1
|
Cools M, Grijp C, Neirinck J, Tavernier SJ, Schelstraete P, Van De Velde J, Morbée L, De Baere E, Bonroy C, van Bever Y, Bruggenwirth H, Vermont C, Hannema SE, De Rijke Y, Abdulhadi-Atwan M, Zangen D, Verdin H, Haerynck F. Spleen function is reduced in individuals with NR5A1 variants with or without a difference of sex development: a cross-sectional study. Eur J Endocrinol 2024; 190:34-43. [PMID: 38128121 DOI: 10.1093/ejendo/lvad174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE NR5A1 is a key regulator of sex differentiation and has been implicated in spleen development through transcription activation of TLX1. Concerns exist about hypo- or asplenism in individuals who have a difference of sex development (DSD) due to an NR5A1 disease-causing variant. We aimed to assess spleen anatomy and function in a clinical cohort of such individuals and in their asymptomatic family member carriers. DESIGN Cross-sectional assessment in 22 patients with a DSD or primary ovarian insufficiency and 5 asymptomatic carriers from 18 families, harboring 14 different NR5A1 variants. METHODS Spleen anatomy was assessed by ultrasound, spleen function by peripheral blood cell count, white blood cell differentiation, percentage of nonswitched memory B cells, specific pneumococcal antibody response, % pitted red blood cells, and Howell-Jolly bodies. RESULTS Patients and asymptomatic heterozygous individuals had significantly decreased nonswitched memory B cells compared to healthy controls, but higher than asplenic patients. Thrombocytosis and spleen hypoplasia were present in 50% of heterozygous individuals. Four out of 5 individuals homozygous for the previously described p.(Arg103Gln) variant had asplenia. CONCLUSIONS Individuals harboring a heterozygous NR5A1 variant that may cause DSD have a considerable risk for functional hyposplenism, irrespective of their gonadal phenotype. Splenic function should be assessed in these individuals, and if affected or unknown, prophylaxis is recommended to prevent invasive encapsulated bacterial infections. The splenic phenotype associated with NR5A1 variants is more severe in homozygous individuals and is, at least for the p.(Arg103Gln) variant, associated with asplenism.
Collapse
Affiliation(s)
- Martine Cools
- Department of Internal Medicine and Pediatrics, Pediatric Endocrinology Service, Ghent University, Ghent University Hospital, 9000 Ghent, Belgium
| | - Celien Grijp
- Department of Internal Medicine and Pediatrics, Pediatric Endocrinology Service, Ghent University, Ghent University Hospital, 9000 Ghent, Belgium
| | - Jana Neirinck
- Department of Diagnostic Science, Ghent University, Department of Laboratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Simon J Tavernier
- Department of Internal Medicine and Pediatrics, PID Research Lab, Ghent University, 9000 Ghent, Belgium
- Laboratory of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, VIB, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Petra Schelstraete
- Department of Internal Medicine and Pediatrics, Pediatric Pulmonology and Infectious Diseases, Ghent University, Ghent University Hospital, 9000 Ghent, Belgium
| | - Julie Van De Velde
- Department of Internal Medicine and Pediatrics, Pediatric Endocrinology Service, Ghent University, Ghent University Hospital, 9000 Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Lieve Morbée
- Department of Radiology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Elfride De Baere
- Center for Medical Genetics, Ghent University Hospital, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Carolien Bonroy
- Department of Diagnostic Science, Ghent University, Department of Laboratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Yolande van Bever
- Department of Clinical Genetics, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Hennie Bruggenwirth
- Department of Clinical Genetics, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Clementien Vermont
- Department of Pediatric Infectious Diseases and Immunology, Erasmus Medical Center-Sophia Children's Hospital, 3015 GD Rotterdam, The Netherlands
| | - Sabine E Hannema
- Department of Pediatric Endocrinology, Erasmus Medical Center-Sophia Children's Hospital, 3015 GD Rotterdam, The Netherlands
- Department of Paediatric Endocrinology, Gastroenterology Endocrinology Metabolism, Reproduction and Development, Amsterdam UMC location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yolanda De Rijke
- Department of Clinical Chemistry, Erasmus MC, University Medical Center 3015 GD Rotterdam, The Netherlands
| | - Maha Abdulhadi-Atwan
- Department of Pediatrics, Pediatric Endocrinology Service, Palestine Red Crescent Society Hospital, PO Box 421, Hebron, Palestine
| | - David Zangen
- Division of Pediatric Endocrinology, Faculty of Medicine, Hadassah University Hospital, Hebrew University of Jerusalem, 91120 Jerusalem, Israel
| | - Hannah Verdin
- Center for Medical Genetics, Ghent University Hospital, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Filomeen Haerynck
- Department of Internal Medicine and Pediatrics, PID Research Lab, Ghent University, 9000 Ghent, Belgium
- Department of Pediatric Pulmonology and Immunology, Centre for Primary Immune Deficiency, Jeffrey Modell Diagnostic and Research Centre for PID, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
2
|
Guerrero-Fernández J, González-Peramato P, Rodríguez Estévez A, Alcázar Villar MJ, Audí Parera L, Azcona San Julián MC, Carcavilla Urquí A, Castaño González LA, Martos Tello JM, Mora Palma C, Moreno Macián MF, Yeste Fernández D, Nistal M. Guía de consenso sobre la gonadectomía profiláctica en el desarrollo sexual diferente. ENDOCRINOL DIAB NUTR 2022. [DOI: 10.1016/j.endinu.2021.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
3
|
Guerrero-Fernández J, González-Peramato P, Rodríguez Estévez A, Alcázar Villar MJ, Audí Parera L, Azcona San Julián MC, Carcavilla Urquí A, Castaño González LA, Martos Tello JM, Mora Palma C, Moreno Macián MF, Yeste Fernández D, Nistal M. Consensus guide on prophylactic gonadectomy in different sex development. ENDOCRINOL DIAB NUTR 2022; 69:629-645. [PMID: 36369235 DOI: 10.1016/j.endien.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 11/06/2021] [Indexed: 06/16/2023]
Abstract
The risk of suffering from gonadal germ cell tumors (GCT) is increased in some patients with different sexual development (DSD), mainly in those with Y chromosome material. This risk, however, varies considerably depending on a multitude of factors that make the decision for prophylactic gonadectomy extremely difficult. In order to make informed recommendations on the convenience of this procedure in cases where there is potential for malignancy, this consensus guide evaluates the latest clinical evidence, which is generally low, and updates the existing knowledge in this field.
Collapse
Affiliation(s)
- Julio Guerrero-Fernández
- Grupo de Trabajo Sobre ADS/DSD de la Sociedad Española de Endocrinología Pediátrica (SEEP), Spain; Servicio de Endocrinología Pediátrica, Hospital Infantil La Paz, Madrid, Spain.
| | - Pilar González-Peramato
- Departamento de Anatomía Patológica, Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - María José Alcázar Villar
- Grupo de Trabajo Sobre ADS/DSD de la Sociedad Española de Endocrinología Pediátrica (SEEP), Spain; Unidad de Endocrinología Pediátrica, Hospital de Fuenlabrada, Fuenlabrada, Spain
| | - Laura Audí Parera
- Grupo de Trabajo Sobre ADS/DSD de la Sociedad Española de Endocrinología Pediátrica (SEEP), Spain; Vall d'Hebron Institut de Recerca (VHIR), CIBER de Enfermedades Raras (CIBERER), Hospital Vall d'Hebron, Barcelona, Spain
| | - María Cristina Azcona San Julián
- Grupo de Trabajo Sobre ADS/DSD de la Sociedad Española de Endocrinología Pediátrica (SEEP), Spain; Unidad de Endocrinología Pediátrica, Departamento de Pediatría, Clínica Universidad de Navarra, Pamplona, Spain
| | - Atilano Carcavilla Urquí
- Grupo de Trabajo Sobre ADS/DSD de la Sociedad Española de Endocrinología Pediátrica (SEEP), Spain; Servicio de Endocrinología Pediátrica, Hospital Infantil La Paz, Madrid, Spain
| | - Luis Antonio Castaño González
- Grupo de Trabajo Sobre ADS/DSD de la Sociedad Española de Endocrinología Pediátrica (SEEP), Spain; Instituto BioCruces - Endocrinología Pediátrica, Hospital Universitario Cruces, Barakaldo, Spain
| | - José María Martos Tello
- Grupo de Trabajo Sobre ADS/DSD de la Sociedad Española de Endocrinología Pediátrica (SEEP), Spain; Unidad de Endocrinología Pediátrica, Hospital Universitario Virgen de La Arrixaca, Murcia, Spain
| | - Cristina Mora Palma
- Grupo de Trabajo Sobre ADS/DSD de la Sociedad Española de Endocrinología Pediátrica (SEEP), Spain; Servicio de Endocrinología Pediátrica, Hospital Infantil La Paz, Madrid, Spain
| | - Maria Francisca Moreno Macián
- Grupo de Trabajo Sobre ADS/DSD de la Sociedad Española de Endocrinología Pediátrica (SEEP), Spain; Servicio de Endocrinología Pediátrica, Hospital La Fe, Valencia, Spain
| | - Diego Yeste Fernández
- Grupo de Trabajo Sobre ADS/DSD de la Sociedad Española de Endocrinología Pediátrica (SEEP), Spain; Servicio de Endocrinología Pediátrica, Hospital Materno Infantil Vall d'Hebron, CIBER de Enfermedades Raras (CIBERER), EndoERN, Barcelona, Spain
| | - Manuel Nistal
- Departamento de Anatomía, Histología y Neurociencias. Universidad Autónoma de Madrid, Madrid, Spain
| |
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
|
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
|
6
|
Kalinchenko NY, Kolodkina AA, Raygorodskaya NY, Tiulpakov AN. [Clinical and molecular characteristics of patients with 46,XY DSD due to NR5A1 gene mutations]. ACTA ACUST UNITED AC 2020; 66:62-69. [PMID: 33351340 DOI: 10.14341/probl12445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 01/18/2023]
Abstract
Steroidogenic factor 1 (SF1, NR5A1) is a nuclear receptor that regulates multiple genes involved in adrenal and gonadal development, steroidogenesis, and the reproductive axis. Human mutations in SF1 were initially found in patients with severe gonadal dysgenesis and primary adrenal failure. However, more recent case reports have suggested that heterozygous mutations in SF1 may also be found in patients with 46,XY partial gonadal dysgenesis and underandrogenization but normal adrenal function. We have analyzed the gene encoding SF1 (NR5A1) in a cohort of 310 Russian patients with 46,XY disorders of sex development (DSD). Heterozygous SF1 variants were found in 36 out of 310 (11.6%) of cases, among them 15 were not previously described. We have not found any phenotype-genotype correlations and any clinical and laboratory markers that would allow to suspect this type of before conducting molecular genetic analysis.
Collapse
|
7
|
Andrade JG, Fabbri-Scallet H, dos Santos AP, Cools M, Werner R, Hiort O, de Mello MP, Guerra-Júnior G, Maciel-Guerra AT. Clinical Findings and Follow-Up of 46,XY and 45,X/46,XY Testicular Dysgenesis. Sex Dev 2019; 13:171-177. [DOI: 10.1159/000504239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2019] [Indexed: 12/12/2022] Open
|
8
|
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
|
9
|
Faure Conter C, Brindusa Gorduza D, Mure PY, Pracros JP, Mouriquand P, Bouvattier C, Siffroi JP, Plotton I, Gay CL, Cools M, Dijoud F. [Atypical genital development and tumor risk]. Bull Cancer 2019; 106:461-467. [PMID: 30910228 DOI: 10.1016/j.bulcan.2019.01.018] [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: 11/06/2018] [Revised: 01/14/2019] [Accepted: 01/23/2019] [Indexed: 11/16/2022]
Abstract
Atypical genital development (AGD), also called disorders of sex development are a set of miscellaneous pathologies who have in common a morphological and/or functional abnormality of the internal and/or external genital organs. The Chicago classification identifies 3 major groups based on karyotype, hormone balance and genetic studies. Some AGD predispose to the occurrence of tumors, mainly malignant germ cell tumors. The tumor risk depends on many factors: the type of AGD, the position of the gonad, the age of the patient, the phenotype, the function of the gonad and the presence of germ cells in the gonad. AGD with the highest tumor risk are those with gonadal dysgenesis, implying an incomplete differentiation of the bipotential gonad (dysplasia). Monitoring of patients with AGD and indication of prophylactic gonadectomies should be individualized according to tumor risk.
Collapse
Affiliation(s)
- Cécile Faure Conter
- Institut d'hématologie et d'oncologie pédiatrique, IHOPe, 69008 Lyon, France.
| | - Daniela Brindusa Gorduza
- Groupement hospitalier Est, Hospices Civils de Lyon, département de chirurgie pédiatrique, 69500 Bron, France
| | - Pierre-Yves Mure
- Groupement hospitalier Est, Hospices Civils de Lyon, département de chirurgie pédiatrique, 69500 Bron, France
| | - Jean-Pierre Pracros
- Groupement hospitalier Est, Hospices Civils de Lyon, département de radiologie pédiatrique, 69500 Bron, France
| | - Pierre Mouriquand
- Groupement hospitalier Est, Hospices Civils de Lyon, département de chirurgie pédiatrique, 69500 Bron, France
| | - Claire Bouvattier
- Hôpital Bicêtre, département d'endocrinologie pédiatrique, 94270 Le Kremlin Bicetre, France
| | - Jean-Pierre Siffroi
- Hôpital d'Enfants Armand Trousseau, département de génétique médicale, 75012 Paris, France
| | - Ingrid Plotton
- Groupement hospitalier Est, Hospices Civils de Lyon, Centre de biologie moléculaire, 69500 Bron, France
| | - Claire-Lise Gay
- Groupement hospitalier Est, département d'endocrinologie pédiatrique, 69500 Bron, France
| | - Martine Cools
- Hôpital universitaire de Gand, département de médecine interne et de pédiatrie université de Gand, 9000 Gand, Belgique
| | - Frédérique Dijoud
- Groupement hospitalier Est, hospices Civils de Lyon, institut Multisite de pathologie, 69500 Bron, France
| |
Collapse
|
10
|
Adachi M, Hasegawa T, Tanaka Y, Asakura Y, Hanakawa J, Muroya K. Spontaneous virilization around puberty in NR5A1-related 46,XY sex reversal: additional case and a literature review. Endocr J 2018; 65:1187-1192. [PMID: 30224582 DOI: 10.1507/endocrj.ej18-0218] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A heterozygous NR5A1 mutation is one of the most frequent causes of 46,XY DSD (disorders of sex development). We here reported a NR5A1-related 46,XY DSD patient, who first received endocrinological attention at 10 years of age for clitoromegaly. The patient had been reared as a girl, and no signs of virilization had been detected before. On examination, her clitoris was 35 mm long and 10 mm wide, with Tanner 3° pubic hair. Urogenital sinus and labial fusion was absent, while her uterus was found to be severely hypoplastic. Her basal testosterone level was 94.8 ng/dL, suggesting the presence of functioning Leydig cells. Gonadal histology revealed bilateral dysplastic testes consisting of mostly Sertoli cell-only tubules and Leydig cell hyperplasia. Novel heterozygous Arg313Leu substitution in NR5A1 was identified in the patient. Literature search confirmed twelve other cases of this scenario, namely, severe under-virilization in utero followed by spontaneous virilization around puberty in NR5A1-related 46,XY DSD. Of interest, Leydig cell hyperplasia was documented in 6 out of 9 patients for whom testicular histology was available. To keep in mind about the possible restoration of Leydig cell function around puberty, even in patients without discernible in utero androgen effect, may be of clinical significance, because it will give a great impact on the judgement about sex assignment.
Collapse
Affiliation(s)
- Masanori Adachi
- Department of Endocrinology and Metabolism, Kanagawa Children's Medical Center, Yokohama 232-8555, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yukichi Tanaka
- Division of Diagnostic Pathology, Kanagawa Children's Medical Center, Yokohama 232-8555, Japan
| | - Yumi Asakura
- Department of Endocrinology and Metabolism, Kanagawa Children's Medical Center, Yokohama 232-8555, Japan
| | - Junko Hanakawa
- Department of Endocrinology and Metabolism, Kanagawa Children's Medical Center, Yokohama 232-8555, Japan
| | - Koji Muroya
- Department of Endocrinology and Metabolism, Kanagawa Children's Medical Center, Yokohama 232-8555, Japan
| |
Collapse
|
11
|
Orekhova AS, Kalinchenko N, Morozov IA, Vasilyev EV, Rubtsov PM, Dedov II, Tiulpakov A. A Novel Mutation in the Critical P-Box Residue of Steroidogenic Factor-1 Presenting with XY Sex Reversal and Transient Adrenal Failure. Horm Res Paediatr 2018; 89:450-454. [PMID: 29151085 DOI: 10.1159/000481776] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 09/22/2017] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Although the importance of steroidogenic factor-1 (SF1, NR5A1) for adrenal development is supported by numerous in vitro and in vivo studies, cases of SF1 deficiency associated with adrenal failure are exceptionally rare. The first human NR5A1 mutation was a heterozygous de novo p.G35E variant identified in a patient with disorder of sex development (DSD) 46,XY and primary adrenal insufficiency. Here we describe another association of the "classic" SF1 phenotype with a novel NR5A1 mutation affecting G35 residue. METHODS We describe the clinical characteristics of a phenotypically female patient presenting at 2 months with signs of adrenal insufficiency. DSD 46,XY was diagnosed at 4 years. The NR5A1 gene was analyzed by Sanger sequencing. Minigene splicing and dual luciferase reporter assays were used to characterize effects of the novel mutation on splicing and transcription, respectively. RESULTS Sequencing of the NR5A1 gene revealed a de novo heterozygous c.104G>A:p.G35D substitution. The minigene experiments demonstrated that c.104G>A substitution did not affect splicing. However, transactivation activity of the p.G35D mutant was clearly impaired, which was comparable with the effect of the p.G35E mutation. CONCLUSIONS The findings stress the importance of G35 residue for adrenal development. The current observation also suggests that some patients with SF1 deficiency may present with transient adrenal failure.
Collapse
Affiliation(s)
- Anna S Orekhova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation
| | | | - Ivan A Morozov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation
| | | | - Petr M Rubtsov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation
| | - Ivan I Dedov
- Endocrinology Research Centre, Moscow, Russian Federation
| | | |
Collapse
|
12
|
Ożegowska K, Dyszkiewicz-Konwińska M, Celichowski P, Nawrocki MJ, Bryja A, Jankowski M, Kranc W, Brązert M, Knap S, Jeseta M, Skowroński MT, Bukowska D, Antosik P, Brüssow KP, Bręborowicz A, Bruska M, Nowicki M, Pawelczyk L, Zabel M, Kempisty B. Expression pattern of new genes regulating female sex differentiation and in vitro maturational status of oocytes in pigs. Theriogenology 2018; 121:122-133. [PMID: 30145542 DOI: 10.1016/j.theriogenology.2018.08.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023]
Abstract
The processes underlying maturation of mammalian oocytes are considered crucial for the oocytes ability to undergo monospermic fertilization. The same factors of influence are suggested to impact the development of sex associated characteristics, allowing sex differentiation to progress during embryonic growth. The primary aim of the study was to analyze the gene ontology groups involved in regulation of porcine oocytes' response to endogenous stimuli. The results obtained would indicate potential genes influencing sex differentiation. Additionally, they could help to determine new genetic markers, expression profile of which is substantially regulated during porcine oocytes' in vitro maturation. To achieve that, porcine oocytes were collected for analysis before and after in vitro maturation. Pigs were used as they are a readily available model that presents significant similarity to humans in terms of physiology and anatomy. Microarray analysis of oocytes, before and after in vitro maturation was performed and later validated by RT-qPCR. We have particularly detected and analyzed genes belonging to gene ontology groups associated with hormonal stimulation during maturation of the oocytes, that exhibited significant change in expression (fold change ≥ |2|; p < 0.05) namely "Female sex differentiation" (CCND2, MMP14, VEGFA, FST, INHBA, NR5A1), "Response to endogenous stimulus" (INSR, ESR1, CCND2, TXNIP, TACR3, MMP14, FOS, AR, EGR2, IGFBP7, TGFBR3, BTG2, PLD1, PHIP, UBE2B) and "Response to estrogen stimulus" (INSR, ESR1, CCND2, IHH, TXNIP, TACR3, MMP14). Some of them were characteristic for just one of the described ontologies, while some belonged into multiple ontological terms. The genes were analyzed, with their relation to the processes of interest explained. Overall, the study provides us with a range of genes that might serve as molecular markers of in vitro maturation associated processes of the oocytes. This knowledge might serve as a reference for further studies and, after further validation, as a potentially useful knowledge in assessment of the oocytes during assisted reproduction processes.
Collapse
Affiliation(s)
- Katarzyna Ożegowska
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marta Dyszkiewicz-Konwińska
- Department of Biomaterials and Experimental Dentistry, Poznań University of Medical Sciences, Poznań, Poland; Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland
| | - Piotr Celichowski
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Mariusz J Nawrocki
- Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland
| | - Artur Bryja
- Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland
| | - Maurycy Jankowski
- Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland
| | - Wiesława Kranc
- Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Brązert
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Sandra Knap
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland; Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland
| | - Michal Jeseta
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Czech Republic
| | - Mariusz T Skowroński
- Department of Animal Physiology University of Warmia and Mazury, Olsztyn, Poland
| | - Dorota Bukowska
- Veterinary Center, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Paweł Antosik
- Veterinary Center, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Klaus P Brüssow
- Veterinary Center, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Andrzej Bręborowicz
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznan, Poland
| | - Małgorzata Bruska
- Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Leszek Pawelczyk
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Zabel
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland; Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland
| | - Bartosz Kempisty
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland; Department of Anatomy, Poznan University of Medical Sciences, Poznan, Poland; Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Czech Republic.
| |
Collapse
|
13
|
Gomes NL, Lerário AM, Machado AZ, Moraes DRD, Silva TED, Arnhold IJP, Batista RL, Faria Júnior JAD, Costa EF, Nishi MY, Inacio M, Domenice S, Mendonca BB. Long-term outcomes and molecular analysis of a large cohort of patients with 46,XY disorder of sex development due to partial gonadal dysgenesis. Clin Endocrinol (Oxf) 2018; 89:164-177. [PMID: 29668062 DOI: 10.1111/cen.13717] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 04/06/2018] [Accepted: 04/06/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND Follow-up data on patients with 46,XY partial gonadal dysgenesis (PGD) until adulthood are scarce, making information on prognosis difficult. OBJECTIVE To analyse the long-term outcomes of patients with 46,XY PGD regarding testosterone production, germ cell tumour risk, genotype and psychosexual adaptation. METHODS A retrospective longitudinal study of 33 patients (20 assigned male and 13 patients assigned female at birth). Molecular diagnosis was performed by Sanger sequencing or by targeted massively parallel sequencing of 63 genes related to disorders of sex development (DSDs). RESULTS Age at first and last visit ranged from 0.1 to 43 and from 17 to 53 years, respectively. Spontaneous puberty was observed in 57% of the patients. During follow-up, six of them had a gonadectomy (four due to female gender, and two because of a gonadal tumour). At last evaluation, five of six patients had adult male testosterone levels (median 16.7 nmol/L, range 15.3-21.7 nmol/L) and elevated LH and FSH levels. Germ cell tumours were found in two postpubertal patients (one with an abdominal gonad and one patient with Frasier syndrome). Molecular diagnosis was possible in 11 patients (33%). NR5A1 variants were the most prevalent molecular defects (n = 6), and four of five patients harbouring them developed spontaneous puberty. Gender change was observed in four patients, two from each sex assignment group; all patients reported satisfaction with their gender at final evaluation. Sexual intercourse was reported by 81% of both gender and 82% of them reported satisfaction with their sexual lives. CONCLUSION Spontaneous puberty was observed in 57% of the patients with 46,XY PGD, being NR5A1 defects the most prevalent ones among all the patients and in those with spontaneous puberty. Gender change due to gender dysphoria was reported by 12% of the patients. All the patients reported satisfaction with their final gender, and most of them with their sexual life.
Collapse
Affiliation(s)
- Nathalia L Gomes
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Antônio Marcondes Lerário
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Aline Zamboni Machado
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Daniela Rodrigues de Moraes
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Thatiana Evilen da Silva
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ivo J P Arnhold
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Rafael Loch Batista
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - José Antônio Diniz Faria Júnior
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Elaine F Costa
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Mirian Y Nishi
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marlene Inacio
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sorahia Domenice
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Berenice B Mendonca
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
14
|
Berberoğlu M, Şıklar Z, of Sexual Development Ethics Committee AUFOMD. The Evaluation of Cases with Y-Chromosome Gonadal Dysgenesis: Clinical Experience over 18 Years. J Clin Res Pediatr Endocrinol 2018; 10:30-37. [PMID: 28825592 PMCID: PMC5838370 DOI: 10.4274/jcrpe.4826] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVE Y-chromosome gonadal dysgenesis (GD) is a rare subgroup of disorders of sexual development (DSD) which results from underdeveloped testis and may exhibit heterogenous symptoms. These patients are phenotypically classified into two groups - complete and partial, and their karyotypic description is either 46,XY GD or 45,X/46,XY GD. In this study; we aimed to evaluate the characteristics of cases with Y-chromosome GD. METHODS Thirty eight cases were followed-up between 1998 and 2016. The age of admission ranged between 0 and 17 years. Clinical and laboratory findings as well as follow-up characteristics of the cases were evaluated retrospectively from the patient files. RESULTS There were 26 cases (four complete, 22 partial) in the 46,XY GD group, and 12 cases (four complete, 8 patients with complete GD in the 45,X/46,XY. Mean age at admission was 6.2±4.6 years for all cases. Patients with complete GD in the 45,X/46,XY GD group were diagnosed earlier that the patients with complete GD in the 46,XY group [11 years vs. 14.31 years of age (p<0.01)]. There were no additional findings in 55% of all patients. In the remaining 45% additional clinical findings, mainly short stature, were detected in 75% of the patients in the 45,X/46,XY GD and 30% of the patients in the 46,XY GD groups. All patients with complete 46,XY and 45,X/46,XY GD were raised as females. There was no gender dysphoria in patients that were raised as females, except for one case. Gonadectomy was performed in 14 patients, at a mean age of 8.75±2.3 years and pathological assessment of the gonads was reported as normal in all cases. CONCLUSION Y-chromosome GD is a very heterogenous clinical and genetic disorder and requires a multifaceted approach to management. Whether including syndromic features or not, associated clinical features may lead to earlier diagnosis, especially in complete forms of GD. Due to difficulties encountered in the long-term follow-up of these patients, evaluation of appropriateness of sex of rearing decision is not truly possible. Performance of gonadectomy during the first decade appears be a preventive factor for tumor development since these tumors are usually seen during the second decade.
Collapse
Affiliation(s)
- Merih Berberoğlu
- Ankara University Faculty of Medicine, Department of Pediatric Endocrinology, Ankara, Turkey
| | - Zeynep Şıklar
- Ankara University Faculty of Medicine, Department of Pediatric Endocrinology, Ankara, Turkey,* Address for Correspondence: Ankara University Faculty of Medicine, Department of Pediatric Endocrinology, Ankara, Turkey Phone: +90 312 595 66 35 E-mail:
| | | |
Collapse
|
15
|
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
|
16
|
Domenice S, Machado AZ, Ferreira FM, Ferraz-de-Souza B, Lerario AM, Lin L, Nishi MY, Gomes NL, da Silva TE, Silva RB, Correa RV, Montenegro LR, Narciso A, Costa EMF, Achermann JC, Mendonca BB. Wide spectrum of NR5A1-related phenotypes in 46,XY and 46,XX individuals. BIRTH DEFECTS RESEARCH. PART C, EMBRYO TODAY : REVIEWS 2017; 108:309-320. [PMID: 28033660 PMCID: PMC5347970 DOI: 10.1002/bdrc.21145] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Steroidogenic factor 1 (NR5A1, SF‐1, Ad4BP) is a transcriptional regulator of genes involved in adrenal and gonadal development and function. Mutations in NR5A1 have been among the most frequently identified genetic causes of gonadal development disorders and are associated with a wide phenotypic spectrum. In 46,XY individuals, NR5A1‐related phenotypes may range from disorders of sex development (DSD) to oligo/azoospermia, and in 46,XX individuals, from 46,XX ovotesticular and testicular DSD to primary ovarian insufficiency (POI). The most common 46,XY phenotype is atypical or female external genitalia with clitoromegaly, palpable gonads, and absence of Müllerian derivatives. Notably, an undervirilized external genitalia is frequently seen at birth, while spontaneous virilization may occur later, at puberty. In 46,XX individuals, NR5A1 mutations are a rare genetic cause of POI, manifesting as primary or secondary amenorrhea, infertility, hypoestrogenism, and elevated gonadotropin levels. Mothers and sisters of 46,XY DSD patients carrying heterozygous NR5A1 mutations may develop POI, and therefore require appropriate counseling. Moreover, the recurrent heterozygous p.Arg92Trp NR5A1 mutation is associated with variable degrees of testis development in 46,XX patients. A clear genotype‐phenotype correlation is not seen in patients bearing NR5A1 mutations, suggesting that genetic modifiers, such as pathogenic variants in other testis/ovarian‐determining genes, may contribute to the phenotypic expression. Here, we review the published literature on NR5A1‐related disease, and discuss our findings at a single tertiary center in Brazil, including ten novel NR5A1 mutations identified in 46,XY DSD patients. The ever‐expanding phenotypic range associated with NR5A1 variants in XY and XX individuals confirms its pivotal role in reproductive biology, and should alert clinicians to the possibility of NR5A1 defects in a variety of phenotypes presenting with gonadal dysfunction. Birth Defects Research (Part C) 108:309–320, 2016. © 2016 The Authors Birth Defects Research Part C: Embryo Today: Reviews Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sorahia Domenice
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Aline Zamboni Machado
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Frederico Moraes Ferreira
- Frederico Moraes Ferreira is from the Ciências da Saúde, Universidade Santo Amaro, São Paulo, Brasil and Laboratorio de Imunologia, Instituto do Coração, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Bruno Ferraz-de-Souza
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Antonio Marcondes Lerario
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Lin Lin
- Lin Lin and John C. Achermann are form the Genetics & Genomic Medicine, University College London (UCL) Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Mirian Yumie Nishi
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Nathalia Lisboa Gomes
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Thatiana Evelin da Silva
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Rosana Barbosa Silva
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Rafaela Vieira Correa
- Rafaela V. Correa is from the Núcleo de Atenção Médica Integrada (NAMI), Universidade de Fortaleza, Ceará, Brasil
| | - Luciana Ribeiro Montenegro
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Amanda Narciso
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Elaine Maria Frade Costa
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - John C Achermann
- Lin Lin and John C. Achermann are form the Genetics & Genomic Medicine, University College London (UCL) Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Berenice Bilharinho Mendonca
- Sorahia Domenice, Aline Zamboni Machado, Bruno Ferraz-de-Souza, Antonio Marcondes Lerario, Mirian Yumie Nishi, Nathalia Lisboa Gomes, Thatiana Evelin da Silva, Rosana Barbosa Silva, Luciana R. Montenegro, Amanda Narciso, Elaine Maria Frade Costa, and Berenice Bilharinho Mendonca are from the Laboratório de Hormônios e Genética Molecular (LIM/42), Unidade de Endocrinologia do Desenvolvimento, Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| |
Collapse
|
17
|
Chen MJ, McCann-Crosby B, Gunn S, Georgiadis P, Placencia F, Mann D, Axelrad M, Karaviti L, McCullough LB. Fluidity models in ancient Greece and current practices of sex assignment. Semin Perinatol 2017; 41:206-213. [PMID: 28478088 PMCID: PMC5950726 DOI: 10.1053/j.semperi.2017.03.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Disorders of sexual differentiation such as androgen insensitivity and gonadal dysgenesis can involve an intrinsic fluidity at different levels, from the anatomical and biological to the social (gender) that must be considered in the context of social constraints. Sex assignment models based on George Engel's biopsychosocial aspects model of biology accept fluidity of gender as a central concept and therefore help establish expectations within the uncertainty of sex assignment and anticipate potential changes. The biology underlying the fluidity inherent to these disorders should be presented to parents at diagnosis, an approach that the gender medicine field should embrace as good practice. Greek mythology provides many accepted archetypes of change, and the ancient Greek appreciation of metamorphosis can be used as context with these patients. Our goal is to inform expertise and optimal approaches, knowing that this fluidity may eventually necessitate sex reassignment. Physicians should provide sex assignment education based on different components of sexual differentiation, prepare parents for future hormone-triggered changes in their children, and establish a sex-assignment algorithm.
Collapse
Affiliation(s)
- Min-Jye Chen
- Divison of Pediatric Endocrinology, Baylor College of Medicine, Texas Children’s Hospital, 6701 Fannin Street, Suite 1020, Houston, TX 77030
| | - Bonnie McCann-Crosby
- Divison of Pediatric Endocrinology, Baylor College of Medicine, Texas Children's Hospital, 6701 Fannin Street, Suite 1020, Houston, TX 77030.
| | - Sheila Gunn
- Divison of Pediatric Endocrinology, Baylor College of Medicine, Texas Children’s Hospital, 6701 Fannin Street, Suite 1020, Houston, TX 77030
| | - Paraskevi Georgiadis
- Section of Neonatology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX
| | - Frank Placencia
- Section of Neonatology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX
| | - David Mann
- Department of Anesthesiology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX
| | - Marni Axelrad
- Section of Psychology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX
| | - L.P Karaviti
- Divison of Pediatric Endocrinology, Baylor College of Medicine, Texas Children’s Hospital, 6701 Fannin Street, Suite 1020, Houston, TX 77030
| | | |
Collapse
|
18
|
Baetens D, Stoop H, Peelman F, Todeschini AL, Rosseel T, Coppieters F, Veitia RA, Looijenga LHJ, De Baere E, Cools M. NR5A1 is a novel disease gene for 46,XX testicular and ovotesticular disorders of sex development. Genet Med 2016; 19:367-376. [PMID: 27490115 PMCID: PMC5392598 DOI: 10.1038/gim.2016.118] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/11/2016] [Indexed: 01/10/2023] Open
Abstract
PURPOSE We aimed to identify the genetic cause in a cohort of 11 unrelated cases and two sisters with 46,XX SRY-negative (ovo)testicular disorders of sex development (DSD). METHODS Whole-exome sequencing (n = 9), targeted resequencing (n = 4), and haplotyping were performed. Immunohistochemistry of sex-specific markers was performed on patients' gonads. The consequences of mutation were investigated using luciferase assays, localization studies, and RNA-seq. RESULTS We identified a novel heterozygous NR5A1 mutation, c.274C>T p.(Arg92Trp), in three unrelated patients. The Arg92 residue is highly conserved and located in the Ftz-F1 region, probably involved in DNA-binding specificity and stability. There were no consistent changes in transcriptional activation or subcellular localization. Transcriptomics in patient-derived lymphocytes showed upregulation of MAMLD1, a direct NR5A1 target previously associated with 46,XY DSD. In gonads of affected individuals, ovarian FOXL2 and testicular SRY-independent SOX9 expression observed. CONCLUSIONS We propose NR5A1, previously associated with 46,XY DSD and 46,XX primary ovarian insufficiency, as a novel gene for 46,XX (ovo)testicular DSD. We hypothesize that p.(Arg92Trp) results in decreased inhibition of the male developmental pathway through downregulation of female antitestis genes, thereby tipping the balance toward testicular differentiation in 46,XX individuals. In conclusion, our study supports a role for NR5A1 in testis differentiation in the XX gonad.Genet Med 19 4, 367-376.
Collapse
Affiliation(s)
- Dorien Baetens
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Hans Stoop
- Department of Pathology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Frank Peelman
- Flanders Institute for Biotechnology (VIB), Department of Medical Protein Research, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Anne-Laure Todeschini
- Molecular Oncology and Pathology, Institut Jacques Monod, France; Université Paris Diderot, Paris VII, France
| | - Toon Rosseel
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Frauke Coppieters
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Reiner A Veitia
- Molecular Oncology and Pathology, Institut Jacques Monod, France; Université Paris Diderot, Paris VII, France
| | - Leendert H J Looijenga
- Department of Pathology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Elfride De Baere
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Martine Cools
- Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University Hospital and Ghent University, Ghent, Belgium
| |
Collapse
|
19
|
Fabbri HC, Ribeiro de Andrade JG, Maciel-Guerra AT, Guerra-Júnior G, de Mello MP. NR5A1 Loss-of-Function Mutations Lead to 46,XY Partial Gonadal Dysgenesis Phenotype: Report of Three Novel Mutations. Sex Dev 2016; 10:191-199. [DOI: 10.1159/000448013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Indexed: 11/19/2022] Open
|
20
|
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
|
21
|
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
|
22
|
Tuhan H, Abaci A, Aykut A, Anik A, Onay H, Bober E. A Novel Androgen Receptor Gene Mutation in a 46,XY Patient: Complete Androgen Insensitivity Syndrome. AACE Clin Case Rep 2016. [DOI: 10.4158/ep15734.cr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
23
|
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
|
24
|
Burckhardt MA, Udhane SS, Marti N, Schnyder I, Tapia C, Nielsen JE, Mullis PE, Rajpert-De Meyts E, Flück CE. Human 3β-hydroxysteroid dehydrogenase deficiency seems to affect fertility but may not harbor a tumor risk: lesson from an experiment of nature. Eur J Endocrinol 2015; 173:K1-K12. [PMID: 26290012 DOI: 10.1530/eje-15-0599] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/19/2015] [Indexed: 11/08/2022]
Abstract
CONTEXT 3β-hydroxysteroid dehydrogenase deficiency (3βHSD) is a rare disorder of sexual development and steroidogenesis. There are two isozymes of 3βHSD, HSD3B1 and HSD3B2. Human mutations are known for the HSD3B2 gene which is expressed in the gonads and the adrenals. Little is known about testis histology, fertility and malignancy risk. OBJECTIVE To describe the molecular genetics, the steroid biochemistry, the (immuno-)histochemistry and the clinical implications of a loss-of-function HSD3B2 mutation. METHODS Biochemical, genetic and immunohistochemical investigations on human biomaterials. RESULTS A 46,XY boy presented at birth with severe undervirilization of the external genitalia. Steroid profiling showed low steroid production for mineralocorticoids, glucocorticoids and sex steroids with typical precursor metabolites for HSD3B2 deficiency. The genetic analysis of the HSD3B2 gene revealed a homozygous c.687del27 deletion. At pubertal age, he showed some virilization of the external genitalia and some sex steroid metabolites appeared likely through conversion of precursors secreted by the testis and converted by unaffected HSD3B1 in peripheral tissues. However, he also developed enlarged breasts through production of estrogens in the periphery. Testis histology in late puberty revealed primarily a Sertoli-cell-only pattern and only few tubules with arrested spermatogenesis, presence of few Leydig cells in stroma, but no neoplastic changes. CONCLUSIONS The testis with HSD3B2 deficiency due to the c.687del27 deletion does not express the defective protein. This patient is unlikely to be fertile and his risk for gonadal malignancy is low. Further studies are needed to obtain firm knowledge on malignancy risk for gonads harboring defects of androgen biosynthesis.
Collapse
Affiliation(s)
- Marie-Anne Burckhardt
- Pediatric Endocrinology and DiabetologyDepartments of Pediatrics and Clinical ResearchPediatric SurgeryInstitute of PathologyUniversity of Bern, CH-3010 Bern, SwitzerlandDepartment of Growth and ReproductionRigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sameer S Udhane
- Pediatric Endocrinology and DiabetologyDepartments of Pediatrics and Clinical ResearchPediatric SurgeryInstitute of PathologyUniversity of Bern, CH-3010 Bern, SwitzerlandDepartment of Growth and ReproductionRigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Nesa Marti
- Pediatric Endocrinology and DiabetologyDepartments of Pediatrics and Clinical ResearchPediatric SurgeryInstitute of PathologyUniversity of Bern, CH-3010 Bern, SwitzerlandDepartment of Growth and ReproductionRigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Isabelle Schnyder
- Pediatric Endocrinology and DiabetologyDepartments of Pediatrics and Clinical ResearchPediatric SurgeryInstitute of PathologyUniversity of Bern, CH-3010 Bern, SwitzerlandDepartment of Growth and ReproductionRigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Coya Tapia
- Pediatric Endocrinology and DiabetologyDepartments of Pediatrics and Clinical ResearchPediatric SurgeryInstitute of PathologyUniversity of Bern, CH-3010 Bern, SwitzerlandDepartment of Growth and ReproductionRigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - John E Nielsen
- Pediatric Endocrinology and DiabetologyDepartments of Pediatrics and Clinical ResearchPediatric SurgeryInstitute of PathologyUniversity of Bern, CH-3010 Bern, SwitzerlandDepartment of Growth and ReproductionRigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Primus E Mullis
- Pediatric Endocrinology and DiabetologyDepartments of Pediatrics and Clinical ResearchPediatric SurgeryInstitute of PathologyUniversity of Bern, CH-3010 Bern, SwitzerlandDepartment of Growth and ReproductionRigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- Pediatric Endocrinology and DiabetologyDepartments of Pediatrics and Clinical ResearchPediatric SurgeryInstitute of PathologyUniversity of Bern, CH-3010 Bern, SwitzerlandDepartment of Growth and ReproductionRigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christa E Flück
- Pediatric Endocrinology and DiabetologyDepartments of Pediatrics and Clinical ResearchPediatric SurgeryInstitute of PathologyUniversity of Bern, CH-3010 Bern, SwitzerlandDepartment of Growth and ReproductionRigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
25
|
Tremblay JJ. Molecular regulation of steroidogenesis in endocrine Leydig cells. Steroids 2015; 103:3-10. [PMID: 26254606 DOI: 10.1016/j.steroids.2015.08.001] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 07/19/2015] [Accepted: 08/04/2015] [Indexed: 02/06/2023]
Abstract
Steroid hormones regulate essential physiological processes and inadequate levels are associated with various pathological conditions. Consequently, the process of steroid hormone biosynthesis is finely regulated. In the testis, the main steroidogenic cells are the Leydig cells. There are two distinct populations of Leydig cells that arise during development: fetal and adult Leydig cells. Fetal Leydig cells are responsible for masculinizing the male urogenital tract and inducing testis descent. These cells atrophy shortly after birth and do not contribute to the adult Leydig cell population. Adult Leydig cells derive from undifferentiated precursors present after birth and become fully steroidogenic at puberty. The differentiation of both Leydig cell populations is controlled by locally produced paracrine factors and by endocrine hormones. In fully differentially and steroidogenically active Leydig cells, androgen production and hormone-responsiveness involve various signaling pathways and downstream transcription factors. This review article focuses on recent developments regarding the origin and function of Leydig cells, the regulation of their differentiation by signaling molecules, hormones, and structural changes, the signaling pathways, kinases, and transcription factors involved in their differentiation and in mediating LH-responsiveness, as well as the fine-tuning mechanisms that ensure adequate production steroid hormones.
Collapse
Affiliation(s)
- Jacques J Tremblay
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Québec City, Québec G1V 4G2, Canada; Centre for Research in Biology of Reproduction, Department of Obstetrics, Gynaecology, and Reproduction, Faculty of Medicine, Université Laval, Québec City, Québec G1V 0A6, Canada.
| |
Collapse
|
26
|
Karpova T, Ravichandiran K, Insisienmay L, Rice D, Agbor V, Heckert LL. Steroidogenic factor 1 differentially regulates fetal and adult leydig cell development in male mice. Biol Reprod 2015; 93:83. [PMID: 26269506 DOI: 10.1095/biolreprod.115.131193] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/05/2015] [Indexed: 12/17/2022] Open
Abstract
The nuclear receptor steroidogenic factor 1 (SF-1, AD4BP, NR5A1) is a key regulator of the endocrine axes and is essential for adrenal and gonad development. Partial rescue of Nr5a1(-/-) mice with an SF-1-expressing transgene caused a hypomorphic phenotype that revealed its roles in Leydig cell development. In contrast to controls, all male rescue mice (Nr5a1(-/-);tg(+/0)) showed varying signs of androgen deficiency, including spermatogenic arrest, cryptorchidism, and poor virilization. Expression of various Leydig cell markers measured by immunohistochemistry, Western blot analysis, and RT-PCR indicated fetal and adult Leydig cell development were differentially impaired. Whereas fetal Leydig cell development was delayed in Nr5a1(-/-);tg(+/0) embryos, it recovered to control levels by birth. In contrast, Sult1e1, Vcam1, and Hsd3b6 transcript levels in adult rescue testes indicated complete blockage in adult Leydig cell development. In addition, between Postnatal Days 8 and 12, peritubular cells expressing PTCH1, SF-1, and CYP11A1 were observed in control testes but not in rescue testes, indicating SF-1 is needed for either survival or differentiation of adult Leydig cell progenitors. Cultured prepubertal rat peritubular cells also expressed SF-1 and PTCH1, but Cyp11a1 was expressed only after treatment with cAMP and retinoic acid. Together, data show SF-1 is needed for proper development of fetal and adult Leydig cells but with distinct primary functions; in fetal Leydig cells, it regulates differentiation, whereas in adult Leydig cells it regulates progenitor cell formation and/or survival.
Collapse
Affiliation(s)
- Tatiana Karpova
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Kumarasamy Ravichandiran
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Lovella Insisienmay
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Daren Rice
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Valentine Agbor
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Leslie L Heckert
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
27
|
Hussain S, Amar A, Najeeb MN, Khaliq S. Two novel mutations in theNR5A1gene as a cause of disorders of sex development in a Pakistani cohort of 46,XY patients. Andrologia 2015; 48:509-17. [DOI: 10.1111/and.12470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2015] [Indexed: 11/29/2022] Open
Affiliation(s)
- S. Hussain
- Department of Biochemistry; University of Health Sciences; Lahore Pakistan
| | - A. Amar
- Department Human Genetics & Molecular Biology; University of Health Sciences; Lahore Pakistan
| | - M. N. Najeeb
- Department of Biochemistry; Quaid-e-Azam Medical Collage; Bahawalpur Pakistan
| | - S. Khaliq
- Department Human Genetics & Molecular Biology; University of Health Sciences; Lahore Pakistan
| |
Collapse
|
28
|
Suntharalingham JP, Buonocore F, Duncan AJ, Achermann JC. DAX-1 (NR0B1) and steroidogenic factor-1 (SF-1, NR5A1) in human disease. Best Pract Res Clin Endocrinol Metab 2015; 29:607-19. [PMID: 26303087 PMCID: PMC5159745 DOI: 10.1016/j.beem.2015.07.004] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
DAX-1 (NR0B1) and SF-1 (NR5A1) are two nuclear receptor transcription factors that play a key role in human adrenal and reproductive development. Loss of DAX-1 function is classically associated with X-linked adrenal hypoplasia congenita. This condition typically affects boys and presents as primary adrenal insufficiency in early infancy or childhood, hypogonadotropic hypogonadism at puberty and impaired spermatogenesis. Late onset forms of this condition and variant phenotypes are increasingly recognized. In contrast, disruption of SF-1 only rarely causes adrenal insufficiency, usually in combination with testicular dysgenesis. Variants in SF-1/NR5A1 more commonly cause a spectrum of reproductive phenotypes ranging from 46,XY DSD (partial testicular dysgenesis or reduced androgen production) and hypospadias to male factor infertility or primary ovarian insufficiency. Making a specific diagnosis of DAX-1 or SF-1 associated conditions is important for long-term monitoring of endocrine and reproductive function, appropriate genetic counselling for family members, and for providing appropriate informed support for young people.
Collapse
Affiliation(s)
| | - Federica Buonocore
- Genetics & Genomic Medicine, UCL Institute of Child Health, University College London, London, UK.
| | - Andrew J Duncan
- Genetics & Genomic Medicine, UCL Institute of Child Health, University College London, London, UK.
| | - John C Achermann
- Genetics & Genomic Medicine, UCL Institute of Child Health, University College London, London, UK.
| |
Collapse
|
29
|
Bertelloni S, Dati E, Baldinotti F, Toschi B, Marrocco G, Sessa MR, Michelucci A, Simi P, Baroncelli GI. NR5A1 gene mutations: clinical, endocrine and genetic features in two girls with 46,XY disorder of sex development. Horm Res Paediatr 2015; 81:104-8. [PMID: 24434652 DOI: 10.1159/000354990] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 08/06/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Steroidogenic factor 1, encoded by the NR5A1 gene, is a key regulator of endocrine function within the hypothalamic-pituitary-steroidogenic axis. Both homozygous, compound heterozygous and heterozygous mutations in the NR5A1 gene may determine 46,XY disorders of sex development (DSD). PATIENTS AND METHODS NR5A1 gene sequencing was performed in a cohort of 6 patients with 46,XY DSD without specific diagnosis. RESULTS Heterozygous NR5A1 gene mutations were found in 2 girls, aged 0.5 years and 14 years. The older girl harbored the c.250C>T transition in exon 4 (p.Arg84Cys), previously reported in a Japanese girl. The younger girl presented a de novo novel exon 6 heterozygous frameshift mutation (c.1074dupG) in codon 359 associated with the p.Gly146Ala polymorphism the latter inherited from her father. This baby showed severe impairment of androgen secretion from the first months of life. Overt adrenal insufficiency did not occur, but the older girl showed subnormal cortisol peak after ACTH stimulation. CONCLUSIONS NR5A1 gene mutations are a relatively frequent cause of 46,XY DSD in humans. Clear indications for management of these individuals remain elusive, mainly when diagnosis is made in infancy. Long-term monitoring of adrenal function should be recommended.
Collapse
Affiliation(s)
- Silvano Bertelloni
- Adolescent Medicine, Division of Pediatrics, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Tuhan HU, Anik A, Catli G, Ceylaner S, Dundar B, Bober E, Abaci A. A novel missense mutation in HSD17B3 gene in a 46, XY adolescent presenting with primary amenorrhea and virilization at puberty. Clin Chim Acta 2015; 438:154-6. [DOI: 10.1016/j.cca.2014.07.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/20/2014] [Indexed: 11/25/2022]
|
31
|
Hiort O, Birnbaum W, Marshall L, Wünsch L, Werner R, Schröder T, Döhnert U, Holterhus PM. Management of disorders of sex development. Nat Rev Endocrinol 2014; 10:520-9. [PMID: 25022812 DOI: 10.1038/nrendo.2014.108] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The medical term disorders of sex development (DSDs) is used to describe individuals with an atypical composition of chromosomal, gonadal and phenotypic sex, which leads to differences in the development of the urogenital tract and reproductive system. A variety of genetic factors have been identified that affect sex development during gonadal differentiation or in specific disorders associated with altered androgen biosynthesis or action. The diagnosis of DSDs in individuals and the subsequent management of patients and their families requires a targeted and structured approach, involving a multidisciplinary team with effective communication between the disciplines. This approach includes distinct clinical, imaging, laboratory and genetic evaluations of patients with DSDs. Although treatment of patients with DSDs can include endocrine and surgical options, many patients have concerns that arise from past incorrect treatments that were founded on the traditional binary concept of the sexes. To dispel these concerns, it is necessary to create centres of expertise for DSDs that include physicians, surgeons, psychologists and specialists in diagnostic procedures to manage patients and their families. Additionally, the inclusion of trained peer support in the multidisciplinary DSD team seems to be integral to the supportive management of patients with DSDs. Most importantly, dealing with DSDs requires acceptance of the fact that deviation from the traditional definitions of gender is not necessarily pathologic.
Collapse
Affiliation(s)
- Olaf Hiort
- Division of Experimental Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Wiebke Birnbaum
- Division of Experimental Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Louise Marshall
- Division of Experimental Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Lutz Wünsch
- Department of Paediatric Surgery, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Ralf Werner
- Division of Experimental Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Tatjana Schröder
- Department of Gynaecology and Obstetrics, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Ulla Döhnert
- Division of Experimental Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Paul-Martin Holterhus
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics, Christian-Albrechts-University, Schwanenweg 20, 24105 Kiel, Germany
| |
Collapse
|
32
|
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
|
33
|
|
34
|
McCann-Crosby B, Mansouri R, Dietrich JE, McCullough LB, Sutton VR, Austin EG, Schlomer B, Roth DR, Karaviti L, Gunn S, Hicks MJ, Macias CG. State of the art review in gonadal dysgenesis: challenges in diagnosis and management. INTERNATIONAL JOURNAL OF PEDIATRIC ENDOCRINOLOGY 2014; 2014:4. [PMID: 24731683 PMCID: PMC3995514 DOI: 10.1186/1687-9856-2014-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 03/14/2014] [Indexed: 11/25/2022]
Abstract
Gonadal dysgenesis, a condition in which gonadal development is interrupted leading to gonadal dysfunction, is a unique subset of disorders of sexual development (DSD) that encompasses a wide spectrum of phenotypes ranging from normally virilized males to slightly undervirilized males, ambiguous phenotype, and normal phenotypic females. It presents specific challenges in diagnostic work-up and management. In XY gonadal dysgenesis, the presence of a Y chromosome or Y-chromosome material renders the patient at increased risk for developing gonadal malignancy. No universally accepted guidelines exist for identifying the risk of developing a malignancy or for determining either the timing or necessity of performing a gonadectomy in patients with XY gonadal dysgenesis. Our goal was to evaluate the literature and develop evidence-based medicine guidelines with respect to the diagnostic work-up and management of patients with XY gonadal dysgenesis. We reviewed the published literature and used the Grading of Recommendation, Assessment, Development, and Evaluation (GRADE) system when appropriate to grade the evidence and to provide recommendations for the diagnostic work-up, malignancy risk stratification, timing or necessity of gonadectomy, role of gonadal biopsy, and ethical considerations for performing a gonadectomy. Individualized health care is needed for patients with XY gonadal dysgenesis, and the decisions regarding gonadectomy should be tailored to each patient based on the underlying diagnosis and risk of malignancy. Our recommendations, based on the evidence available, add an important component to the diagnostic and management armament of physicians who treat patients with these conditions.
Collapse
Affiliation(s)
- Bonnie McCann-Crosby
- Division of Pediatric Endocrinology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Roshanak Mansouri
- Division of Pediatric and Adolescent Gynecology, Department of Obstetrics and Gynecology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Jennifer E Dietrich
- Division of Pediatric and Adolescent Gynecology, Department of Obstetrics and Gynecology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Laurence B McCullough
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - V Reid Sutton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Elise G Austin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Bruce Schlomer
- Division of Pediatric Urology, Department of Surgery, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - David R Roth
- Division of Pediatric Urology, Department of Surgery, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Lefkothea Karaviti
- Division of Pediatric Endocrinology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Sheila Gunn
- Division of Pediatric Endocrinology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - M John Hicks
- Department of Pathology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Charles G Macias
- Evidence-Based Outcomes Center, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| |
Collapse
|
35
|
Sıklar Z, Berberoğlu M, Ceylaner S, Çamtosun E, Kocaay P, Göllü G, Sertçelik A, Öcal G. A novel heterozygous mutation in steroidogenic factor-1 in pubertal virilization of a 46,XY female adolescent. J Pediatr Adolesc Gynecol 2014; 27:98-101. [PMID: 24231572 DOI: 10.1016/j.jpag.2013.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 05/23/2013] [Accepted: 06/04/2013] [Indexed: 11/17/2022]
Abstract
BACKGROUND Steroidogenic factor-1 (SF-1) gene (NR5A1) mutations cause disorders of sexual development due to gonadal dysgenesis, particularly in 46,XY individuals. In cases exhibiting this mutation, the phenotype is heterogeneous, and it may vary within a spectrum ranging from complete female appearance to an infertile male. Virilization observed in some cases in the pubertal age group may lead to diagnostic difficulties. CASE The present case report describes the clinical, histopathologic, and genetic characteristics of a 46,XY case, who was born with a female phenotype and raised as a girl, presented with findings of virilization in the pubertal period. She had no germ cells and very few Leydig cells with atrophic testis on biopsy and in whom a novel heterozygous mutation in the SF-1 gene (a heterozygous 7-bp deletion mutation in exon 7 [c.1308-1314del7bp] causing frameshift) was identified. SUMMARY AND CONCLUSION Although the gonads are very dysgentic in patient with SF-1 mutations, sufficient androgen synthesis can cause severe virilization during puberty.
Collapse
Affiliation(s)
- Zeynep Sıklar
- Department of Pediatric Endocrinology, Ankara University School of Medicine, Ankara, Turkey.
| | - Merih Berberoğlu
- Department of Pediatric Endocrinology, Ankara University School of Medicine, Ankara, Turkey
| | | | - Emine Çamtosun
- Department of Pediatric Endocrinology, Ankara University School of Medicine, Ankara, Turkey
| | - Pınar Kocaay
- Department of Pediatric Endocrinology, Ankara University School of Medicine, Ankara, Turkey
| | - Gülnur Göllü
- Department of Pediatric Surgery, Ankara University School of Medicine, Ankara, Turkey
| | - Ayşe Sertçelik
- Department of Pathology, Ankara University School of Medicine, Ankara, Turkey
| | - Gönül Öcal
- Department of Pediatric Endocrinology, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
36
|
Nishina-Uchida N, Fukuzawa R, Numakura C, Suwanai AS, Hasegawa T, Hasegawa Y. Characteristic testicular histology is useful for the identification of NR5A1 gene mutations in prepubertal 46,XY patients. Horm Res Paediatr 2014; 80:119-28. [PMID: 23969951 DOI: 10.1159/000353763] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 06/06/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Individuals with NR5A1 mutations encoding steroidogenic factor-1 (SF1) develop a phenotypically broad range of disorders of sexual development (DSD). Based on a literature review, we noted that hypoplastic seminiferous tubules and the emergence of Leydig cells with vacuolar cytoplasms are seen predominantly in the majority of individuals with NR5A1 mutations. AIM The aim of this study was to address whether the histopathological characteristics of the testis can be a biomarker for 46,XY individuals with NR5A1 mutations. DESIGN In order to ascertain whether or not the histological features were the characteristics of NR5A1 mutations, we screened the testicular histology of 242 patients with 46,XY DSD and then subsequently assessed NR5A1 mutations. RESULT Of 242 patients with 46,XY DSD, 6 patients matched histological testicular features: a reduced number of thin seminiferous tubules and focal aggregations of Leydig cells that contained cytoplasmic lipid droplets. All 6 patients had NR5A1 mutations. These histological features were distinct from those of other DSD. Thus, this unique testicular histology is useful for identifying NR5A1 mutations in 46,XY patients with DSD before puberty.
Collapse
Affiliation(s)
- Noriko Nishina-Uchida
- Department of Endocrinology and Metabolism, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Suwanai AS, Ishii T, Haruna H, Yamataka A, Narumi S, Fukuzawa R, Ogata T, Hasegawa T. A report of two novel NR5A1 mutation families: possible clinical phenotype of psychiatric symptoms of anxiety and/or depression. Clin Endocrinol (Oxf) 2013; 78:957-65. [PMID: 23095176 DOI: 10.1111/cen.12054] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 08/09/2012] [Accepted: 09/03/2012] [Indexed: 10/27/2022]
Abstract
OBJECTIVE NR5A1 or steroidogenic factor 1 is a nuclear receptor that plays important roles in the hypothalamus-pituitary-steroidogenic axis. The clinical phenotype of most 46,XY mutation carriers includes disorders of sex development (DSD) without adrenal insufficiency, whereas 46,XX mutation carriers have phenotypes ranging from no symptoms to ovarian insufficiency. Although genetically engineered ventromedial hypothalamus-specific Nr5a1 knockout mice show anxiety behaviour, no psychiatric symptoms have been reported in human NR5A1 mutation carriers. We report clinical and molecular findings for individuals (from two families) with NR5A1 mutations, showing psychiatric symptoms. DESIGN AND METHODS We screened for NR5A1 mutations in a cohort of 34 patients with 46,XY DSD using PCR-based sequencing. Psychiatric symptoms were assessed using mental health assessment tools and structured clinical interviews. Functional properties of detected mutant NR5A1s were studied in silico and in vitro, including three-dimensional (3D) mutation models, subcellular NR5A1 protein localization and transactivation assays. RESULTS We found 2 (46,XY) patients with NR5A1 heterozygous novel mutations (p.D257fs and p.V424del), which were transmitted from their respective mothers. The patients' clinical findings indicated DSD without adrenal insufficiency. Both mothers showed psychiatric symptoms, including excessive anxiety and/or depression. The mother and grandmother of one patient had premature ovarian insufficiency. Functional studies showed altered 3D models of p.V424del and normal subcellular NR5A1 localization and impaired transcriptional activation without dominant-negative effects in both mutations. CONCLUSIONS We found 2 (46,XX) NR5A1 mutation carriers with excessive anxiety and/or depression. These results suggest that excessive anxiety and/or depression are possible clinical phenotypes of 46,XX NR5A1 mutations.
Collapse
Affiliation(s)
- Ayuko S Suwanai
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Wu JY, McGown IN, Lin L, Achermann JC, Harris M, Cowley DM, Aftimos S, Neville KA, Choong CS, Cotterill AM. A novel NR5A1 variant in an infant with elevated testosterone from an Australasian cohort of 46,XY patients with disorders of sex development. Clin Endocrinol (Oxf) 2013; 78:545-50. [PMID: 22909003 PMCID: PMC3613751 DOI: 10.1111/cen.12012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 07/09/2012] [Accepted: 08/08/2012] [Indexed: 12/30/2022]
Abstract
BACKGROUND NR5A1 loss-of-function mutations are increasingly found to be the cause of 46,XY disorders of sex development (DSD). OBJECTIVE To determine the presence of NR5A1 mutations in an Australasian cohort of 17 46,XY DSD patients with presumed androgen insensitivity syndrome (AIS) who were negative for androgen receptor gene (AR) mutation. DESIGN Exons 2-7 of NR5A1 were PCR amplified and sequenced. Gene expression and cellular localization studies were performed on a novel NR5A1 variant c.74A>G (p.Y25C) identified in this study. RESULTS We identified one novel mutation, c.74A>G (p.Y25C) in a patient characterized by penoscrotal hypospadias with bifid scrotum. He had elevated testosterone and gonadotropins in early infancy. Functional analysis of p.Y25C in vitro demonstrated reduced transcriptional activation by SF-1 and partially impaired nuclear localization in a proportion of transfected human adrenal NCI-H295R cells. CONCLUSION This is the first reported case of a DSD patient with a NR5A1 mutation and elevated testosterone levels. Our finding supports evaluation of NR5A1 mutations in 46,XY DSD patients with a range of testosterone levels.
Collapse
Affiliation(s)
- Joyce Y Wu
- Department of Clinical Chemistry, Mater Hospital, South Brisbane, QLD, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
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
|