Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Clin Genet ; 103(3): 277-287, 2023 03.
Article in English | MEDLINE | ID: mdl-36349847

ABSTRACT

46,XY gonadal dysgenesis (GD) is a Disorder/Difference of Sex Development (DSD) that can present with phenotypes ranging from ambiguous genitalia to complete male-to-female sex reversal. Around 50% of 46,XY DSD cases receive a molecular diagnosis. In mice, Fibroblast growth factor 9 (FGF9) is an important component of the male sex-determining pathway. Two FGF9 variants reported to date disrupt testis development in mice, but not in humans. Here, we describe a female patient with 46,XY GD harbouring the rare FGF9 variant (missense mutation), NM_002010.2:c.583G > A;p.(Asp195Asn) (D195N). By biochemical and cell-based approaches, the D195N variant disrupts FGF9 protein homodimerisation and FGF9-heparin-binding, and reduces both Sertoli cell proliferation and Wnt4 repression. XY Fgf9D195N/D195N foetal mice show a transient disruption of testicular cord development, while XY Fgf9D195N/- foetal mice show partial male-to-female gonadal sex reversal. In the general population, the D195N variant occurs at an allele frequency of 2.4 × 10-5 , suggesting an oligogenic basis for the patient's DSD. Exome analysis of the patient reveals several known and novel variants in genes expressed in human foetal Sertoli cells at the time of sex determination. Taken together, our results indicate that disruption of FGF9 homodimerization impairs testis determination in mice and, potentially, also in humans in combination with other variants.


Subject(s)
Fibroblast Growth Factor 9 , Gonadal Dysgenesis, 46,XY , Humans , Male , Female , Mice , Animals , Dimerization , Fibroblast Growth Factor 9/genetics , Testis , Gonads , Gonadal Dysgenesis, 46,XY/genetics
2.
Hum Mutat ; 43(3): 362-379, 2022 03.
Article in English | MEDLINE | ID: mdl-34918413

ABSTRACT

SRY is the Y-chromosomal gene that determines male sex development in humans and most other mammals. After three decades of study, we still lack a detailed understanding of which domains of the SRY protein are required to engage the pathway of gene activity leading to testis development. Some insight has been gained from the study of genetic variations underlying differences/disorders of sex determination (DSD), but the lack of a system of experimentally generating SRY mutations and studying their consequences in vivo has limited progress in the field. To address this issue, we generated a mouse model carrying a human SRY transgene able to drive testis determination in XX mice. Using CRISPR-Cas9 gene editing, we generated novel genetic modifications in each of SRY's three domains (N-terminal, HMG box, and C-terminal) and performed a detailed analysis of their molecular and cellular effects on embryonic testis development. Our results provide new functional insights unique to human SRY and present a versatile and powerful system in which to functionally analyze variations of SRY including known and novel pathogenic variants found in DSD.


Subject(s)
Gene Editing , Mice, Transgenic , Sex-Determining Region Y Protein , Testis , Animals , Humans , Male , Mice , Protein Domains , Sex-Determining Region Y Protein/genetics , Testis/metabolism
3.
Hum Mol Genet ; 29(13): 2148-2161, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32452519

ABSTRACT

In mice, male sex determination depends on FGF9 signalling via FGFR2c in the bipotential gonads to maintain the expression of the key testis gene SOX9. In humans, however, while FGFR2 mutations have been linked to 46,XY disorders of sex development (DSD), the role of FGF9 is unresolved. The only reported pathogenic mutations in human FGF9, FGF9S99N and FGF9R62G, are dominant and result in craniosynostosis (fusion of cranial sutures) or multiple synostoses (fusion of limb joints). Whether these synostosis-causing FGF9 mutations impact upon gonadal development and DSD etiology has not been explored. We therefore examined embryonic gonads in the well-characterized Fgf9 missense mouse mutants, Fgf9S99N and Fgf9N143T, which phenocopy the skeletal defects of FGF9S99N and FGF9R62G variants, respectively. XY Fgf9S99N/S99N and XY Fgf9N143T/N143T fetal mouse gonads showed severely disorganized testis cords and partial XY sex reversal at 12.5 days post coitum (dpc), suggesting loss of FGF9 function. By 15.5 dpc, testis development in both mutants had partly recovered. Mitotic analysis in vivo and in vitro suggested that the testicular phenotypes in these mutants arise in part through reduced proliferation of the gonadal supporting cells. These data raise the possibility that human FGF9 mutations causative for dominant skeletal conditions can also lead to loss of FGF9 function in the developing testis, at least in mice. Our data suggest that, in humans, testis development is largely tolerant of deleterious FGF9 mutations which lead to skeletal defects, thus offering an explanation as to why XY DSDs are rare in patients with pathogenic FGF9 variants.


Subject(s)
Fibroblast Growth Factor 9/genetics , Ovotesticular Disorders of Sex Development/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Synostosis/genetics , Animals , Disease Models, Animal , Female , Gene Expression Regulation, Developmental/genetics , Gonads/growth & development , Gonads/pathology , Humans , Male , Mice , Mutation, Missense/genetics , Ovotesticular Disorders of Sex Development/pathology , SOX9 Transcription Factor/genetics , Sex Determination Processes/genetics , Sexual Development/genetics
4.
Clin Genet ; 99(2): 325-329, 2021 02.
Article in English | MEDLINE | ID: mdl-33174625

ABSTRACT

Human multiple synostoses syndrome 3 is an autosomal dominant disorder caused by pathogenic variants in FGF9. Only two variants have been described in FGF9 in humans so far, and one in mice. Here we report a novel missense variant c.566C > G, p.(Pro189Arg) in FGF9. Functional studies showed this variant impairs FGF9 homodimerization, but not FGFR3c binding. We also review the findings of cases reported previously and report on additional features not described previously.


Subject(s)
Fibroblast Growth Factor 9/genetics , Mutation, Missense , Synostosis/genetics , Abnormalities, Multiple/genetics , Adolescent , Fibroblast Growth Factor 9/metabolism , Heterozygote , Humans , Male , Phenotype , Protein Binding , Radiography , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Synostosis/diagnostic imaging , Synostosis/pathology
5.
Hum Mutat ; 40(2): 207-216, 2019 02.
Article in English | MEDLINE | ID: mdl-30350900

ABSTRACT

Several recent reports have described a missense variant in the gene NR5A1 (c.274C>T; p.Arg92Trp) in a significant number of 46,XX ovotesticular or testicular disorders of sex development (DSDs) cases. The affected residue falls within the DNA-binding domain of the NR5A1 protein, however the exact mechanism by which it causes testicular development in 46,XX individuals remains unclear. We have screened a cohort of 26 patients with 46,XX (ovo)testicular DSD and identified three unrelated individuals with this NR5A1 variant (p.Arg92Trp), as well as one patient with a novel NR5A1 variant (c.779C>T; p.Ala260Val). We examined the functional effect of these changes, finding that while protein levels and localization were unaffected, variant NR5A1 proteins repress the WNT signaling pathway and have less ability to upregulate the anti-testis gene NR0B1. These findings highlight how NR5A1 variants impact ovarian differentiation across multiple pathways, resulting in a switch from ovarian to testis development in genetic females.


Subject(s)
46, XX Disorders of Sex Development/genetics , Disorders of Sex Development/genetics , Steroidogenic Factor 1/genetics , Testis/pathology , 46, XX Disorders of Sex Development/pathology , Adolescent , Adult , Child, Preschool , DNA-Binding Proteins/genetics , Disorders of Sex Development/pathology , Female , Humans , Infant , Male , Mutation, Missense/genetics , Pedigree , Phenotype , Protein Domains/genetics , Testis/growth & development , Wnt Signaling Pathway/genetics
6.
Hum Mutat ; 39(12): 1861-1874, 2018 12.
Article in English | MEDLINE | ID: mdl-30067310

ABSTRACT

Nuclear receptor subfamily 5 group A member 1/Steroidogenic factor 1 (NR5A1; SF-1; Ad4BP) mutations cause 46,XY disorders of sex development (DSD), with phenotypes ranging from developmentally mild (e.g., hypospadias) to severe (e.g., complete gonadal dysgenesis). The molecular mechanism underlying this spectrum is unclear. During sex determination, SF-1 regulates SOX9 (SRY [sex determining region Y]-box 9) expression. We hypothesized that SF-1 mutations in 46,XY DSD patients affect SOX9 expression via the Testis-specific Enhancer of Sox9 core element, TESCO. Our objective was to assess the ability of 20 SF-1 mutants found in 46,XY DSD patients to activate TESCO. Patient DNA was sequenced for SF-1 mutations and mutant SF-1 proteins were examined for transcriptional activity, protein expression, sub-cellular localization and in silico structural defects. Fifteen of the 20 mutants showed reduced SF-1 activation on TESCO, 11 with atypical sub-cellular localization. Fourteen SF-1 mutants were predicted in silico to alter DNA, ligand or cofactor interactions. Our study may implicate aberrant SF-1-mediated transcriptional regulation of SOX9 in 46,XY DSDs.


Subject(s)
Disorder of Sex Development, 46,XY/genetics , Enhancer Elements, Genetic , Mutation , SOX9 Transcription Factor/genetics , Steroidogenic Factor 1/genetics , Adolescent , Adult , Child , Child, Preschool , Computer Simulation , Gene Expression Regulation , HEK293 Cells , Humans , Infant , Infant, Newborn , Ligands , Male , Protein Binding , Sequence Analysis, DNA/methods , Steroidogenic Factor 1/chemistry , Steroidogenic Factor 1/metabolism
7.
Birth Defects Res C Embryo Today ; 108(4): 337-350, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28033663

ABSTRACT

Disorders of Sex Development (DSDs) are a major paediatric concern and are estimated to occur in around 1.7% of all live births (Fausto-Sterling, Sexing the Body: Gender Politics and the Construction of Sexuality, Basic Books, New York, 2000). They are often caused by the breakdown in the complex genetic mechanisms that underlie gonadal development and differentiation. Having a genetic diagnosis can be important for patients with a DSD: it can increase acceptance of a disorder often surrounded by stigma, alter clinical management and it can assist in reproductive planning. While Massively Parallel Sequencing (MPS) is advancing the genetic diagnosis of rare Mendelian disorders, it is not yet clear which MPS assay is best suited for the clinical diagnosis of DSD patients and to what extent other established methods are still relevant. To complicate matters, DSDs represent a wide spectrum of disorders caused by an array of different genetic changes, many of which are yet unknown. Here we discuss the different genetic lesions that are known to contribute to different DSDs, and review the utility of a range of MPS approaches for diagnosing DSD patients. Birth Defects Research (Part C) 108:337-350, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Disorders of Sex Development/diagnosis , Disorders of Sex Development/genetics , Animals , DNA Copy Number Variations , Disorders of Sex Development/metabolism , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Testing , Genomics/methods , Gonadal Disorders/genetics , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/statistics & numerical data , Humans , Sexual Development/genetics
8.
Endocrinology ; 164(5)2023 03 13.
Article in English | MEDLINE | ID: mdl-36786658

ABSTRACT

During sex determination in the mouse, fibroblast growth factor 9 signals through the fibroblast growth factor receptor 2c isoform (FGFR2c) to trigger Sertoli cell and testis development from 11.5 days post coitum (dpc). In the XX gonad, the FOXL2 and WNT4/RSPO1 pathways drive granulosa cell and ovarian development. The function of FGFR2 in the developing ovary, and whether FGFR2 is required in the testis after sex determination, is not clear. In fetal mouse gonads from 12.5 dpc, FGFR2 shows sexually dimorphic expression. In XX gonads, FGFR2c is coexpressed with FOXL2 in pregranulosa cells, whereas XY gonads show FGFR2b expression in germ cells. Deletion of Fgfr2c in XX mice led to a marked decrease/absence of germ cells by 13.5 dpc in the ovary. This indicates that FGFR2c in the somatic pregranulosa cells is required for the maintenance of germ cells. Surprisingly, on the Fgfr2c-/- background, the germ cell phenotype could be rescued by ablation of Foxl2, suggesting a novel mechanism whereby FGFR2 and FOXL2 act antagonistically during germ cell development. Consistent with low/absent FGFR2 expression in the Sertoli cells of 12.5 and 13.5 dpc XY gonads, XY AMH:Cre; Fgfr2flox/flox mice showed normal testis morphology and structures during fetal development and in adulthood. Thus, FGFR2 is not essential for maintaining Sertoli cell fate after sex determination. Combined, these data show that FGFR2 is not necessary for Sertoli cell function after sex determination but does play an important role in the ovary.


Subject(s)
Ovary , Receptor, Fibroblast Growth Factor, Type 2 , Male , Female , Mice , Animals , Ovary/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Gonads/metabolism , Testis/metabolism , Germ Cells/metabolism , Sex Determination Processes
9.
Maturitas ; 131: 78-86, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31787151

ABSTRACT

Ovarian deficiency, including diminished ovarian reserve and premature ovarian insufficiency, represents one of the main causes of female infertility. Little is known of the genetic basis of diminished ovarian reserve, while premature ovarian insufficiency often has a genetic basis, with genes affecting various processes. NR5A1 is a key gene required for gonadal function, and variants are associated with a wide phenotypic spectrum of disorders of sexual development, and are found in 0.26-8% of patients with premature ovarian insufficiency. As there is some debate about the extent of involvement of NR5A1 in the pathogenesis of ovarian deficiency, we performed an in-depth analysis of NR5A1 variants detected in a cohort of 142 patients with premature ovarian insufficiency, diminished ovarian reserve, or unexplained infertility associated with normal ovarian function. We identified rare non-synonymous protein-altering variants in 2.8 % of women with ovarian deficiency and no such variants in our small cohort of women with infertility but normal ovarian function. We observed previously reported variants associated with premature ovarian insufficiency in patients with diminished ovarian reserve, highlighting a genetic relationship between these conditions. We confirmed functional impairment resulting from a p.Val15Met variant, detected for the first time in a patient with premature ovarian insufficiency. The remaining variants were associated with preserved transcriptional activity and localization of NR5A1, indicating that rare NR5A1 variants may be incorrectly curated if functional studies are not undertaken, and/or that NR5A1 variants may have only a subtle impact on protein function and/or confer risk of ovarian deficiency via oligogenic inheritance.


Subject(s)
Infertility, Female/genetics , Menopause, Premature/genetics , Ovarian Reserve , Primary Ovarian Insufficiency/genetics , Steroidogenic Factor 1/genetics , Adult , Alleles , Black People , Cohort Studies , Female , Gene Expression Regulation , HEK293 Cells , Humans , Infertility, Female/ethnology , Menopause, Premature/ethnology , Mutation , Primary Ovarian Insufficiency/ethnology
11.
Sex Dev ; 12(1-3): 19-29, 2018.
Article in English | MEDLINE | ID: mdl-29145200

ABSTRACT

Despite considerable research effort and significant advances in sequencing technologies, the majority of disorders of sex development (DSD) cases still lack a molecular genetic diagnosis. While coding variants have been discovered in known and candidate DSD genes, comparatively little is known about copy number variations (CNVs) affecting both coding and noncoding regions. Due to rapidly falling costs of whole genome sequencing, many more CNVs in individuals with DSD will be identified. These CNVs may explain a significant number of hitherto undiagnosed cases of DSD. In this review, we provide an overview of CNVs that are known to cause DSD and discuss approaches to identify and verify causative CNVs.


Subject(s)
DNA Copy Number Variations/genetics , Disorders of Sex Development/genetics , Chromosome Mapping , High-Throughput Nucleotide Sequencing , Humans , Open Reading Frames/genetics , Regulatory Sequences, Nucleic Acid/genetics
12.
Nat Commun ; 9(1): 5319, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30552336

ABSTRACT

Disorders of sex development (DSDs) are conditions affecting development of the gonads or genitalia. Variants in two key genes, SRY and its target SOX9, are an established cause of 46,XY DSD, but the genetic basis of many DSDs remains unknown. SRY-mediated SOX9 upregulation in the early gonad is crucial for testis development, yet the regulatory elements underlying this have not been identified in humans. Here, we identified four DSD patients with overlapping duplications or deletions upstream of SOX9. Bioinformatic analysis identified three putative enhancers for SOX9 that responded to different combinations of testis-specific regulators. All three enhancers showed synergistic activity and together drive SOX9 in the testis. This is the first study to identify SOX9 enhancers that, when duplicated or deleted, result in 46,XX or 46,XY sex reversal, respectively. These enhancers provide a hitherto missing link by which SRY activates SOX9 in humans, and establish SOX9 enhancer mutations as a significant cause of DSD.


Subject(s)
Disorders of Sex Development/genetics , Gene Duplication/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sequence Deletion/genetics , Sex-Determining Region Y Protein/genetics , 46, XX Disorders of Sex Development/genetics , Animals , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Disorder of Sex Development, 46,XY/genetics , Female , Gene Knockout Techniques , Genitalia/metabolism , Gonads/metabolism , Humans , Male , Mice , Mutation , RNA Splicing Factors/metabolism , Regulatory Sequences, Nucleic Acid/genetics , SOX Transcription Factors/genetics , Sex Differentiation , Testis/growth & development , Testis/metabolism
13.
Sex Dev ; 10(2): 59-65, 2016.
Article in English | MEDLINE | ID: mdl-27078861

ABSTRACT

Despite recent advances in our understanding, most cases of disorders of sex development (DSD) cannot be explained by mutations in known genes. In genome-wide screens of DSD patients, we and others detected duplications or deletions of potential regulatory regions of known or suspected DSD genes. It is therefore likely that a significant proportion of DSD cases may be explained by disrupted transcriptional regulation of gonad genes. Despite many recent technological advances, limited availability of relevant tissues - especially human embryonic material - can make the identification of long-range regulatory elements extremely difficult. In an attempt to overcome this limitation, we evaluated the usefulness of publicly available DNaseI hypersensitivity data from the Roadmap Epigenomics Project. For this feasibility study we used the 'gene desert' around the SOX9 gene and a genomic locus downstream of GATA4. Over 60% of our selected candidate regions had significant enhancer activity in luciferase assays. We show that this approach facilitates the detection of strong enhancer candidates worthy of further analysis.


Subject(s)
Databases, Genetic , Disorders of Sex Development/genetics , Gene Expression Regulation/physiology , Female , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation/genetics , Genome, Human/genetics , Humans , Male , Regulatory Sequences, Nucleic Acid/genetics , Regulatory Sequences, Nucleic Acid/physiology , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL