1
|
Luppino G, Wasniewska M, Coco R, Pepe G, Morabito LA, Li Pomi A, Corica D, Aversa T. Role of NR5A1 Gene Mutations in Disorders of Sex Development: Molecular and Clinical Features. Curr Issues Mol Biol 2024; 46:4519-4532. [PMID: 38785542 PMCID: PMC11119465 DOI: 10.3390/cimb46050274] [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: 03/30/2024] [Revised: 05/04/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Disorders/differences of sex development (DSDs) are defined as broad, heterogenous groups of congenital conditions characterized by atypical development of genetic, gonadal, or phenotypic sex accompanied by abnormal development of internal and/or external genitalia. NR5A1 gene mutation is one of the principal genetic alterations implicated in causing DSD. This review outlines the role of NR5A1 gene during the process of gonadal development in humans, provides an overview of the molecular and functional characteristics of NR5A1 gene, and discusses potential clinical phenotypes and additional organ diseases due to NR5A1 mutations. NR5A1 mutations were analyzed in patients with 46,XY DSD and 46,XX DSD both during the neonatal and pubertal periods. Loss of function of the NR5A1 gene causes several different phenotypes, including some associated with disease in additional organs. Clinical phenotypes may vary, even among patients carrying the same NR5A1 variant, indicating that there is no specific genotype-phenotype correlation. Genetic tests are crucial diagnostic tools that should be used early in the diagnostic pathway, as early as the neonatal period, when gonadal dysgenesis is the main manifestation of NR5A1 mutation. NR5A1 gene mutations could be mainly associated with amenorrhea, ovarian failure, hypogonadism, and infertility during puberty. Fertility preservation techniques should be considered as early as possible.
Collapse
Affiliation(s)
- Giovanni Luppino
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (R.C.); (G.P.); (A.L.P.); (D.C.); (T.A.)
| | - Malgorzata Wasniewska
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (R.C.); (G.P.); (A.L.P.); (D.C.); (T.A.)
- Pediatric Unit, AOU Policlinico G. Martino, Via Consolare Valeria 1, 98125 Messina, Italy;
| | - Roberto Coco
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (R.C.); (G.P.); (A.L.P.); (D.C.); (T.A.)
| | - Giorgia Pepe
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (R.C.); (G.P.); (A.L.P.); (D.C.); (T.A.)
- Pediatric Unit, AOU Policlinico G. Martino, Via Consolare Valeria 1, 98125 Messina, Italy;
| | - Letteria Anna Morabito
- Pediatric Unit, AOU Policlinico G. Martino, Via Consolare Valeria 1, 98125 Messina, Italy;
| | - Alessandra Li Pomi
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (R.C.); (G.P.); (A.L.P.); (D.C.); (T.A.)
| | - Domenico Corica
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (R.C.); (G.P.); (A.L.P.); (D.C.); (T.A.)
- Pediatric Unit, AOU Policlinico G. Martino, Via Consolare Valeria 1, 98125 Messina, Italy;
| | - Tommaso Aversa
- Department of Human Pathology of Adulthood and Childhood, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (R.C.); (G.P.); (A.L.P.); (D.C.); (T.A.)
- Pediatric Unit, AOU Policlinico G. Martino, Via Consolare Valeria 1, 98125 Messina, Italy;
| |
Collapse
|
2
|
Teoli J, Mallet D, Renault L, Gay CL, Labrune E, Bretones P, Giscard D’Estaing S, Cuzin B, Dijoud F, Roucher-Boulez F, Plotton I. Case Report: Longitudinal follow-up and testicular sperm extraction in a patient with a pathogenic NR5A1 (SF-1) frameshift variant: p.(Phe70Ser fs*5). Front Endocrinol (Lausanne) 2023; 14:1171822. [PMID: 37409232 PMCID: PMC10319352 DOI: 10.3389/fendo.2023.1171822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/26/2023] [Indexed: 07/07/2023] Open
Abstract
Background Steroidogenic factor 1 (SF-1), encoded by the nuclear receptor subfamily 5 group A member 1 (NR5A1) gene, is a transcriptional factor crucial for adrenal and gonadal organogenesis. Pathogenic variants of NR5A1 are responsible for a wide spectrum of phenotypes with autosomal dominant inheritance including disorders of sex development and oligospermia-azoospermia in 46,XY adults. Preservation of fertility remains challenging in these patients. Objective The aim was to offer fertility preservation at the end of puberty in an NR5A1 mutated patient. Case report The patient was born of non-consanguineous parents, with a disorder of sex development, a small genital bud, perineal hypospadias, and gonads in the left labioscrotal fold and the right inguinal region. Neither uterus nor vagina was detected. The karyotype was 46,XY. Anti-Müllerian hormone (AMH) and testosterone levels were low, indicating testicular dysgenesis. The child was raised as a boy. At 9 years old, he presented with precocious puberty treated by triptorelin. At puberty, follicle-stimulating hormone (FSH), luteinising hormone (LH), and testosterone levels increased, whereas AMH, inhibin B, and testicular volume were low, suggesting an impaired Sertoli cell function and a partially preserved Leydig cell function. A genetic study performed at almost 15 years old identified the new frameshift variant NM_004959.5: c.207del p.(Phe70Serfs*5) at a heterozygous state. He was thus addressed for fertility preservation. No sperm cells could be retrieved from three semen collections between the ages of 16 years 4 months and 16 years 10 months. A conventional bilateral testicular biopsy and testicular sperm extraction were performed at 17 years 10 months of age, but no sperm cells were found. Histological analysis revealed an aspect of mosaicism with seminiferous tubules that were either atrophic, with Sertoli cells only, or presenting an arrest of spermatogenesis at the spermatocyte stage. Conclusion We report a case with a new NR5A1 variant. The fertility preservation protocol proposed at the end of puberty did not allow any sperm retrieval for future parenthood.
Collapse
Affiliation(s)
- Jordan Teoli
- Service de Biochimie et Biologie Moléculaire, Unité Médicale de Biologie Endocrinienne, Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France
- Département des sciences biomédicales B, Institut des sciences pharmaceutiques et biologiques, Université Claude Bernard Lyon 1, Lyon, France
- Institut Cellule Souche et Cerveau (SBRI), Unité de Institut national de la recherche médicale (INSERM) 1208, Centre de Recherche INSERM, Bron, France
| | - Delphine Mallet
- Service de Biochimie et Biologie Moléculaire, Unité Médicale de Biologie Endocrinienne, Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France
- Centre de Référence Maladies Rares du Développement Génital: du Fœtus à l’Adulte, Filière Maladies Rares Endocriniennes, Bron, France
| | - Lucie Renault
- Service de médecine de la reproduction, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron, France
| | - Claire-Lise Gay
- Centre de Référence Maladies Rares du Développement Génital: du Fœtus à l’Adulte, Filière Maladies Rares Endocriniennes, Bron, France
- Service d’endocrinologie pédiatrique, Institut Saint-Pierre, Palavas-Les-Flots, France
| | - Elsa Labrune
- Institut Cellule Souche et Cerveau (SBRI), Unité de Institut national de la recherche médicale (INSERM) 1208, Centre de Recherche INSERM, Bron, France
- Service de médecine de la reproduction, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron, France
- Faculté de médecine, Université Claude Bernard Lyon 1, Lyon, France
| | - Patricia Bretones
- Centre de Référence Maladies Rares du Développement Génital: du Fœtus à l’Adulte, Filière Maladies Rares Endocriniennes, Bron, France
- Service d’endocrinologie pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron, France
| | - Sandrine Giscard D’Estaing
- Institut Cellule Souche et Cerveau (SBRI), Unité de Institut national de la recherche médicale (INSERM) 1208, Centre de Recherche INSERM, Bron, France
- Service de médecine de la reproduction, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron, France
- Faculté de médecine, Université Claude Bernard Lyon 1, Lyon, France
| | - Béatrice Cuzin
- Chirurgie Urologique, Centre Lyonnais d’Urologie Bellecour, Lyon, France
| | - Frédérique Dijoud
- Institut Cellule Souche et Cerveau (SBRI), Unité de Institut national de la recherche médicale (INSERM) 1208, Centre de Recherche INSERM, Bron, France
- Faculté de médecine, Université Claude Bernard Lyon 1, Lyon, France
- Service d’Anatomie Pathologique, Centre de Biologie et de Pathologie Est, Hospices Civils de Lyon, Bron, France
| | - Florence Roucher-Boulez
- Service de Biochimie et Biologie Moléculaire, Unité Médicale de Biologie Endocrinienne, Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France
- Centre de Référence Maladies Rares du Développement Génital: du Fœtus à l’Adulte, Filière Maladies Rares Endocriniennes, Bron, France
- Faculté de médecine, Université Claude Bernard Lyon 1, Lyon, France
- Institut Génétique, Reproduction & Développement (iGReD), Centre national de la recherche scientifique (CNRS), INSERM, Université Clermont Auvergne, Clermont–Ferrand, France
| | - Ingrid Plotton
- Service de Biochimie et Biologie Moléculaire, Unité Médicale de Biologie Endocrinienne, Centre de Biologie et Pathologie Est, Hospices Civils de Lyon, Bron, France
- Institut Cellule Souche et Cerveau (SBRI), Unité de Institut national de la recherche médicale (INSERM) 1208, Centre de Recherche INSERM, Bron, France
- Centre de Référence Maladies Rares du Développement Génital: du Fœtus à l’Adulte, Filière Maladies Rares Endocriniennes, Bron, France
- Service de médecine de la reproduction, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron, France
- Faculté de médecine, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
3
|
Shi JW, Zhou YW, Chen YF, Ye M, Qiao F, Tian JW, Zhang MY, Lin HC, Xie GC, Fok KL, Jiang H, Liu Y, Chen H. Epididymis cell atlas in a patient with a sex development disorder and a novel NR5A1 gene mutation. Asian J Androl 2023; 25:103-112. [PMID: 35546286 PMCID: PMC9933965 DOI: 10.4103/aja202226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/24/2022] [Indexed: 01/19/2023] Open
Abstract
This study aims to characterize the cell atlas of the epididymis derived from a 46,XY disorders of sex development (DSD) patient with a novel heterozygous mutation of the nuclear receptor subfamily 5 group A member 1 (NR5A1) gene. Next-generation sequencing found a heterozygous c.124C>G mutation in NR5A1 that resulted in a p.Q42E missense mutation in the conserved DNA-binding domain of NR5A1. The patient demonstrated feminization of external genitalia and Tanner stage 1 breast development. The surgical procedure revealed a morphologically normal epididymis and vas deferens but a dysplastic testis. Microfluidic-based single-cell RNA sequencing (scRNA-seq) analysis found that the fibroblast cells were significantly increased (approximately 46.5%), whereas the number of main epididymal epithelial cells (approximately 9.2%), such as principal cells and basal cells, was dramatically decreased. Bioinformatics analysis of cell-cell communications and gene regulatory networks at the single-cell level inferred that epididymal epithelial cell loss and fibroblast occupation are associated with the epithelial-to-mesenchymal transition (EMT) process. The present study provides a cell atlas of the epididymis of a patient with 46,XY DSD and serves as an important resource for understanding the pathophysiology of DSD.
Collapse
Affiliation(s)
- Jian-Wu Shi
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong 226000, China
| | - Yi-Wen Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yu-Fei Chen
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong 226000, China
| | - Mei Ye
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong 226000, China
| | - Feng Qiao
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong 226000, China
| | - Jia-Wei Tian
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong 226000, China
| | - Meng-Ya Zhang
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong 226000, China
| | - Hao-Cheng Lin
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Gang-Cai Xie
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong 226000, China
| | - Kin Lam Fok
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hui Jiang
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Yang Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Hao Chen
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong 226000, China
| |
Collapse
|
4
|
Mönig I, Schneidewind J, Johannsen TH, Juul A, Werner R, Lünstedt R, Birnbaum W, Marshall L, Wünsch L, Hiort O. Pubertal development in 46,XY patients with NR5A1 mutations. Endocrine 2022; 75:601-613. [PMID: 34613524 PMCID: PMC8816419 DOI: 10.1007/s12020-021-02883-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/15/2021] [Indexed: 11/25/2022]
Abstract
PURPOSE Mutations in the NR5A1 gene, encoding the transcription factor Steroidogenic Factor-1, are associated with a highly variable genital phenotype in patients with 46,XY differences of sex development (DSD). Our objective was to analyse the pubertal development in 46,XY patients with NR5A1 mutations by the evaluation of longitudinal clinical and hormonal data at pubertal age. METHODS We retrospectively studied a cohort of 10 46,XY patients with a verified NR5A1 mutation and describe clinical features including the external and internal genitalia, testicular volumes, Tanner stages and serum concentrations of LH, FSH, testosterone, AMH, and inhibin B during pubertal transition. RESULTS Patients who first presented in early infancy due to ambiguous genitalia showed spontaneous virilization at pubertal age accompanied by a significant testosterone production despite the decreased gonadal volume. Patients with apparently female external genitalia at birth presented later in life at pubertal age either with signs of virilization and/or absence of female puberty. Testosterone levels were highly variable in this group. In all patients, gonadotropins were constantly in the upper reference range or elevated. Neither the extent of virilization at birth nor the presence of Müllerian structures reliably correlated with the degree of virilization during puberty. CONCLUSION Patients with NR5A1 mutations regardless of phenotype at birth may demonstrate considerable virilization at puberty. Therefore, it is important to consider sex assignment carefully and avoid irreversible procedures during infancy.
Collapse
Affiliation(s)
- Isabel Mönig
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, Lübeck, Germany.
| | - Julia Schneidewind
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, Lübeck, Germany
| | - Trine H Johannsen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ralf Werner
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, Lübeck, Germany
- Institute for Molecular Medicine, University of Lübeck, Lübeck, Germany
| | - Ralf Lünstedt
- Catholic Children's Hospital Wilhelmstift, Hamburg, Germany
| | - Wiebke Birnbaum
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, Lübeck, Germany
| | - Louise Marshall
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, Lübeck, Germany
| | - Lutz Wünsch
- Department of Paediatric Surgery, University of Lübeck, Lübeck, Germany
| | - Olaf Hiort
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, Lübeck, Germany
| |
Collapse
|
5
|
Identification and functional analysis of fourteen NR5A1 variants in patients with the 46 XY disorders of sex development. Gene 2020; 760:145004. [PMID: 32738419 DOI: 10.1016/j.gene.2020.145004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/21/2020] [Accepted: 07/25/2020] [Indexed: 10/23/2022]
Abstract
Human sex determination and differentiation is a complex process, during which NR5A1 plays a central role via the transcriptional regulation of key modulators involved in steroidogenesis. Approximately 8-15% of 46,XY DSD are caused by variants in the NR5A1 gene. Therefore, screening for variants in the NR5A1 gene was performed in a Chinese cohort of sixty-two 46,XY DSD patients with no AR or SRD5A2 variants via next-generation sequencing (NGS). Fourteen variants in the NR5A1 gene were identified in 16 patients from 14 unrelated families, including nine novel variants. These variants included eight heterozygote missense variants, two heterozygote frameshift variants, two heterozygote nonsense variants, one heterozygote nonframeshift deletion-insertion variant, and one homozygous missense variant. Functional assays showed that the transcriptional activity of the 11 variants was significantly reduced. In this study, 11 NR5A1 pathogenic variants were identified. These novel variants further expand the existing spectrum of the NR5A1 variants associated with 46,XY DSD, which will, in turn, assist in the molecular diagnosis of DSD.
Collapse
|
6
|
Canalichio KL, Shnorhavorian M, Oelschlager AMA, Ramsdell L, Fisher C, Adam MP, Fechner PY. A non-surgical approach to 46,XY differences in sex development through hormonal suppression at puberty: a single-center case series study. Endocrine 2020; 70:170-177. [PMID: 32643049 DOI: 10.1007/s12020-020-02409-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 06/27/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE We aim to report outcomes and safety with hormonal suppression to facilitate gonadal preservation in a select group of patients with 46,XY differences in sex development (DSD) who are raised and identify as female yet have diagnoses with potential for androgenization at puberty. METHODS We performed a retrospective review of the past 10 years of DSD patients treated by a multidisciplinary program. Inclusion criteria were 46,XY DSD, female sex of rearing, risk of androgenization at puberty, and plan for hormonal suppression at puberty. Patients on hormonal suppression had at least 6 months of follow-up from initiation. We excluded those with complete gonadal dysgenesis or complete androgen insensitivity. RESULTS Four patients met inclusion criteria. Initial evaluation by DSD team was at a mean age of 6.6 years (3 weeks-16 years). All patients were evaluated in a coordinated multidisciplinary clinic. The diagnoses are listed in Table 1. Mean follow-up was 5.7 years (1.2-10.9 years). One patient presented as an infant, and is being monitored until Tanner stage 2 and/or serum hormonal evidence to initiate hormonal suppression. Three patients have been receiving hormonal suppression for 1.4 years (1.1-1.9 years) without side effects or complication. Three patients were initiated with estrogen replacement to promote desired breast development. At last follow-up, all patients had retained their gonads, all have female gender identity with no reported gender dysphoria, and no progression of androgenization. CONCLUSIONS In our initial experience, gonadal preservation with hormonal suppression is a tool in multidisciplinary management of select DSD patients with female gender identity with conditions associated with androgenization at puberty. Patients' growth, bone health, and overall psychosocial well-being will need to be monitored closely.
Collapse
Affiliation(s)
- Katie L Canalichio
- Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA, 98105, USA.
- University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA.
| | - Margarett Shnorhavorian
- Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA, 98105, USA
- University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Anne-Marie Amies Oelschlager
- Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA, 98105, USA
- University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Linda Ramsdell
- Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA, 98105, USA
| | - Christina Fisher
- Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA, 98105, USA
| | - Margaret P Adam
- Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA, 98105, USA
- University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Patricia Y Fechner
- Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA, 98105, USA
- University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| |
Collapse
|
7
|
Xu C, Dai Y, Mohsin A, Hang H, Zhuang Y, Guo M. Mapping molecular pathways for embryonic Sertoli cells derivation based on differentiation model of mouse embryonic stem cells. Stem Cell Res Ther 2020; 11:85. [PMID: 32102677 PMCID: PMC7045406 DOI: 10.1186/s13287-020-01600-2] [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: 12/16/2019] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 12/27/2022] Open
Abstract
Background Embryonic Sertoli cells (eSCs) have been known for playing important roles in male reproductive development system. In current studies, eSCs were mainly generated from induced intermediate mesoderm. The deriving mechanism of eSCs has been unclear so far. Therefore, this work was aimed to reveal the molecular pathways during derivation of eSCs. Methods In this scenario, a differentiation model from mouse embryonic stem cells (mESCs) to eSCs was established through spatiotemporal control of 5 key factors, Wilms tumor 1 homolog (Wt1), GATA binding protein 4 (Gata4), nuclear receptor subfamily 5, group A, member 1 (Nr5a1, i.e., Sf1), SRY (sex determining region Y)-box 9 (Sox9), doublesex, and mab-3 related transcription factor 1 (Dmrt1). To investigate the molecular mechanism, these key factors were respectively manipulated through a light-switchable (light-on) system, tetracycline-switchable (Tet-on) system, and CRISPR/Cas9 knock out (KO) system. Results Via the established approach, some embryonic Sertoli-like cells (eSLCs) were induced from mESCs and formed ring-like or tubular-like structures. The key factors were respectively manipulated and revealed their roles in the derivation of these eSLCs. Based on these results, some molecular pathways were mapped during the development of coelomic epithelial somatic cells to eSCs. Conclusions This differentiation model provided a high controllability of some key factors and brought a novel insight into the deriving mechanism of Sertoli cells. Supplementary information accompanies this paper at 10.1186/s13287-020-01600-2.
Collapse
Affiliation(s)
- Chenze Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Yichen Dai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Ali Mohsin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Haifeng Hang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China.,Engineering Research Centre of Processes System, Ministry of Education, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Yingping Zhuang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China.,Engineering Research Centre of Processes System, Ministry of Education, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China. .,Engineering Research Centre of Processes System, Ministry of Education, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
8
|
Bertrand-Delepine J, Manouvrier-Hanu S, Cartigny M, Paris F, Mallet D, Philibert P, Morel Y, Lefevre C, Dewailly D, Catteau-Jonard S. In cases of familial primary ovarian insufficiency and disorders of gonadal development, consider NR5A1/SF-1 sequence variants. Reprod Biomed Online 2020; 40:151-159. [PMID: 31831369 DOI: 10.1016/j.rbmo.2019.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 09/24/2019] [Accepted: 10/02/2019] [Indexed: 11/21/2022]
Abstract
RESEARCH QUESTION Primary ovarian insufficiency (POI) is defined as the early exhaustion of ovarian function, before the age of 40 years. Its origin is genetic in 20-25% of cases. In rare cases, sequence variants of the NR5A1/SF-1 gene may result in POI, or in various disorders of gonadal development (DGD) or adrenal insufficiency. DESIGN This study describes the cases of two families in which the association of DGD and POI enabled a diagnosis of NR5A1 deleterious variations. Their clinical, hormonal, ultrasound and genetic characteristics are reported. RESULTS The mothers of the affected children were 21 and 29 years when POI was diagnosed. Each nonetheless had two spontaneous pregnancies. The children have different phenotypes and different forms of DGD. None of the affected family members had adrenal insufficiency. A new sequence variant of the NR5A1 gene was identified in one family: p.Cys283Phe (c.848G>T), and the NR5A1 sequence variant c.86G>C was found in the other family. CONCLUSION Sequence variation of the NR5A1 gene is a possibility that must be considered when a woman with POI or a diminished ovarian reserve has a family member or child with DGD. If a variant is identified, genetic counselling is essential for the patient and his/her family.
Collapse
Affiliation(s)
| | - Sylvie Manouvrier-Hanu
- Université de Lille. Lille, CHU Lille, Clinique de Génétique, EA 7364-RADEME, Lille F-59000, France
| | - Maryse Cartigny
- CHU Lille, Centre de Référence DEV-GEN, Lille F-59000, France
| | - Françoise Paris
- Département d'Endocrinologie et de Gynécologie Pédiatrique CHU Arnaud de Villeneuve, 34090 Montpellier, Département de Génétique, IURC, Equipe DEV-GEN, Montpellier 34090, France
| | - Delphine Mallet
- CHU Lyon, Laboratoire de Biochimie et Biologie Moléculaire Grand Est, UM Pathologies Endocriniennes Rénales Musculaires et Mucoviscidose - Centre de Référence DEV-GEN, Bron F-69677, France
| | - Pascal Philibert
- Département d'Endocrinologie et de Gynécologie Pédiatrique CHU Arnaud de Villeneuve, 34090 Montpellier, Département de Génétique, IURC, Equipe DEV-GEN, Montpellier 34090, France
| | - Yves Morel
- CHU Lyon, Laboratoire de Biochimie et Biologie Moléculaire Grand Est, UM Pathologies Endocriniennes Rénales Musculaires et Mucoviscidose - Centre de Référence DEV-GEN, Bron F-69677, France; Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Didier Dewailly
- Université de Lille, CHU Lille, INSERM U1172, Lille F-59000, France
| | - Sophie Catteau-Jonard
- Université de Lille, CHU Lille, INSERM U1172, Lille F-59000, France; Université de Lille, CHU Lille, Department of Reproductive Medicine, Lille F-59000, France.
| |
Collapse
|
9
|
da Silva TE, Gomes NL, Lerário AM, Keegan CE, Nishi MY, Carvalho FM, Vilain E, Barseghyan H, Martinez-Aguayo A, Forclaz MV, Papazian R, Pedroso de Paula LC, Costa EC, Carvalho LR, Jorge AAL, Elias FM, Mitchell R, Costa EMF, Mendonca BB, Domenice S. Genetic Evidence of the Association of DEAH-Box Helicase 37 Defects With 46,XY Gonadal Dysgenesis Spectrum. J Clin Endocrinol Metab 2019; 104:5923-5934. [PMID: 31287541 DOI: 10.1210/jc.2019-00984] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/02/2019] [Indexed: 01/07/2023]
Abstract
CONTEXT 46,XY Gonadal dysgenesis (GD) is a heterogeneous group of disorders with a wide phenotypic spectrum, including embryonic testicular regression syndrome (ETRS). OBJECTIVE To report a gene for 46,XY GD etiology, especially for ETRS. DESIGN Screening of familial cases of 46,XY GD using whole-exome sequencing and sporadic cases by target gene-panel sequencing. SETTING Tertiary Referral Center for differences/disorders of sex development (DSD). PATIENTS AND INTERVENTIONS We selected 87 patients with 46,XY DSD (17 familial cases from 8 unrelated families and 70 sporadic cases); 55 patients had GD (among them, 10 patients from 5 families and 8 sporadic cases had ETRS), and 32 patients had 46,XY DSD of unknown etiology. RESULTS We identified four heterozygous missense rare variants, classified as pathogenic or likely pathogenic in the Asp-Glu-Ala-His-box (DHX) helicase 37 (DHX37) gene in five families (n = 11 patients) and in six sporadic cases. Two variants were recurrent: p.Arg308Gln (in two families and in three sporadic cases) and p.Arg674Trp (in two families and in two sporadic cases). The variants were specifically associated with ETRS (7/14 index cases; 50%). The frequency of rare, predicted-to-be-deleterious DHX37 variants in this cohort (14%) is significantly higher than that observed in the Genome Aggregation Database (0.4%; P < 0.001). Immunohistochemistry analysis in human testis showed that DHX37 is mainly expressed in germ cells at different stages of testis maturation, in Leydig cells, and rarely in Sertoli cells. CONCLUSION This strong genetic evidence identifies DHX37 as a player in the complex cascade of male gonadal differentiation and maintenance.
Collapse
Affiliation(s)
- Thatiana Evilen da Silva
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Nathalia Lisboa Gomes
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Antonio Marcondes Lerário
- Laboratório de Sequenciamento em Larga Escala, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Catherine Elizabeth Keegan
- Department of Pediatric Genetics, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Mirian Yumi Nishi
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Eric Vilain
- Center for Genetic Medicine Research, The Children's Research Institute, Children's National Medical Center, Children's National Health System, Washington, DC
| | - Hayk Barseghyan
- Center for Genetic Medicine Research, The Children's Research Institute, Children's National Medical Center, Children's National Health System, Washington, DC
| | - Alejandro Martinez-Aguayo
- Division de Pediatria, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | | | - Regina Papazian
- Servicio de Pediatría, Hospital Nacional Prof. Dr. A. Posadas, Buenos Aires, Argentina
| | - Leila Cristina Pedroso de Paula
- Programa de Atendimento às Desordens do Desenvolvimento Sexual, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduardo Corrêa Costa
- Programa de Atendimento às Desordens do Desenvolvimento Sexual, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luciani Renata Carvalho
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alexander Augusto Lima Jorge
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Felipe Martins Elias
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Rod Mitchell
- Medical Research Council Centre for Reproductive Health, Queens Medical Research Institute, Edinburgh, United Kingdom
| | - Elaine Maria Frade Costa
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Berenice Bilharinho Mendonca
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Sequenciamento em Larga Escala, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Sorahia Domenice
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Buonocore F, Clifford-Mobley O, King TFJ, Striglioni N, Man E, Suntharalingham JP, del Valle I, Lin L, Lagos CF, Rumsby G, Conway GS, Achermann JC. Next-Generation Sequencing Reveals Novel Genetic Variants (SRY, DMRT1, NR5A1, DHH, DHX37) in Adults With 46,XY DSD. J Endocr Soc 2019; 3:2341-2360. [PMID: 31745530 PMCID: PMC6855215 DOI: 10.1210/js.2019-00306] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/04/2019] [Indexed: 12/11/2022] Open
Abstract
CONTEXT The genetic basis of human sex development is slowly being elucidated, and >40 different genetic causes of differences (or disorders) of sex development (DSDs) have now been reported. However, reaching a specific diagnosis using traditional approaches can be difficult, especially in adults where limited biochemical data may be available. OBJECTIVE We used a targeted next-generation sequencing approach to analyze known and candidate genes for DSDs in individuals with no specific molecular diagnosis. PARTICIPANTS AND DESIGN We studied 52 adult 46,XY women attending a single-center adult service, who were part of a larger cohort of 400 individuals. Classic conditions such as17β-hydroxysteroid dehydrogenase deficiency type 3, 5α-reductase deficiency type 2, and androgen insensitivity syndrome were excluded. The study cohort had broad working diagnoses of complete gonadal dysgenesis (CGD) (n = 27) and partially virilized 46,XY DSD (pvDSD) (n = 25), a group that included partial gonadal dysgenesis and those with a broad "partial androgen insensitivity syndrome" label. Targeted sequencing of 180 genes was undertaken. RESULTS Overall, a likely genetic cause was found in 16 of 52 (30.8%) individuals (22.2% CGD, 40.0% pvDSD). Pathogenic variants were found in sex-determining region Y (SRY; n = 3), doublesex and mab-3-related transcription factor 1 (DMRT1; n = 1), NR5A1/steroidogenic factor-1 (SF-1) (n = 1), and desert hedgehog (DHH; n = 1) in the CGD group, and in NR5A1 (n = 5), DHH (n = 1), and DEAH-box helicase 37 (DHX37; n = 4) in the pvDSD group. CONCLUSIONS Reaching a specific diagnosis can have clinical implications and provides insight into the role of these proteins in sex development. Next-generation sequencing approaches are invaluable, especially in adult populations or where diagnostic biochemistry is not possible.
Collapse
Affiliation(s)
- Federica Buonocore
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | | | - Tom F J King
- Reproductive Medicine Unit, University College London Hospitals, London, United Kingdom
| | - Niccolò Striglioni
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Elim Man
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Jenifer P Suntharalingham
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Ignacio del Valle
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Lin Lin
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Carlos F Lagos
- Chemical Biology and Drug Discovery Laboratory, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Gill Rumsby
- Clinical Biochemistry, University College London Hospitals, London, United Kingdom
| | - Gerard S Conway
- Reproductive Medicine Unit, University College London Hospitals, London, United Kingdom
| | - John C Achermann
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
11
|
Fabbri‐Scallet H, Sousa LM, Maciel‐Guerra AT, Guerra‐Júnior G, Mello MP. Mutation update for theNR5A1gene involved in DSD and infertility. Hum Mutat 2019; 41:58-68. [DOI: 10.1002/humu.23916] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/29/2019] [Accepted: 09/09/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Helena Fabbri‐Scallet
- Center for Molecular Biology and Genetic Engineering‐CBMEGState University of Campinas São Paulo Brazil
| | - Lizandra Maia Sousa
- Center for Molecular Biology and Genetic Engineering‐CBMEGState University of Campinas São Paulo Brazil
| | - Andréa Trevas Maciel‐Guerra
- Department of Medical Genetics and Genomic Medicine, Faculty of Medical SciencesState University of Campinas São Paulo Brazil
- Interdisciplinary Group for the Study of Sex Determination and Differentiation‐GIEDDSState University of Campinas São Paulo Brazil
| | - Gil Guerra‐Júnior
- Interdisciplinary Group for the Study of Sex Determination and Differentiation‐GIEDDSState University of Campinas São Paulo Brazil
- Department of Pediatrics, Faculty of Medical SciencesState University of Campinas São Paulo Brazil
| | - Maricilda Palandi Mello
- Center for Molecular Biology and Genetic Engineering‐CBMEGState University of Campinas São Paulo Brazil
| |
Collapse
|
12
|
Parivesh A, Barseghyan H, Délot E, Vilain E. Translating genomics to the clinical diagnosis of disorders/differences of sex development. Curr Top Dev Biol 2019; 134:317-375. [PMID: 30999980 PMCID: PMC7382024 DOI: 10.1016/bs.ctdb.2019.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The medical and psychosocial challenges faced by patients living with Disorders/Differences of Sex Development (DSD) and their families can be alleviated by a rapid and accurate diagnostic process. Clinical diagnosis of DSD is limited by a lack of standardization of anatomical and endocrine phenotyping and genetic testing, as well as poor genotype/phenotype correlation. Historically, DSD genes have been identified through positional cloning of disease-associated variants segregating in families and validation of candidates in animal and in vitro modeling of variant pathogenicity. Owing to the complexity of conditions grouped under DSD, genome-wide scanning methods are better suited for identifying disease causing gene variant(s) and providing a clinical diagnosis. Here, we review a number of established genomic tools (karyotyping, chromosomal microarrays and exome sequencing) used in clinic for DSD diagnosis, as well as emerging genomic technologies such as whole-genome (short-read) sequencing, long-read sequencing, and optical mapping used for novel DSD gene discovery. These, together with gene expression and epigenetic studies can potentiate the clinical diagnosis of DSD diagnostic rates and enhance the outcomes for patients and families.
Collapse
Affiliation(s)
- Abhinav Parivesh
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, United States
| | - Hayk Barseghyan
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, United States; Department of Genomics and Precision Medicine, The George Washington University, Washington, DC, United States
| | - Emmanuèle Délot
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, United States; Department of Genomics and Precision Medicine, The George Washington University, Washington, DC, United States.
| | - Eric Vilain
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, United States; Department of Genomics and Precision Medicine, The George Washington University, Washington, DC, United States.
| |
Collapse
|
13
|
Rocca MS, Ortolano R, Menabò S, Baronio F, Cassio A, Russo G, Balsamo A, Ferlin A, Baldazzi L. Mutational and functional studies on NR5A1 gene in 46,XY disorders of sex development: identification of six novel loss of function mutations. Fertil Steril 2019; 109:1105-1113. [PMID: 29935645 DOI: 10.1016/j.fertnstert.2018.02.123] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/30/2018] [Accepted: 02/19/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To study the functional properties of six novel missense mutations of the NR5A1 gene encoding the steroidogenic factor 1 (SF-1) identified in six patients with 46,XY disorders of sex development (DSD) and to describe their relative phenotype-genotype relationship. DESIGN Genetic and functional studies. SETTING University department. PATIENT(S) Six 46,XY DSD patients. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Sanger sequencing and multiplex ligation-dependent probe amplification analysis to identify the mutations or deletions/duplications of the NR5A1 gene. Functional studies by transactivation assays to predict the impact of mutations on molecular function. RESULT(S) NR5A1 exons sequencing identified in six 46,XY DSD patients six novel mutations: p.T40R, p.T47C, p.G328W, p.A351E, p.R427W, and p.Q460R. Five missense variants were heterozygous, and one was homozygous (p.R427W). Functional analysis revealed a significant loss of DNA-binding and transactivation ability for all variants, except for p.Q460R, which showed a modest reduced activity compared with that of the wild-type protein. Phenotypes associated with these mutations varied from males with spontaneous puberty, substantial T production, and possible fertility, to females with and without müllerian structures and primary amenorrhea. CONCLUSION(S) We describe six novel mutations in NR5A1 gene and showed that they might affect protein structure, therefore compromising seriously the SF-1 role in regulating gonadal development. Clinically, we suggest that NR5A1 analysis should be performed whenever atypical sex organs are evidenced or there is an abnormal sexual development, to have proper diagnosis and better management of patients.
Collapse
Affiliation(s)
- Maria Santa Rocca
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | - Rita Ortolano
- Centre for Rare Endocrine Conditions, Department of Women, Children and Urological Diseases, S. Orsola Malpighi University Hospital, Bologna, Italy
| | - Soara Menabò
- Program of Endocrinology, Pediatric Unit, Department of Medical and Surgical Sciences, S. Orsola Malpighi University Hospital, Bologna, Italy
| | - Federico Baronio
- Centre for Rare Endocrine Conditions, Department of Women, Children and Urological Diseases, S. Orsola Malpighi University Hospital, Bologna, Italy
| | - Alessandra Cassio
- Program of Endocrinology, Pediatric Unit, Department of Medical and Surgical Sciences, S. Orsola Malpighi University Hospital, Bologna, Italy
| | - Gianni Russo
- Centre for Rare Endocrine Conditions, Scientific Institute San Raffaele, Milan, Italy
| | - Antonio Balsamo
- Program of Endocrinology, Pediatric Unit, Department of Medical and Surgical Sciences, S. Orsola Malpighi University Hospital, Bologna, Italy
| | - Alberto Ferlin
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | - Lilia Baldazzi
- Centre for Rare Endocrine Conditions, Department of Women, Children and Urological Diseases, S. Orsola Malpighi University Hospital, Bologna, Italy.
| |
Collapse
|
14
|
Song Y, Fan L, Gong C. Phenotype and Molecular Characterizations of 30 Children From China With NR5A1 Mutations. Front Pharmacol 2018; 9:1224. [PMID: 30425642 PMCID: PMC6218886 DOI: 10.3389/fphar.2018.01224] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/08/2018] [Indexed: 01/31/2023] Open
Abstract
Background: Patients harboring NR5A1 mutations have a wide spectrum of phenotypes. Objective: To investigate the phenotype of patients with NR5A1 gene mutations from a 30 Chinese patient cohort. Methods: We reported the clinical features of children with NR5A1 gene mutations and compared them between two groups of patients with social genders of male (boys group) and female (girls group). Results: Thirty patients with NR5A1 mutations ranging from 2 months to 17 years of age were studied. There were 11 boys and 19 girls who were identified when they visited the hospital. The patients were verified as having testes without a uterus and ovaries by B-mode ultrasound. There was no difference between boys and girls in terms of the Prader stage (p = 0.086), but the position of the testes was higher in girls than in boys (p = 0.013). The patients’ average height is −0.43 SDS according to the normal boys’ height with SDS (while their average target height was 0.07 SDS). However, there was no such difference between boys and girls (p > 0.05). Although the basal LH and post-hCG testosterone (T) levels were not different (p > 0.05), but the basal FSH level, LH/FSH ratio, and INHB level were decreased in girls (p = 0.002; p = 0.001; p = 0.006). All of the mothers of the patients reported to have normal pregnancies. We found 24 patients (80%) with de novo mutations in the NR5A1 gene; 5 patients had inherited mutations from their mothers, and one inherited from the father. Only the mothers of patients 16 and 18 showed premature ovarian failure at the time of reporting. Among 26 disease associated mutations, 14 novel mutations that have been reported the first time and p.R87C is the most common Among the other 12 had had been reported,the p.R313C is the most common. Conclusion: Patients with 46, XY NR5A1 mutations presented a wide spectrum of external genitalia characteristics and severe Sertoli cell impairment. The p.R87C and p.R313C mutations appeared to be common (10%) in this group, and 14 new mutations were identified, improving our understanding the genotype phenotype correlations.
Collapse
Affiliation(s)
- Yanning Song
- Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Lijun Fan
- Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chunxiu Gong
- Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.,Beijing Key Laboratory for Genetics of Birth Defects, Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Sreenivasan R, Ludbrook L, Fisher B, Declosmenil F, Knower KC, Croft B, Bird AD, Ryan J, Bashamboo A, Sinclair AH, Koopman P, McElreavey K, Poulat F, Harley VR. Mutant NR5A1/SF-1 in patients with disorders of sex development shows defective activation of the SOX9 TESCO enhancer. Hum Mutat 2018; 39:1861-1874. [PMID: 30067310 DOI: 10.1002/humu.23603] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 07/19/2018] [Accepted: 07/19/2018] [Indexed: 11/09/2022]
Abstract
Nuclear receptor subfamily 5 group A member 1/Steroidogenic factor 1 (NR5A1; SF-1; Ad4BP) mutations cause 46,XY disorders of sex development (DSD), with phenotypes ranging from developmentally mild (e.g., hypospadias) to severe (e.g., complete gonadal dysgenesis). The molecular mechanism underlying this spectrum is unclear. During sex determination, SF-1 regulates SOX9 (SRY [sex determining region Y]-box 9) expression. We hypothesized that SF-1 mutations in 46,XY DSD patients affect SOX9 expression via the Testis-specific Enhancer of Sox9 core element, TESCO. Our objective was to assess the ability of 20 SF-1 mutants found in 46,XY DSD patients to activate TESCO. Patient DNA was sequenced for SF-1 mutations and mutant SF-1 proteins were examined for transcriptional activity, protein expression, sub-cellular localization and in silico structural defects. Fifteen of the 20 mutants showed reduced SF-1 activation on TESCO, 11 with atypical sub-cellular localization. Fourteen SF-1 mutants were predicted in silico to alter DNA, ligand or cofactor interactions. Our study may implicate aberrant SF-1-mediated transcriptional regulation of SOX9 in 46,XY DSDs.
Collapse
Affiliation(s)
- Rajini Sreenivasan
- Hudson Institute of Medical Research, Victoria, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia
| | - Louisa Ludbrook
- Hudson Institute of Medical Research, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Brett Fisher
- Hudson Institute of Medical Research, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | | | - Kevin C Knower
- Hudson Institute of Medical Research, Victoria, Australia
| | - Brittany Croft
- Hudson Institute of Medical Research, Victoria, Australia.,Department of Molecular Translational Science, Monash University, Victoria, Australia
| | - Anthony D Bird
- Hudson Institute of Medical Research, Victoria, Australia
| | - Janelle Ryan
- Hudson Institute of Medical Research, Victoria, Australia
| | | | - Andrew H Sinclair
- Murdoch Children's Research Institute, Royal Children's Hospital and Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | | | - Francis Poulat
- Department of Molecular Translational Science, Monash University, Victoria, Australia
| | - Vincent R Harley
- Hudson Institute of Medical Research, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia.,Department of Anatomy and Developmental Biology, Monash University, Victoria, Australia
| |
Collapse
|
16
|
Robevska G, van den Bergen JA, Ohnesorg T, Eggers S, Hanna C, Hersmus R, Thompson EM, Baxendale A, Verge CF, Lafferty AR, Marzuki NS, Santosa A, Listyasari NA, Riedl S, Warne G, Looijenga L, Faradz S, Ayers KL, Sinclair AH. Functional characterization of novel NR5A1 variants reveals multiple complex roles in disorders of sex development. Hum Mutat 2017; 39:124-139. [PMID: 29027299 PMCID: PMC5765430 DOI: 10.1002/humu.23354] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/05/2017] [Accepted: 10/09/2017] [Indexed: 12/23/2022]
Abstract
Variants in the NR5A1 gene encoding SF1 have been described in a diverse spectrum of disorders of sex development (DSD). Recently, we reported the use of a targeted gene panel for DSD where we identified 15 individuals with a variant in NR5A1, nine of which are novel. Here, we examine the functional effect of these changes in relation to the patient phenotype. All novel variants tested had reduced trans‐activational activity, while several had altered protein level, localization, or conformation. In addition, we found evidence of new roles for SF1 protein domains including a region within the ligand binding domain that appears to contribute to SF1 regulation of Müllerian development. There was little correlation between the severity of the phenotype and the nature of the NR5A1 variant. We report two familial cases of NR5A1 deficiency with evidence of variable expressivity; we also report on individuals with oligogenic inheritance. Finally, we found that the nature of the NR5A1 variant does not inform patient outcomes (including pubertal androgenization and malignancy risk). This study adds nine novel pathogenic NR5A1 variants to the pool of diagnostic variants. It highlights a greater need for understanding the complexity of SF1 function and the additional factors that contribute.
Collapse
Affiliation(s)
| | | | | | | | - Chloe Hanna
- Murdoch Children's Research Institute, Melbourne, Australia.,Royal Children's Hospital, Melbourne, Australia
| | - Remko Hersmus
- Department of Pathology, Josephine Nefkens Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Elizabeth M Thompson
- SA Clinical Genetics Service, SA Pathology at the Women's and Children's Hospital, Adelaide, Australia.,School of Medicine, University of Adelaide, Adelaide, Australia
| | - Anne Baxendale
- SA Clinical Genetics Service, SA Pathology at the Women's and Children's Hospital, Adelaide, Australia
| | - Charles F Verge
- Sydney Children's Hospital, Sydney, Australia.,School of Women's and Children's Health, UNSW, Sydney, Australia
| | - Antony R Lafferty
- Centenary Hospital for Women and Children, Canberra, Australia.,ANU Medical School, Canberra, Australia
| | | | - Ardy Santosa
- Division of Urology, Department of Surgery, Dr. Kariadi Hospital, Semarang, Indonesia
| | - Nurin A Listyasari
- Division of Human Genetics, Centre for Biomedical Research Faculty of Medicine Diponegoro University (FMDU), Semarang, Indonesia
| | - Stefan Riedl
- St Anna Children's Hospital, Department of Paediatrics, Medical University of Vienna, Wien, Austria.,Division of Paediatric Pulmology, Allergology, and Endocrinology, Department of Paediatrics, Medical University of Vienna, Wien, Austria
| | - Garry Warne
- Murdoch Children's Research Institute, Melbourne, Australia.,Royal Children's Hospital, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Leendert Looijenga
- Department of Pathology, Josephine Nefkens Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Sultana Faradz
- Division of Human Genetics, Centre for Biomedical Research Faculty of Medicine Diponegoro University (FMDU), Semarang, Indonesia
| | - Katie L Ayers
- Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Andrew H Sinclair
- Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia
| |
Collapse
|
17
|
Werner R, Mönig I, Lünstedt R, Wünsch L, Thorns C, Reiz B, Krause A, Schwab KO, Binder G, Holterhus PM, Hiort O. New NR5A1 mutations and phenotypic variations of gonadal dysgenesis. PLoS One 2017; 12:e0176720. [PMID: 28459839 PMCID: PMC5411087 DOI: 10.1371/journal.pone.0176720] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/15/2017] [Indexed: 02/01/2023] Open
Abstract
Mutations in NR5A1 have been reported as a frequent cause of 46,XY disorders of sex development (DSD) associated to a broad phenotypic spectrum ranging from infertility, ambiguous genitalia, anorchia to gonadal dygenesis and female genitalia. Here we present the clinical follow up of four 46,XY DSD patients with three novel heterozygous mutations in the NR5A1 gene leading to a p.T40P missense mutation and a p.18DKVSG22del nonframeshift deletion in the DNA-binding domain and a familiar p.Y211Tfs*83 frameshift mutation. Functional analysis of the missense and nonframeshift mutation revealed a deleterious character with loss of DNA-binding and transactivation capacity. Both, the mutations in the DNA-binding domain, as well as the familiar frameshift mutation are associated with highly variable endocrine values and phenotypic appearance. Phenotypes vary from males with spontaneous puberty, substantial testosterone production and possible fertility to females with and without Müllerian structures and primary amenorrhea. Exome sequencing of the sibling’s family revealed TBX2 as a possible modifier of gonadal development in patients with NR5A1 mutations.
Collapse
Affiliation(s)
- Ralf Werner
- Department of Paediatrics and Adolescent Medicine, Division of Experimental Paediatric Endocrinology and Diabetes, University of Lübeck, Lübeck, Germany
| | - Isabel Mönig
- Department of Paediatrics and Adolescent Medicine, Division of Experimental Paediatric Endocrinology and Diabetes, University of Lübeck, Lübeck, Germany
| | - Ralf Lünstedt
- Department of Paediatrics and Adolescent Medicine, Division of Experimental Paediatric Endocrinology and Diabetes, University of Lübeck, Lübeck, Germany
| | - Lutz Wünsch
- Department of Paediatric Surgery, University Hospital of Lübeck, Germany
| | - Christoph Thorns
- Department of Pathology, University Hospital of Lübeck, Lübeck, Germany
| | - Benedikt Reiz
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Alexandra Krause
- Department of Paediatrics and Adolescent Medicine, Paediatric Endocrinology and Diabetes, University Hospital Freiburg, Freiburg, Germany
| | - Karl Otfried Schwab
- Department of Paediatrics and Adolescent Medicine, Paediatric Endocrinology and Diabetes, University Hospital Freiburg, Freiburg, Germany
| | - Gerhard Binder
- Department of Paediatrics and Adolescent Medicine, Eberhard-Karls-University of Tübingen, Tübingen, Germany
| | - Paul-Martin Holterhus
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics, Christian-Albrechts-University, Kiel, Germany
| | - Olaf Hiort
- Department of Paediatrics and Adolescent Medicine, Division of Experimental Paediatric Endocrinology and Diabetes, University of Lübeck, Lübeck, Germany
- * E-mail:
| |
Collapse
|
18
|
Swartz JM, Ciarlo R, Guo MH, Abrha A, Diamond DA, Chan YM, Hirschhorn JN. Two Unrelated Undervirilized 46,XY Males with Inherited NR5A1 Variants Identified by Whole-Exome Sequencing. Horm Res Paediatr 2016; 87:264-270. [PMID: 27553487 PMCID: PMC5325809 DOI: 10.1159/000448754] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/28/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Undervirilized 46,XY males with bifid scrotum often pose a diagnostic challenge, and the majority of cases typically do not receive a genetic diagnosis. NR5A1 mutations can be seen in 10-20% of the cases and are a relatively common cause of undervirilization. METHODS Whole-exome sequencing was utilized to study 10 undervirilized 46,XY subjects with bifid scrotum. RESULTS Exome sequencing identified novel NR5A1 variants, both affecting exon 7, in 2 of the 10 subjects with bifid scrotum. Subject 1 had a heterozygous frameshift variant, c.1150delC, p.Leu384fsTer1, within the ligand-binding domain inherited from his unaffected father. Subject 2 had a novel splice-site variant c.1139-2T>C, affecting the canonical splice acceptor site for exon 7 and also disrupting the ligand-binding domain. Both subjects had serum testosterone levels within the normal range as infants. CONCLUSIONS We describe two novel NR5A1 variants, demonstrating mutations in this gene as a common cause of milder cases of 46,XY undervirilization. Whole-exome sequencing results yielded the diagnosis in 2 out of 10 cases without a previous diagnosis, supporting the value of this approach. Significant genotype-phenotype variability was also noted with Subject 1's paternal inheritance from his unaffected father.
Collapse
Affiliation(s)
- Jonathan M Swartz
- Division of Endocrinology, Boston Children's Hospital, Boston, Mass., USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Woo KH, Cheon B, Kim JH, Cho J, Kim GH, Yoo HW, Choi JH. Novel Heterozygous Mutations of NR5A1 and Their Functional Characteristics in Patients with 46,XY Disorders of Sex Development without Adrenal Insufficiency. Horm Res Paediatr 2016; 84:116-23. [PMID: 26139438 DOI: 10.1159/000431324] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 05/11/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Heterozygous mutations of NR5A1, which encodes steroidogenic factor 1 (SF1), were identified in patients with 46,XY disorders of sex development (DSD) with normal adrenal function. This study was aimed to identify and functionally characterize mutations of NR5A1 in patients with 46,XY DSD. METHODS This study included 51 patients from 49 unrelated families with 46,XY DSD. Genomic DNA was extracted from peripheral blood leukocytes, and direct sequencing of all coding exons and their flanking introns of NR5A1 was performed. Transient transfections and dual-luciferase® reporter assays were performed to evaluate the effect of NR5A1 variants on transcriptional activity. RESULTS Four of 49 patients (8.2%) harbored a novel heterozygous sequence variant of NR5A1: c.80G>C (p.G26A), c.847T>C (p.C283R), c.1151del (p.L384Rfs*7), and c.1333G>T (p.E445*). They presented with female external genitalia with clitoromegaly in infancy or childhood, or primary amenorrhea in adolescence. In vitro functional studies of SF1 activity determined that each variant, except p.E445*, led to a reduced expression of downstream target genes and disturbed the regulation of gonadal development. CONCLUSIONS Loss-of-function mutations of NR5A1 are a relatively common cause of 46,XY DSD. Therefore, genetic defects of NR5A1 should be considered as an etiology in subjects with 46,XY DSD without adrenal insufficiency.
Collapse
Affiliation(s)
- Kyu Ha Woo
- Genome Research Center for Birth Defects and Genetic Diseases, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
20
|
Tuhan H, Anik A, Catli G, Onay H, Aykut A, Abaci A, Bober E. A novel mutation in steroidogenic factor (SF1/NR5A1) gene in a patient with 46 XY DSD without adrenal insufficiency. Andrologia 2016; 49. [PMID: 27135758 DOI: 10.1111/and.12589] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2016] [Indexed: 11/30/2022] Open
Abstract
Steroidogenic factor-1 (SF-1), also known as nuclear receptor subfamily 5 group A member 1 (NR5A1), is a member of orphan receptor subfamily and located on chromosome 9 (9q33). In 46, XY individuals with mutation of SF-1 gene, adrenal failure, testis dysgenesis, androgen synthesis defects, hypospadias and anorchia with microphallus, infertility can occur from severe to mild. We report a case of a 20-day-old male who is admitted to our clinic due to ambiguous genitalia. In this report, we describe a novel heterozygous c.814A > C (p. T272P) NR5A1 mutation in a patient with 46, XY DSD without adrenal insufficiency. We describe a novel missense mutation c.814A > C (p. T272P) in NR5A1 gene which had not previously been reported. Also this report highlights that the potential diagnostic utility of next-generation sequencing is an effective strategy versus Sanger sequencing to identify genetic mosaicism in clinical practice.
Collapse
Affiliation(s)
- H Tuhan
- Department of Pediatric Endocrinology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - A Anik
- Department of Pediatric Endocrinology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - G Catli
- Department of Pediatric Endocrinology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - H Onay
- Department of Medical Genetics, School of Medicine, Ege University, Izmir, Turkey
| | - A Aykut
- Department of Medical Genetics, School of Medicine, Ege University, Izmir, Turkey
| | - A Abaci
- Department of Pediatric Endocrinology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - E Bober
- Department of Pediatric Endocrinology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
21
|
Werner R, M�nig I, August J, Freiberg C, L�nstedt R, Reiz B, W�nsch L, Holterhus PM, Kulle A, D�hnert U, Wudy SA, Richter-Unruh A, Thorns C, Hiort O. Novel Insights into 46,XY Disorders of Sex Development due to NR5A1 Gene Mutation. Sex Dev 2015; 9:260-8. [DOI: 10.1159/000442309] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2015] [Indexed: 11/19/2022] Open
|
22
|
Yagi H, Takagi M, Kon M, Igarashi M, Fukami M, Hasegawa Y. Fertility preservation in a family with a novel NR5A1 mutation. Endocr J 2015; 62:289-95. [PMID: 25502990 DOI: 10.1507/endocrj.ej14-0340] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The common phenotype of nuclear receptor superfamily 5, group A, member 1 (NR5A1) gene mutations in 46,XY is gonadal dysgenesis without adrenal deficiency. Though the phenotype of gonadal dysgenesis is variable, ranging from complete female to normal male genitalia, an asymptomatic 46,XY male is rare. Preserved fertility has so far been described in only three affected 46,XY males with different mutations, but no functional analysis of these mutations has been performed. Here, we report on male siblings with hypospadias and their asymptomatic father in whom we identified a heterozygous NR5A1 mutation of c.910G>A, p.E304K. Western blotting and subcellular localization revealed no significant difference between the wild type (WT) and E304K. Electrophoretic mobility shift assay experiments showed that E304K abrogated DNA-binding ability. E304K reduced transactivation and had no dominant negative effect. In conclusion, we report on a novel hypomorphic NR5A1 mutation, which may be associated with the phenotype of the family.
Collapse
Affiliation(s)
- Hiroko Yagi
- Division of Genetic Research, Tokyo Metropolitan Children's Medical Center, Tokyo 183-8561, Japan
| | | | | | | | | | | |
Collapse
|
23
|
Baetens D, Mladenov W, Delle Chiaie B, Menten B, Desloovere A, Iotova V, Callewaert B, Van Laecke E, Hoebeke P, De Baere E, Cools M. Extensive clinical, hormonal and genetic screening in a large consecutive series of 46,XY neonates and infants with atypical sexual development. Orphanet J Rare Dis 2014; 9:209. [PMID: 25497574 PMCID: PMC4271496 DOI: 10.1186/s13023-014-0209-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 12/05/2014] [Indexed: 01/22/2023] Open
Abstract
Background One in 4500 children is born with ambiguous genitalia, milder phenotypes occur in one in 300 newborns. Conventional time-consuming hormonal and genetic work-up provides a genetic diagnosis in around 20-40% of 46,XY cases with ambiguous genitalia. All others remain without a definitive diagnosis. The investigation of milder cases, as suggested by recent reports remains controversial. Methods Integrated clinical, hormonal and genetic screening was performed in a sequential series of 46, XY children, sex-assigned male, who were referred to our pediatric endocrine service for atypical genitalia (2007–2013). Results A consecutive cohort of undervirilized 46,XY children with external masculinization score (EMS) 2–12, was extensively investigated. In four patients, a clinical diagnosis of Kallmann syndrome or Mowat-Wilson syndrome was made and genetically supported in 2/3 and 1/1 cases respectively. Hormonal data were suggestive of a (dihydro)testosterone biosynthesis disorder in four cases, however no HSD17B3 or SRD5A2 mutations were found. Array-CGH revealed a causal structural variation in 2/6 syndromic patients. In addition, three novel NR5A1 mutations were found in non-syndromic patients. Interestingly, one mutation was present in a fertile male, underlining the inter- and intrafamilial phenotypic variability of NR5A1-associated phenotypes. No AR, SRY or WT1 mutations were identified. Conclusion Overall, a genetic diagnosis could be established in 19% of non-syndromic and 33% of syndromic cases. There is no difference in diagnostic yield between patients with more or less pronounced phenotypes, as expressed by the external masculinisation score (EMS). The clinical utility of array-CGH is high in syndromic cases. Finally, a sequential gene-by-gene approach is time-consuming, expensive and inefficient. Given the low yield and high expense of Sanger sequencing, we anticipate that massively parallel sequencing of gene panels and whole exome sequencing hold promise for genetic diagnosis of 46,XY DSD boys with an undervirilized phenotype. Electronic supplementary material The online version of this article (doi:10.1186/s13023-014-0209-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dorien Baetens
- Center for Medical Genetics, Ghent University Hospital and Ghent University, Ghent, Belgium.
| | - Wilhelm Mladenov
- Department of Pediatric Endocrinology, Ghent University Hospital and Ghent University, Building 3K12D, De Pintelaan 185, 9000, Ghent, Belgium. .,Department of Pediatrics and Medical Genetics, Medical University of Varna, University Hospital "Sveta Marina", Varna, Bulgaria.
| | - Barbara Delle Chiaie
- Center for Medical Genetics, Ghent University Hospital and Ghent University, Ghent, Belgium.
| | - Björn Menten
- Center for Medical Genetics, Ghent University Hospital and Ghent University, Ghent, Belgium.
| | - An Desloovere
- Department of Pediatric Endocrinology, Ghent University Hospital and Ghent University, Building 3K12D, De Pintelaan 185, 9000, Ghent, Belgium.
| | - Violeta Iotova
- Department of Pediatrics and Medical Genetics, Medical University of Varna, University Hospital "Sveta Marina", Varna, Bulgaria.
| | - Bert Callewaert
- Center for Medical Genetics, Ghent University Hospital and Ghent University, Ghent, Belgium.
| | - Erik Van Laecke
- Department of Pediatric Urology, Ghent University Hospital and Ghent University, Ghent, Belgium.
| | - Piet Hoebeke
- Department of Pediatric Urology, Ghent University Hospital and Ghent University, Ghent, Belgium.
| | - Elfride De Baere
- Center for Medical Genetics, Ghent University Hospital and Ghent University, Ghent, Belgium.
| | - Martine Cools
- Department of Pediatric Endocrinology, Ghent University Hospital and Ghent University, Building 3K12D, De Pintelaan 185, 9000, Ghent, Belgium.
| |
Collapse
|
24
|
Pedace L, Laino L, Preziosi N, Valentini MS, Scommegna S, Rapone AM, Guarino N, Boscherini B, De Bernardo C, Marrocco G, Majore S, Grammatico P. Longitudinal hormonal evaluation in a patient with disorder of sexual development, 46,XY karyotype and oneNR5A1mutation. Am J Med Genet A 2014; 164A:2938-46. [DOI: 10.1002/ajmg.a.36729] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/09/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Lucia Pedace
- Medical Genetics, Department of Molecular Medicine; Sapienza University, S. Camillo-Forlanini Hospital; Rome Italy
| | - Luigi Laino
- Medical Genetics, Department of Molecular Medicine; Sapienza University, S. Camillo-Forlanini Hospital; Rome Italy
| | - Nicoletta Preziosi
- Medical Genetics, Department of Molecular Medicine; Sapienza University, S. Camillo-Forlanini Hospital; Rome Italy
| | - Maria Stella Valentini
- Medical Genetics, Department of Molecular Medicine; Sapienza University, S. Camillo-Forlanini Hospital; Rome Italy
| | - Salvatore Scommegna
- Pediatrics and Pediatric Hematology, S. Camillo-Forlanini Hospital; Rome Italy
| | - Anna Maria Rapone
- Medical Genetics, Department of Molecular Medicine; Sapienza University, S. Camillo-Forlanini Hospital; Rome Italy
| | - Nino Guarino
- Pediatric Surgery; S. Camillo-Forlanini Hospital; Rome Italy
| | | | - Carmelilia De Bernardo
- Medical Genetics, Department of Molecular Medicine; Sapienza University, S. Camillo-Forlanini Hospital; Rome Italy
| | | | - Silvia Majore
- Medical Genetics, Department of Molecular Medicine; Sapienza University, S. Camillo-Forlanini Hospital; Rome Italy
| | - Paola Grammatico
- Medical Genetics, Department of Molecular Medicine; Sapienza University, S. Camillo-Forlanini Hospital; Rome Italy
| |
Collapse
|
25
|
46,XY disorder of sex development and developmental delay associated with a novel 9q33.3 microdeletion encompassing NR5A1. Eur J Med Genet 2013; 56:619-23. [PMID: 24056159 DOI: 10.1016/j.ejmg.2013.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 09/10/2013] [Indexed: 11/23/2022]
Abstract
Steroidogenic factor 1 (SF1) is a nuclear receptor encoded by the NR5A1 gene. SF1 affects both sexual and adrenal development through the regulation of target gene expression. Genotypic male and female SF1 knockout mice have adrenal and gonadal agenesis with persistent Müllerian structures and early lethality. There have been several reports of NR5A1 mutations in individuals with 46,XY complete gonadal dysgenesis (CGD) or other disorders of sex development (DSD) with or without an adrenal phenotype. To date microdeletions involving NR5A1 have been reported in only two patients with DSDs. We report a novel microdeletion encompassing NR5A1 in a patient with 46,XY DSD and developmental delay. The phenotypically female patient initially presented with mild developmental delay and dysmorphisms. Chromosome analysis revealed a 46,XY karyotype. A 1.54 Mb microdeletion of chromosome 9q33.3 including NR5A1 was detected by array CGH and confirmed by FISH. Normal maternal FISH results indicated that this was most likely a de novo event. Since most NR5A1 mutations have been ascertained through gonadal or adrenal abnormalities, the additional findings of developmental delay and minor facial dysmorphisms are possibly related to haploinsufficiency of other genes within the 1.54 Mb deleted region. This report further confirms the role of NR5A1 deletions in 46,XY DSD and reinforces the utility of aCGH in the work up of DSDs of unclear etiology.
Collapse
|
26
|
Philibert P, Paris F, Lakhal B, Audran F, Gaspari L, Saâd A, Christin-Maître S, Bouchard P, Sultan C. NR5A1 (SF-1) gene variants in a group of 26 young women with XX primary ovarian insufficiency. Fertil Steril 2012; 99:484-9. [PMID: 23153500 DOI: 10.1016/j.fertnstert.2012.10.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 09/10/2012] [Accepted: 10/12/2012] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To determine whether NR5A1 (SF-1) variants are a cause of primary ovarian insufficiency (POI) in 26 young women with similar genetic background. DESIGN Genetic and functional mutation study. SETTING University hospitals. PATIENT(S) Genetic analysis of the NR5A1 gene in 26 XX girls with POI. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) NR5A1 molecular and functional analysis. RESULT(S) Genetic analysis revealed a new c.763C>T (p.Arg255Cys) mutation and a recurrent c.437G>C (p.Gly146Ala) variant. Functional analysis of the p.Arg255Cys mutant showed a marked decrease in transactivation on the Cyp11a1 and Amh promoters. The p.Gly146Ala variant was identified significantly more often in the patients (46.1%) than in ancestry-matched control subjects (10%). CONCLUSION(S) We identified one new NR5A1 mutation in a patient of our POI cohort (prevalence 3.8%). Moreover, although our study is limited in the number of cases, we report the high frequency of the p.Gly146Ala variant in this cohort compared with the ancestry-matched control subjects. This work highlights the important role of SF-1 in ovarian function.
Collapse
Affiliation(s)
- Pascal Philibert
- Département d'Hormonologie, Hôpital Lapeyronie, CHU de Montpellier and Université Montpellier 1, Montpellier, France
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Steroidogenic factor-1 (SF-1) (Ad4BP, NR5A1) is a nuclear receptor that plays a key role in adrenal and reproductive development and function. Deletion of the gene encoding Sf-1 (Nr5a1) in mice results in severe developmental defects of the adrenal gland and gonad. Consequently, initial work on the potential effects of SF-1 disruption in humans focused on individuals with primary adrenal failure, a 46,XY karyotype, complete gonadal dysgenesis, and Müllerian structures. This is a rare phenotype, but has been reported on two occasions, because of alterations that affect key DNA-binding domains of SF-1. Attention then turned to a potential wider role of SF-1 in human adrenal and reproductive disorders. Although changes in SF-1 only very rarely cause isolated adrenal failure, it is emerging that variations in SF-1 are a surprisingly frequent cause of reproductive dysfunction in humans. In 46,XY disorders of sex development, a spectrum of phenotypes has been reported including severe and partial forms of gonadal (testicular) dysgenesis, hypospadias, anorchia with microphallus, and even male factor infertility. In 46,XX females, alterations in SF-1 are associated with primary ovarian insufficiency. Thus, SF-1 seems be a more significant factor in human reproductive health than was first envisioned, with implications for adults as well as children.
Collapse
Affiliation(s)
- Ranna El-Khairi
- Developmental Endocrinology Research Group, Clinical and Molecular Genetics Unit, UCL Institute of Child Health, University College London, London, United Kingdom
| | - John C. Achermann
- Developmental Endocrinology Research Group, Clinical and Molecular Genetics Unit, UCL Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
28
|
Wisniewski AB. Gender Development in 46,XY DSD: Influences of Chromosomes, Hormones, and Interactions with Parents and Healthcare Professionals. SCIENTIFICA 2012; 2012:834967. [PMID: 24278745 PMCID: PMC3820494 DOI: 10.6064/2012/834967] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 09/18/2012] [Indexed: 06/02/2023]
Abstract
Variables that impact gender development in humans are difficult to evaluate. This difficulty exists because it is not usually possible to tease apart biological influences on gender from social variables. People with disorders of sex development, or DSD, provide important opportunities to study gender within individuals for whom biologic components of sex can be discordant with social components of gender. While most studies of gender development in people with 46,XY DSD have historically emphasized the importance of genes and hormones on gender identity and gender role, more recent evidence for a significant role for socialization exists and is considered here. For example, the influence of parents' perceptions of, and reactions to, DSD are considered. Additionally, the impact of treatments for DSD such as receiving gonadal surgeries or genitoplasty to reduce genital ambiguity on the psychological development of people with 46,XY DSD is presented. Finally, the role of multi-disciplinary care including access to peer support for advancing medical, surgical and psychosexual outcomes of children and adults with 46,XY DSD, regardless of sex of rearing, is discussed.
Collapse
Affiliation(s)
- Amy B Wisniewski
- Department of Urology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
29
|
Tantawy S, Lin L, Akkurt I, Borck G, Klingmüller D, Hauffa BP, Krude H, Biebermann H, Achermann JC, Köhler B. Testosterone production during puberty in two 46,XY patients with disorders of sex development and novel NR5A1 (SF-1) mutations. Eur J Endocrinol 2012; 167:125-30. [PMID: 22474171 PMCID: PMC3381348 DOI: 10.1530/eje-11-0944] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Steroidogenic factor 1 (SF-1, NR5A1) is a key transcriptional regulator of many genes involved in the hypothalamic-pituitary-gonadal axis and mutations in NR5A1 can result in 46,XY disorders of sex development (DSD). Patients with this condition typically present with ambiguous genitalia, partial gonadal dysgenesis, and absent/rudimentary Müllerian structures. In these cases, testosterone is usually low in early infancy, indicating significantly impaired androgen synthesis. Further, Sertoli cell dysfunction is seen (low inhibin B, anti-Müllerian hormone). However, gonadal function at puberty in patients with NR5A1 mutations is unknown. SUBJECTS AND METHODS Clinical assessment, endocrine evaluation, and genetic analysis were performed in one female and one male with 46,XY DSD who showed spontaneous virilization during puberty. The female patient presented at adolescence with clitoral hypertrophy, whereas the male patient presented at birth with severe hypospadias and entered puberty spontaneously. Molecular analysis of NR5A1 was performed followed by in vitro functional analysis of the two novel mutations detected. RESULTS Testosterone levels were normal during puberty in both patients. Analysis of NR5A1 revealed two novel heterozygous missense mutations in the ligand-binding domain of SF-1 (patient 1: p.L376F; patient 2: p.G328V). The mutant proteins showed reduced transactivation of the CYP11A promoter in vitro. CONCLUSION Patients with 46,XY DSD and NR5A1 mutations can produce sufficient testosterone for spontaneous virilization during puberty. Phenotypic females (46,XY) with NR5A1 mutations can present with clitoromegaly at puberty, a phenotype similar to other partial defects of androgen synthesis or action. Testosterone production in 46,XY males with NR5A1 mutations can be sufficient for virilization at puberty. As progressive gonadal dysgenesis is likely, gonadal function should be monitored in adolescence and adulthood, and early sperm cryopreservation considered in male patients if possible.
Collapse
Affiliation(s)
| | - Lin Lin
- UCL Institute of Child HealthUniversity College LondonLondonUK
| | | | - Guntram Borck
- Institute of Human GeneticsUniversity of UlmUlmGermany
| | | | - Berthold P Hauffa
- Department of Pediatric Endocrinology and DiabetesUniversity Children's Hospital, University Duisburg-EssenEssenGermany
| | | | | | | | | |
Collapse
|
30
|
Camats N, Pandey AV, Fernández-Cancio M, Andaluz P, Janner M, Torán N, Moreno F, Bereket A, Akcay T, García-García E, Muñoz MT, Gracia R, Nistal M, Castaño L, Mullis PE, Carrascosa A, Audí L, Flück CE. Ten novel mutations in the NR5A1 gene cause disordered sex development in 46,XY and ovarian insufficiency in 46,XX individuals. J Clin Endocrinol Metab 2012; 97:E1294-306. [PMID: 22549935 DOI: 10.1210/jc.2011-3169] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Steroidogenic factor-1 (SF-1/NR5A1) is a nuclear receptor that regulates adrenal and reproductive development and function. NR5A1 mutations have been detected in 46,XY individuals with disorders of sexual development (DSD) but apparently normal adrenal function and in 46,XX women with normal sexual development yet primary ovarian insufficiency (POI). OBJECTIVE A group of 100 46,XY DSD and two POI was studied for NR5A1 mutations and their impact. DESIGN Clinical, biochemical, histological, genetic, and functional characteristics of the patients with NR5A1 mutations are reported. SETTING Patients were referred from different centers in Spain, Switzerland, and Turkey. Histological and genetic studies were performed in Barcelona, Spain. In vitro studies were performed in Bern, Switzerland. PATIENTS A total of 65 Spanish and 35 Turkish patients with 46,XY DSD and two Swiss 46,XX patients with POI were investigated. MAIN OUTCOME Ten novel heterozygote NR5A1 mutations were detected and characterized (five missense, one nonsense, three frameshift mutations, and one duplication). RESULTS The novel NR5A1 mutations were tested in vitro by promoter transactivation assays showing grossly reduced activity for mutations in the DNA binding domain and variably reduced activity for other mutations. Dominant negative effect of the mutations was excluded. We found high variability and thus no apparent genotype-structure-function-phenotype correlation. Histological studies of testes revealed vacuolization of Leydig cells due to fat accumulation. CONCLUSIONS SF-1/NR5A1 mutations are frequently found in 46,XY DSD individuals (9%) and manifest with a broad phenotype. Testes histology is characteristic for fat accumulation and degeneration over time, similar to findings observed in patients with lipoid congenital adrenal hyperplasia (due to StAR mutations). Genotype-structure-function-phenotype correlation remains elusive.
Collapse
Affiliation(s)
- N Camats
- Pediatric Endocrinology Research Unit, Hospital Vall d’Hebron, Passeig Vall d’Hebron 119, Barcelona 08035, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gaspari L, Paris F, Philibert P, Audran F, Orsini M, Servant N, Maïmoun L, Kalfa N, Sultan C. 'Idiopathic' partial androgen insensitivity syndrome in 28 newborn and infant males: impact of prenatal exposure to environmental endocrine disruptor chemicals? Eur J Endocrinol 2011; 165:579-87. [PMID: 21788424 DOI: 10.1530/eje-11-0580] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE 46,XY disorders of sex differentiation (46,XY DSD) can be due to a testis determination defect, an androgen biosynthesis defect, or androgen resistance (complete or partial androgen insensitivity syndrome (PAIS), or 5α reductase deficiency). We aimed to evaluate the impact of a prenatal contamination by environmental xenoestrogens in 'idiopathic' PAIS-like phenotype. SUBJECTS We investigated 28 newborn/infant males with 46,XY DSD, normal androgen production, and no androgen receptor or steroid-5αR type II enzyme (SRD5A2) gene mutations. METHODS To exclude other genetic defects, we sequenced the steroidogenic factor 1 (SF1) and mastermind-like domain-containing 1 (MAMLD1) genes, which were recently found to be associated with the PAIS-like phenotype. Parents were interviewed about their environmental/occupational exposure to endocrine disrupting chemicals (EDCs) before/during the patients' fetal life. Total estrogenic bioactivity of patient serum was analyzed by ultrasensitive bioassay. RESULTS All the patients had normal SF1 sequence and one patient showed a double polymorphism of MAMLD1. Eleven (39.3%) of the 28 patients had reported parental fetal exposure to EDCs. The mean estrogenic bioactivity in these 11 patients with fetal EDC exposure (6.65 ± 8.07 pg/ml) versus 17 cases without contamination (1.27 ± 0.34 pg/ml) and controls (1.06 ± 0.44 pg/ml; P<0.05) was elevated. CONCLUSIONS Our results indicate that the 'idiopathic' PAIS-like phenotype may in some cases be related to EDC contamination during fetal life.
Collapse
Affiliation(s)
- Laura Gaspari
- Unité d'Endocrinologie-Gynécologie Pédiatrique, Service de Pédiatrie 1, Hôpital Arnaud-de-Villeneuve, Montpellier, France
| | | | | | | | | | | | | | | | | |
Collapse
|