 gene in the family. Sanger sequencing was used to confirm the candidate sequence variants in the proband, and screening of the detected mutation was performed for the other affected and unaffected members of the family. Results A homozygous frameshift mutation, c.349delC, was identified in ficolin-3 (FCN3) gene in the proband and two other patients. The parents and two healthy brothers were heterozygous for the mutation, and an unaffected sister was homozygous for wild type. Conclusion This is the first report of a mutation in FCN3 gene in a family with POF. Our findings can lead to the enhancement of genetic databases of patients with POF, specifically for families with high-risk background.
Collapse
|
41
|
Bhartiya D, Patel H. An overview of FSH-FSHR biology and explaining the existing conundrums. J Ovarian Res 2021; 14:144. [PMID: 34717708 PMCID: PMC8557046 DOI: 10.1186/s13048-021-00880-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 09/12/2021] [Indexed: 12/23/2022] Open
Abstract
FSH was first identified in 1930 and is central to mammalian reproduction. It is indeed intriguing that despite being researched upon for about 90 years, there is still so much more to learn about FSH-FSHR biology. The purpose of this review is to provide an overview of current understanding of FSH-FSHR biology, to review published data on biological and clinical relevance of reported mutations, polymorphisms and alternately spliced isoforms of FSHR. Tissue-resident stem/progenitor cells in multiple adult tissues including ovaries, testes and uterus express FSHR and this observation results in a paradigm shift in the field. The results suggest a direct action of FSH on the stem cells in addition to their well-studied action on Granulosa and Sertoli cells in the ovaries and testes respectively. Present review further addresses various concerns raised in recent times by the scientific community regarding extragonadal expression of FSHR, especially in cancers affecting multiple organs. Similar population of primitive and pluripotent tissue-resident stem cells expressing FSHR exist in multiple adult tissues including bone marrow and reproductive tissues and help maintain homeostasis throughout life. Any dysfunction of these stem cells results in various pathologies and they also most likely get transformed into cancer stem cells and initiate cancer. This explains why multiple solid as well as liquid tumors express OCT-4 and FSHR. More research efforts need to be focused on alternately spliced FSHR isoforms.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Maharashtra, 400012, Mumbai, India. .,Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| | - Hiren Patel
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Maharashtra, 400012, Mumbai, India.,Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
42
|
Rooda I, Kaselt B, Liivrand M, Smolander OP, Salumets A, Velthut-Meikas A. Hsa-mir-548 family expression in human reproductive tissues. BMC Genom Data 2021; 22:40. [PMID: 34625017 PMCID: PMC8501715 DOI: 10.1186/s12863-021-00997-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Hsa-miR-548ba expressed in ovarian granulosa cells targets PTEN and LIFR, which are essential for ovarian follicle activation and growth. The expression pattern of hsa-miR-548ba correlates with its host gene follicle-stimulating hormone receptor (FSHR), and FSH has a positive influence on hsa-miR-548ba expression. However, hsa-miR-548ba is a member of a large hsa-mir-548 family with potentially overlapping targets. The current study aims to investigate the co-expression of hsa-mir-548 family members in FSHR-positive reproductive tissues and to explore the potential co-regulation of pathways. RESULTS For the above-described analysis, small RNA sequencing data from public data repositories were used. Sequencing results revealed that hsa-miR-548ba was expressed at the highest level in the ovarian granulosa cells and uterine myometrial samples together with another twelve and one hsa-miR-548 family members, respectively. Pathway enrichment analysis of microRNA targets in the ovarian samples revealed the hsa-miR-548ba and hsa-miR-548b-5p co-regulation of RAB geranylgeranylation in mural granulosa cells. Moreover, other hsa-mir-548 family members co-regulate pathways essential for ovarian functions (PIP3 activates AKT signalling and signalling by ERBB4). In addition to hsa-miR-548ba, hsa-miR-548o-3p is expressed in the myometrium, which separately targets the peroxisome proliferator-activated receptor alpha (PPARA) pathway. CONCLUSION This study reveals that hsa-mir-548 family members are expressed in variable combinations in the reproductive tract, where they potentially fulfil different regulatory roles. The results provide a reference for further studies of the hsa-mir-548 family role in the reproductive tract.
Collapse
Affiliation(s)
- Ilmatar Rooda
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia.
- Competence Centre on Health Technologies, Teaduspargi 13, 50411, Tartu, Estonia.
| | - Birgitta Kaselt
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Maria Liivrand
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Olli-Pekka Smolander
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Andres Salumets
- Competence Centre on Health Technologies, Teaduspargi 13, 50411, Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 14186, Stockholm, Sweden
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, L. Puusepa St. 8, 50406, Tartu, Estonia
- Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Agne Velthut-Meikas
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| |
Collapse
|
43
|
Monge-Ochoa B, Montoro L, Gil-Arribas E, Montoya J, Ruiz-Pesini E, López-Pérez MJ, de Castro F, Díez-Sánchez C. Variants Ala307Ala and Ser680Ser of 307 and 680 FSHr polymorphisms negatively influence on assisted reproductive techniques outcome and determine high probability of non-pregnancy in Caucasian patients. J Assist Reprod Genet 2021; 38:2769-2779. [PMID: 34346002 PMCID: PMC8581085 DOI: 10.1007/s10815-021-02276-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/07/2021] [Indexed: 10/20/2022] Open
Abstract
PURPOSE To determine the influence of different genotypes of Ala307Thr and Asn680Ser FSHr polymorphisms on controlled ovarian stimulation (COS) outcome and pregnancy. METHODS This study collected blood and physiological and clinical parameters of 517 Caucasian patients (Statistical power ≥ 80%) that underwent COS treatment. Genotypes of Ala307Thr and Asn680Ser polymorphisms were determined using PCR amplification followed by Bsu36I and BsrI digestion, respectively. RESULTS Ala307Ala and Ser680Ser genotypes associated to worse parameters of COS outcome (preovulatory follicles P = 0.05, in both), justifying their lower pregnancy rate than Non-Ala307Ala, P = 0.01 and Non-Ser680Ser, P = 0.004, respectively or together, (P = 0.003). Within the Non-Ala307Ala group, Thr307Thr genotype showed higher number of fertilized oocytes (P = 0.04) and embryos (P = 0.01) than Non-Thr307Thr, but no influence on pregnancy rate. Ala307Ala and Ser680Ser patients doubled probability of non-pregnancy than Non-Ala307Ala (odds ratio = 2.0) and Non-Ser680Ser (odds ratio = 2.11), respectively. Ala307Ala and Ser680Ser genotypes tend to appear together (P < 0.0001), which increases the probability of non-pregnancy. CONCLUSIONS Ala307Ala and Ser680Ser genotypes of 307 and 680 FSHr polymorphisms associate to worse COS outcome than its respective Non-Ala307Ala and Non-Ser680Ser. Within the Non-Ala307Ala genotypes, Thr307Thr, although shows higher Fertilized Oocytes and Embryos, do not influence on pregnancy rate. Ala307Ala and Ser680Ser genotypes double the probability of Non-Pregnancy than their respective Non-Ala307Ala and Non-Ser680Ser genotypes. Furthermore, the strong tendency of these genotypes to appear together worsens the probability of pregnancy in these patients.
Collapse
Affiliation(s)
- Belén Monge-Ochoa
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, Miguel Servet 177, 50013, Zaragoza, Spain
| | - Luis Montoro
- Unidad de Reproducción Asistida, Hospital Universitario Príncipe de Asturias, Universidad Complutense de Madrid, Alcalá de Henares, Madrid, Spain
| | | | - Julio Montoya
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, Miguel Servet 177, 50013, Zaragoza, Spain
- Centro de Investigaciones Biomédicas en Red (CIBER) de Enfermedades Raras, Madrid, Spain
- Instituto de Investigación Sanitaria (IIS) de Aragón, Zaragoza, Spain
| | - Eduardo Ruiz-Pesini
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, Miguel Servet 177, 50013, Zaragoza, Spain
- Centro de Investigaciones Biomédicas en Red (CIBER) de Enfermedades Raras, Madrid, Spain
- Instituto de Investigación Sanitaria (IIS) de Aragón, Zaragoza, Spain
| | - Manuel J López-Pérez
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, Miguel Servet 177, 50013, Zaragoza, Spain
| | - Francisco de Castro
- Unidad de Reproducción Asistida, Hospital Universitario Príncipe de Asturias, Universidad Complutense de Madrid, Alcalá de Henares, Madrid, Spain
| | - Carmen Díez-Sánchez
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, Miguel Servet 177, 50013, Zaragoza, Spain.
| |
Collapse
|
44
|
Recchia K, Jorge AS, Pessôa LVDF, Botigelli RC, Zugaib VC, de Souza AF, Martins DDS, Ambrósio CE, Bressan FF, Pieri NCG. Actions and Roles of FSH in Germinative Cells. Int J Mol Sci 2021; 22:10110. [PMID: 34576272 PMCID: PMC8470522 DOI: 10.3390/ijms221810110] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/21/2022] Open
Abstract
Follicle stimulating hormone (FSH) is produced by the pituitary gland in a coordinated hypothalamic-pituitary-gonadal (HPG) axis event, plays important roles in reproduction and germ cell development during different phases of reproductive development (fetal, neonatal, puberty, and adult life), and is consequently essential for fertility. FSH is a heterodimeric glycoprotein hormone of two dissociable subunits, α and β. The FSH β-subunit (FSHβ) function starts upon coupling to its specific receptor: follicle-stimulating hormone receptor (FSHR). FSHRs are localized mainly on the surface of target cells on the testis and ovary (granulosa and Sertoli cells) and have recently been found in testicular stem cells and extra-gonadal tissue. Several reproduction disorders are associated with absent or low FSH secretion, with mutation of the FSH β-subunit or the FSH receptor, and/or its signaling pathways. However, the influence of FSH on germ cells is still poorly understood; some studies have suggested that this hormone also plays a determinant role in the self-renewal of germinative cells and acts to increase undifferentiated spermatogonia proliferation. In addition, in vitro, together with other factors, it assists the process of differentiation of primordial germ cells (PGCLCs) into gametes (oocyte-like and SSCLCs). In this review, we describe relevant research on the influence of FSH on spermatogenesis and folliculogenesis, mainly in the germ cell of humans and other species. The possible roles of FSH in germ cell generation in vitro are also presented.
Collapse
Affiliation(s)
- Kaiana Recchia
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, Brazil; (K.R.); (F.F.B.)
| | - Amanda Soares Jorge
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Laís Vicari de Figueiredo Pessôa
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Ramon Cesar Botigelli
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
- Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-970, Brazil
| | - Vanessa Cristiane Zugaib
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Aline Fernanda de Souza
- Department Biomedical Science, Ontary Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Daniele dos Santos Martins
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Carlos Eduardo Ambrósio
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Fabiana Fernandes Bressan
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, Brazil; (K.R.); (F.F.B.)
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Naira Caroline Godoy Pieri
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| |
Collapse
|
45
|
Byambaragchaa M, Ahn TY, Choi SH, Kang MH, Min KS. Functional characterization of naturally-occurring constitutively activating/inactivating mutations in equine follicle-stimulating hormone receptor (eFSHR). Anim Biosci 2021; 35:399-409. [PMID: 34474536 PMCID: PMC8902225 DOI: 10.5713/ab.21.0246] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/12/2021] [Indexed: 11/27/2022] Open
Abstract
Objective Follicle-stimulating hormone (FSH) is the central hormone involved in mammalian reproduction, maturation at puberty, and gamete production that mediates its function by control of follicle growth and function. The present study investigated the mutations involved in the regulation of FSH receptor (FSHR) activation. Methods We analyzed seven naturally-occurring mutations that were previously reported in human FSHR (hFSHR), in the context of equine FSHR (eFSHR); these include one constitutively activation variant, one allelic variant, and five inactivating variants. These mutations were introduced into wild-type eFSHR (eFSHR-wt) sequence to generate mutants that were designated as eFSHR-D566G, -A306T, -A189V, -N191I, -R572C, -A574V, and -R633H. Mutants were transfected into PathHunter EA-parental CHO-K1 cells expressing β-arrestin. The biological function of mutants was analyzed by quantitating cAMP accumulation in cells incubated with increasing concentrations of FSH. Results Cells expressing eFSHR-D566G exhibited an 8.6-fold increase in basal cAMP response, as compared to that in eFSHR-wt. The allelic variation mutant eFSHR-A306T was not found to affect the basal cAMP response or EC50 levels. On the other hand, eFSHR-D566G and eFSHR-A306T displayed a 1.5- and 1.4-fold increase in the maximal response, respectively. Signal transduction was found to be completely impaired in case of the inactivating mutants eFSHR-A189V, -R572C, and -A574V. When compared with eFSHR-wt, eFSHR-N191I displayed a 5.4-fold decrease in the EC50 levels (3910 ng/mL) and a 2.3-fold decrease in the maximal response. In contrast, cells expressing eFSHR-R633H displayed in a similar manner to that of the cells expressing the eFSHR-wt on signal transduction and maximal response. Conclusion The activating mutant eFSHR-D566G greatly enhanced the signal transduction in response to FSH, in the absence of agonist treatment. We suggest that the state of activation of the eFSHR can modulate its basal cAMP accumulation.
Collapse
Affiliation(s)
- Munkhzaya Byambaragchaa
- Animal Biotechnology, Graduate School of Future Convergence Technology, Hankyong National University, Ansung 17579, Korea
| | - Tae-Young Ahn
- Animal Biotechnology, Graduate School of Future Convergence Technology, Hankyong National University, Ansung 17579, Korea
| | - Seung-Hee Choi
- Animal Biotechnology, Graduate School of Future Convergence Technology, Hankyong National University, Ansung 17579, Korea
| | - Myung-Hwa Kang
- Department of Food Science and Nutrition, Hoseo University, Asan 31499, Korea
| | - Kwan-Sik Min
- Animal Biotechnology, Graduate School of Future Convergence Technology, Hankyong National University, Ansung 17579, Korea.,School of Animal Life Convergence Science, Institute of Genetic Engineering, Hankyong National University, Ansung 17579, Korea
| |
Collapse
|
46
|
Yang Q, Mumusoglu S, Qin Y, Sun Y, Hsueh AJ. A kaleidoscopic view of ovarian genes associated with premature ovarian insufficiency and senescence. FASEB J 2021; 35:e21753. [PMID: 34233068 DOI: 10.1096/fj.202100756r] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022]
Abstract
Ovarian infertility and subfertility presenting with premature ovarian insufficiency (POI) and diminished ovarian reserve are major issues facing the developed world due to the trend of delaying childbirth. Ovarian senescence and POI represent a continuum of physiological/pathophysiological changes in ovarian follicle functions. Based on advances in whole exome sequencing, evaluation of gene copy variants, together with family-based and genome-wide association studies, we discussed genes responsible for POI and ovarian senescence. We used a gene-centric approach to sort out literature deposited in the Ovarian Kaleidoscope database (http://okdb.appliedbioinfo.net) by sub-categorizing candidate genes as ligand-receptor signaling, meiosis and DNA repair, transcriptional factors, RNA metabolism, enzymes, and others. We discussed individual gene mutations found in POI patients and verification of gene functions in gene-deleted model organisms. Decreased expression of some of the POI genes could be responsible for ovarian senescence, especially those essential for DNA repair, meiosis and mitochondrial functions. We propose to set up a candidate gene panel for targeted sequencing in POI patients together with studies on mitochondria-associated genes in middle-aged subfertile patients.
Collapse
Affiliation(s)
- Qingling Yang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sezcan Mumusoglu
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Obstetrics and Gynecology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Yingying Qin
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yingpu Sun
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Aaron J Hsueh
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
47
|
Benammar A, Fanchin R, Filali-Baba M, Vialard F, Fossard C, Vandame J, Pirtea P, Racowsky C, Ayoubi JM, Poulain M. Utilization of in vitro maturation in cases with a FSH receptor mutation. J Assist Reprod Genet 2021; 38:1311-1321. [PMID: 34089127 DOI: 10.1007/s10815-021-02249-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/26/2021] [Indexed: 10/21/2022] Open
Abstract
PURPOSE To identify the FSH receptor (FSHR) variant and efficacy of in vitro maturation (IVM) in a 28-year-old woman with secondary amenorrhea, primary infertility, and ovarian resistance to FSH, and to analyze the genotype-to-phenotype relationship in cases of FSHR mutation for the development of an IVM algorithm for use in patients with gonadotropin resistance syndrome (GRS). METHODS Oocytes retrieved after menstruation induction with norethisterone, followed by daily estrogen and an ovulatory trigger, underwent IVM, ICSI, and culture in a time-lapse (TL) incubator. Embryo transfers were performed on day 2, and after thawing on day 5. Genes associated with disorders of sex development were sequenced for both the patient and her parents. All reported cases of FSHR mutation were analyzed to investigate genotype/phenotypic relationships. RESULTS After ovum pickup, seven of 16 oocytes matured and all fertilized. After unsuccessful day 2 transfer, our patient delivered with a thawed day 5 blastocyst, the sole embryo without abnormal TL phenotypes. Genetic analysis revealed a new composite heterozygous FSHR variant. Analysis of our patient case with published cases of GRS revealed associations among FSHR variant genotype, location on the FSHR, functionality of tested variants, and type of amenorrhea. An algorithm for application of IVM for GRS patients was developed. CONCLUSIONS We report two novel variants of the FSHR. Although IVM successfully matured some oocytes, only one resulted in an embryo with normal TL phenotypes. We recommend FSHR genetic testing in GRS patients, which will help guide their suitability for IVM.
Collapse
Affiliation(s)
- Achraf Benammar
- Department of Gyneacology, Obstetrics and Reproductive Medicine, Hospital Foch, 92150, Suresnes, France.
| | - Renato Fanchin
- Department of Gyneacology, Obstetrics and Reproductive Medicine, Hospital Foch, 92150, Suresnes, France
| | - Meryem Filali-Baba
- Department of Gyneacology, Obstetrics and Reproductive Medicine, Hospital Foch, 92150, Suresnes, France
| | - François Vialard
- Université Paris-Saclay, INRAE, ENVA, UVSQ, BREED, 78350, Jouy-en-Josas, France.,Genetics Federation, CHI de Poissy St Germain en Laye, 78303, Poissy, France
| | - Camille Fossard
- Department of Gyneacology, Obstetrics and Reproductive Medicine, Hospital Foch, 92150, Suresnes, France
| | - Jessica Vandame
- Department of Gyneacology, Obstetrics and Reproductive Medicine, Hospital Foch, 92150, Suresnes, France
| | - Paul Pirtea
- Department of Gyneacology, Obstetrics and Reproductive Medicine, Hospital Foch, 92150, Suresnes, France
| | - Catherine Racowsky
- Department of Gyneacology, Obstetrics and Reproductive Medicine, Hospital Foch, 92150, Suresnes, France
| | - Jean-Marc Ayoubi
- Department of Gyneacology, Obstetrics and Reproductive Medicine, Hospital Foch, 92150, Suresnes, France.,Université Paris-Saclay, INRAE, ENVA, UVSQ, BREED, 78350, Jouy-en-Josas, France
| | - Marine Poulain
- Department of Gyneacology, Obstetrics and Reproductive Medicine, Hospital Foch, 92150, Suresnes, France.,Université Paris-Saclay, INRAE, ENVA, UVSQ, BREED, 78350, Jouy-en-Josas, France
| |
Collapse
|
48
|
De Vos M, Grynberg M, Ho TM, Yuan Y, Albertini DF, Gilchrist RB. Perspectives on the development and future of oocyte IVM in clinical practice. J Assist Reprod Genet 2021; 38:1265-1280. [PMID: 34218388 PMCID: PMC8266966 DOI: 10.1007/s10815-021-02263-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/15/2021] [Indexed: 12/19/2022] Open
Abstract
Oocyte in vitro maturation (IVM) is an assisted reproductive technology designed to obtain mature oocytes following culture of immature cumulus-oocyte complexes collected from antral follicles. Although IVM has been practiced for decades and is no longer considered experimental, the uptake of IVM in clinical practice is currently limited. The purpose of this review is to ensure reproductive medicine professionals understand the appropriate use of IVM drawn from the best available evidence supporting its clinical potential and safety in selected patient groups. This group of scientists and fertility specialists, with expertise in IVM in the ART laboratory and/or clinic, explore here the development of IVM towards acquisition of a non-experimental status and, in addition, critically appraise the current and future role of IVM in human ART.
Collapse
Affiliation(s)
- Michel De Vos
- Centre for Reproductive Medicine, UZ Brussel, Brussels, Belgium
- Department of Obstetrics, Gynecology, Perinatology and Reproductology, Institute of Professional Education, Sechenov University, Moscow, Russia
| | - Michaël Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Antoine Béclère University Hospital, Clamart, Clamart, France
- Paris-Sud University, Le Kremlin Bicêtre, France
| | - Tuong M Ho
- IVFMD, My Duc Hospital, Ho Chi Minh City, Vietnam
| | - Ye Yuan
- Colorado Center for Reproductive Medicine, Lone Tree, CO, 80124, USA
| | - David F Albertini
- Bedford Research Foundation, 124 South Road, Bedford, MA, 01730, USA
| | - Robert B Gilchrist
- Fertility & Research Centre, School of Women's and Children's Health, University of New South Wales Sydney, Sydney, NSW, Australia.
| |
Collapse
|
49
|
Chon SJ, Umair Z, Yoon MS. Premature Ovarian Insufficiency: Past, Present, and Future. Front Cell Dev Biol 2021; 9:672890. [PMID: 34041247 PMCID: PMC8141617 DOI: 10.3389/fcell.2021.672890] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022] Open
Abstract
Premature ovarian insufficiency (POI) is the loss of normal ovarian function before the age of 40 years, a condition that affects approximately 1% of women under 40 years old and 0.1% of women under 30 years old. It is biochemically characterized by amenorrhea with hypoestrogenic and hypergonadotropic conditions, in some cases, causing loss of fertility. Heterogeneity of POI is registered by genetic and non-genetic causes, such as autoimmunity, environmental toxins, and chemicals. The identification of possible causative genes and selection of candidate genes for POI confirmation remain to be elucidated in cases of idiopathic POI. This review discusses the current understanding and future prospects of heterogeneous POI. We focus on the genetic basis of POI and the recent studies on non-coding RNA in POI pathogenesis as well as on animal models of POI pathogenesis, which help unravel POI mechanisms and potential targets. Despite the latest discoveries, the crosstalk among gene regulatory networks and the possible therapies targeting the same needs to explore in near future.
Collapse
Affiliation(s)
- Seung Joo Chon
- Department of Obstetrics and Gynecology, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon, South Korea
| | - Zobia Umair
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, South Korea
| | - Mee-Sup Yoon
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, South Korea.,Lee Gil Ya Cancer and Diabetes Institute, Incheon, South Korea.,Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, South Korea
| |
Collapse
|
50
|
Eskenazi S, Bachelot A, Hugon-Rodin J, Plu-Bureau G, Gompel A, Catteau-Jonard S, Molina-Gomes D, Dewailly D, Dodé C, Christin-Maitre S, Touraine P. Next Generation Sequencing Should Be Proposed to Every Woman With "Idiopathic" Primary Ovarian Insufficiency. J Endocr Soc 2021; 5:bvab032. [PMID: 34095689 PMCID: PMC8169040 DOI: 10.1210/jendso/bvab032] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Indexed: 12/14/2022] Open
Abstract
Context Primary ovarian insufficiency (POI) affects 1% of women under 40 years of age. POI is idiopathic in more than 70% of cases. Though many candidate genes have been identified in recent years, the prevalence and pathogenicity of abnormalities are still difficult to establish. Objective Our primary objective was to evaluate the prevalence of gene variations in a large prospective multicentric POI cohort. Our secondary objective was to evaluate the correlation between phenotype and genotype. Methods Two hundred and sixty-nine well-phenotyped POI patients were screened for variants of 18 known POI genes (BMP15, DMC1, EIF2S2, FIGLA, FOXL2, FSHR, GDF9, GPR3, HFM1, LHX8, MSH5, NOBOX, NR5A1, PGRMC1, STAG3, XPNPEP2, BHLB, and FSHB) by next generation sequencing (NGS). Abnormalities were classified as "variant" or "variant of unknown signification" (VUS) according to available functional tests or algorithms (SIFT, Polyphen-2, MutationTaster). Results One hundred and two patients (38%) were identified as having at least 1 genetic abnormality. Sixty-seven patients (25%) presented at least 1 variant. Forty-eight patients presented at least 1 VUS (18%). Thirteen patients (5%) had combined abnormalities. NOBOX variants were the most common gene variants involved in POI (9%). Interestingly, we saw no significant differences in the previous family history of POI, ethnic origin, age at onset of POI, primary amenorrhea, or secondary menstrual disturbances between the different genotypes. Conclusion In our study, a high percentage of patients presented gene variants detected by NGS analysis (38%). Every POI patient should undergo NGS analysis to improve medical cares of the patients.
Collapse
Affiliation(s)
- Sarah Eskenazi
- Department of Reproductive Endocrinology, Saint-Antoine Hospital, AP-HP, Paris, France; Center for Rare Growth Disorders and Center for Developmental Disorders: CMERC.,Sorbonne University Medicine, Paris, France
| | - Anne Bachelot
- Sorbonne University Medicine, Paris, France.,Department of Endocrinology and Reproductive Medicine, Pitié-Salpêtrière Hospital, AP-HP, Paris, France; Center for Rare Endocrine Disorders and Center for Rare Gynecological Disorders: CMERC
| | - Justine Hugon-Rodin
- Department of Gynecology and Endocrinology, Cochin/Port-Royal Hospital, AP-HP, Paris, France.,Paris Descartes University, Paris, France.,INSERM UMR 1153, EPOPE group, Paris, France
| | - Genevieve Plu-Bureau
- Department of Gynecology and Endocrinology, Cochin/Port-Royal Hospital, AP-HP, Paris, France.,Paris Descartes University, Paris, France.,INSERM UMR 1153, EPOPE group, Paris, France
| | - Anne Gompel
- Department of Gynecology and Endocrinology, Cochin/Port-Royal Hospital, AP-HP, Paris, France.,Paris Descartes University, Paris, France
| | - Sophie Catteau-Jonard
- Department of Medical Gynaecology, CHU Lille, University of Lillle, F-59000 Lille, France
| | - Denise Molina-Gomes
- Department of Assisted Reproductive Technics, Poissy Saint-Germain-en-Laye Hospital, Poissy, France
| | - Didier Dewailly
- Department of Medical Gynaecology, CHU Lille, University of Lillle, F-59000 Lille, France
| | - Catherine Dodé
- Department of Genetics and Molecular Biology, Cochin/Port-Royal Hospital, AP-HP, Paris, France
| | - Sophie Christin-Maitre
- Department of Reproductive Endocrinology, Saint-Antoine Hospital, AP-HP, Paris, France; Center for Rare Growth Disorders and Center for Developmental Disorders: CMERC.,Sorbonne University Medicine, Paris, France.,INSERM UMR-S933, 75012 Paris, France
| | - Philippe Touraine
- Sorbonne University Medicine, Paris, France.,Department of Endocrinology and Reproductive Medicine, Pitié-Salpêtrière Hospital, AP-HP, Paris, France; Center for Rare Endocrine Disorders and Center for Rare Gynecological Disorders: CMERC
| |
Collapse
|