Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Publication year range
1.
BMC Pediatr ; 23(1): 182, 2023 04 19.
Article in English | MEDLINE | ID: mdl-37072715

ABSTRACT

BACKGROUND: Adolescents with 46,XY disorders of sex development (DSD) face additional medical and psychological challenges. To optimize management and minimize hazards, correct and early clinical and molecular diagnosis is necessary. CASE PRESENTATION: We report a 13-year-old Chinese adolescent with absent Müllerian derivatives and suspected testis in the inguinal area. History, examinations, and assistant examinations were available for clinical diagnosis of 46,XY DSD. The subsequent targeting specific disease-causing genes, comprising 360 endocrine disease-causing genes, was employed for molecular diagnosis. A novel variation in nuclear receptor subfamily 5 group A member 1 (NR5A1) [c.64G > T (p.G22C)] was identified in the patient. In vitro functional analyses of the novel variant suggested no impairment to NR5A1 mRNA or protein expression relative to wild-type, and immunofluorescence confirmed similar localization of NR5A1 mutant to the cell nucleus. However, we observed decreased DNA-binding affinity by the NR5A1 variant, while dual-luciferase reporter assays showed that the mutant effectively downregulated the transactivation capacity of anti-Müllerian hormone. We described a novel NR5A1 variant and demonstrated its adverse effects on the functional integrity of the NR5A1 protein resulting in serious impairment of its modulation of gonadal development. CONCLUSIONS: This study adds one novel NR5A1 variant to the pool of pathogenic variants and enriches the adolescents of information available about the mutation spectrum of this gene in Chinese population.


Subject(s)
Disorder of Sex Development, 46,XY , Steroidogenic Factor 1 , Adolescent , Humans , Male , Disorder of Sex Development, 46,XY/diagnosis , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/pathology , East Asian People/genetics , Mutation , Steroidogenic Factor 1/genetics
2.
Hum Mol Genet ; 28(14): 2319-2329, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30985895

ABSTRACT

Disorders of sex development (DSDs) are defined as congenital conditions in which chromosomal, gonadal or anatomical sex is atypical. In many DSD cases, genetic causes remain to be elucidated. Here, we performed a case-control exome sequencing study comparing gene-based burdens of rare damaging variants between 26 DSD cases and 2625 controls. We found exome-wide significant enrichment of rare heterozygous truncating variants in the MYRF gene encoding myelin regulatory factor, a transcription factor essential for oligodendrocyte development. All three variants occurred de novo. We identified an additional 46,XY DSD case of a de novo damaging missense variant in an independent cohort. The clinical symptoms included hypoplasia of Müllerian derivatives and ovaries in 46,XX DSD patients, defective development of Sertoli and Leydig cells in 46,XY DSD patients and congenital diaphragmatic hernia in one 46,XY DSD patient. As all of these cells and tissues are or partly consist of coelomic epithelium (CE)-derived cells (CEDC) and CEDC developed from CE via proliferaiton and migration, MYRF might be related to these processes. Consistent with this hypothesis, single-cell RNA sequencing of foetal gonads revealed high expression of MYRF in CE and CEDC. Reanalysis of public chromatin immunoprecipitation sequencing data for rat Myrf showed that genes regulating proliferation and migration were enriched among putative target genes of Myrf. These results suggested that MYRF is a novel causative gene of 46,XY and 46,XX DSD and MYRF is a transcription factor regulating CD and/or CEDC proliferation and migration, which is essential for development of multiple organs.


Subject(s)
46, XX Disorders of Sex Development/genetics , Disorder of Sex Development, 46,XY/genetics , Membrane Proteins/genetics , Transcription Factors/genetics , 46, XX Disorders of Sex Development/pathology , Adolescent , Case-Control Studies , Cell Movement , Cell Proliferation , Child, Preschool , Cohort Studies , Computational Biology , Disorder of Sex Development, 46,XY/pathology , Female , Gene Ontology , Gonads/growth & development , Haploinsufficiency , Humans , Male , Membrane Proteins/metabolism , Mutation , Mutation, Missense , Single-Cell Analysis , Transcription Factors/metabolism , Exome Sequencing , Young Adult
3.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768925

ABSTRACT

Congenital anomalies of the female reproductive tract that present with primary amenorrhea involve Müllerian aplasia, also known as Mayer-Rokitansky-Küster-Hauser syndrome (MRKHS), and cervical and vaginal anomalies that completely obstruct the reproductive tract. Karyotype abnormalities do not exclude the diagnosis of MRKHS. Familial cases of Müllerian anomalies and associated malformations of the urinary and skeletal systems strongly suggest a complex genetic etiology, but so far, the molecular mechanism in the vast majority of cases remains unknown. Primary amenorrhea may also be the first presentation of complete androgen insensitivity syndrome, steroid 5α-reductase type 2 deficiency, 17ß-hydroxysteroid dehydrogenase type 3 deficiency, and Leydig cells hypoplasia type 1; therefore, these disorders should be considered in the differential diagnosis of the congenital absence of the uterus and vagina. The molecular diagnosis in the majority of these cases can be established.


Subject(s)
46, XX Disorders of Sex Development/pathology , Amenorrhea/genetics , Amenorrhea/pathology , Cervix Uteri/abnormalities , Congenital Abnormalities/pathology , Mullerian Ducts/abnormalities , Vagina/abnormalities , 17-Hydroxysteroid Dehydrogenases/deficiency , 17-Hydroxysteroid Dehydrogenases/genetics , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/pathology , Cervix Uteri/embryology , Cholestenone 5 alpha-Reductase/deficiency , Cholestenone 5 alpha-Reductase/genetics , Congenital Abnormalities/diagnosis , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/pathology , Female , Humans , Male , Mullerian Ducts/pathology , Testis/abnormalities , Testis/pathology , Vagina/embryology
4.
Ann Hum Genet ; 84(2): 151-160, 2020 03.
Article in English | MEDLINE | ID: mdl-31613402

ABSTRACT

5α-Reductase type 2 deficiency causes a 46,XY disorder of sex development (DSD) characterized by ambiguous external genitalia, rudimentary prostate, and normal internal genitalia. The disease prevalence worldwide is low, but in a small and isolated village of the Venezuelan Andes, a higher incidence has been found. DNA analysis of the SRD5A2 gene was performed in three inbred affected individuals clinically diagnosed with DSD. The entire coding regions, the p.L89V polymorphism (rs523349) and five intragenic SNPs (rs2300702, rs2268797, rs2268796, rs4952220, rs12470196) used to construct haplotypes were analyzed by Sanger sequencing. To assess the probable ethnic origin of the mutation in this geographic isolate, a population structure analysis was performed. Homozygosis for the p.N193S mutation was found in all patients, with a mutation carrier frequency of 1:80 chromosomes (0.0125) in the geographic focus, suggesting a founder phenomenon. The results of the population structure analysis suggested a mutation origin closer to the Spanish populations, according to the clusters grouping. The genotype-phenotype correlation in the patients was not absolute, being hypospadias and cryptorchidism the main traits that differentiate affected individuals.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Disorder of Sex Development, 46,XY/genetics , Membrane Proteins/genetics , Mutation , Polymorphism, Genetic , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/deficiency , Adolescent , Case-Control Studies , Child , Disorder of Sex Development, 46,XY/enzymology , Disorder of Sex Development, 46,XY/epidemiology , Disorder of Sex Development, 46,XY/pathology , Female , Humans , Infant , Male , Membrane Proteins/deficiency , Phenotype , Prognosis , Venezuela/epidemiology
5.
Am J Med Genet A ; 182(12): 2887-2890, 2020 12.
Article in English | MEDLINE | ID: mdl-32949097

ABSTRACT

Differences in sex development (DSD) are a group of rare conditions involving genes, hormones and reproductive organs, including genitals. Although these disorders are common, information about the molecular causes remain limited. Many genes have been identified in association with DSD but in many cases the causative gene could not be identified. The Lhx9 gene has been studied in mice and birds, and biallelic mutations in this gene have been found to cause 46,XY DSD and limb abnormalities. So far two variants of LHX9 have been identified in 46,XY individuals with testicular regression, micropenis and hypospadias. We report a de novo heterozygous missense variant in LHX9 in a girl with 46,XY DSD and finger and toe abnormalities. It was previously predicted that a mutation in LHX9 would not cause extragenital anomalies in light of prior animal studies, but our report adds to the limited knowledge of the phenotype observed in humans with a variant in LHX9. To the best of our knowledge this is the first reported case with this combination of abnormalities.


Subject(s)
Disorder of Sex Development, 46,XY/pathology , LIM-Homeodomain Proteins/genetics , Limb Deformities, Congenital/pathology , Mutation, Missense , Transcription Factors/genetics , Adult , Child , Disorder of Sex Development, 46,XY/complications , Disorder of Sex Development, 46,XY/genetics , Female , Humans , Infant, Newborn , Limb Deformities, Congenital/complications , Limb Deformities, Congenital/genetics , Male , Phenotype , Young Adult
6.
J Assist Reprod Genet ; 37(2): 477-486, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31938931

ABSTRACT

PURPOSE: This study aimed to present the clinical features and gene mutation characteristics of a child with 46,XY disorders of sex development (DSD) caused by a novel heterozygous mutation in the NR5A1 gene to determine the potential association between this heterozygous mutation and the pathogenesis of 46,XY DSD. METHODS: We present the case of a Chinese child with ambiguous genitalia at birth but a normal adrenal gland. Targeted next-generation sequencing, comprising 163 candidate genes involved in sexual differentiation and development, was performed, followed by the functional evaluation of the novel NR5A1 mutation. RESULT: The patient had a novel heterozygous mutation in the NR5A1 gene, c.630delG (p.Y211Tfs*85). Results revealed that overexpression of p.Y211Tfs*85 impaired steroidogenic factor-1 (SF-1) protein synthesis. Immunofluorescence analysis revealed that both SF-1 wild-type and p.Y211Tfs*85 mutation proteins were localized in the cell nucleus. Furthermore, dual-luciferase reporter assay results revealed that the p.Y211Tfs*85 mutation could effectively downregulate the transcriptional activation of anti-Müllerian hormone and steroidogenic acute regulatory protein genes (P < 0.01). Additionally, the p.Y211Tfs*85 mutation changed three-dimensional conformation of SF-1, and three conformations could be constructed with the mutated amino acid sequences. Therefore, the novel frameshift mutation could result in decreased protein expression of SF-1. CONCLUSION: We described a novel mutation in NR5A1 and showed that it might affect protein structure, thereby seriously compromising the role of SF-1 in regulating gonadal development. The novel p.Y211Tfs*85 mutation in the NR5A1 gene enriches the boy of information available regarding the mutation spectrum of this gene in the Chinese population.


Subject(s)
Disorder of Sex Development, 46,XY/genetics , Genetic Predisposition to Disease , Steroidogenic Factor 1/genetics , Amino Acid Sequence/genetics , Child , China/epidemiology , Disorder of Sex Development, 46,XY/pathology , Female , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Male , Mutation
7.
Int J Mol Sci ; 21(22)2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33202802

ABSTRACT

Variants of NR5A1 are often found in individuals with 46,XY disorders of sex development (DSD) and manifest with a very broad spectrum of clinical characteristics and variable sex hormone levels. Such complex phenotypic expression can be due to the inheritance of additional genetic hits in DSD-associated genes that modify sex determination, differentiation and organ function in patients with heterozygous NR5A1 variants. Here we describe the clinical, biochemical and genetic features of a series of seven patients harboring monoallelic variants in the NR5A1 gene. We tested the transactivation activity of novel NR5A1 variants. We additionally included six of these patients in a targeted diagnostic gene panel for DSD and identified a second genetic hit in known DSD-causing genes STAR, AMH and ZFPM2/FOG2 in three individuals. Our study increases the number of NR5A1 variants related to 46,XY DSD and supports the hypothesis that a digenic mode of inheritance may contribute towards the broad spectrum of phenotypes observed in individuals with a heterozygous NR5A1 variation.


Subject(s)
DNA-Binding Proteins/genetics , Disorder of Sex Development, 46,XY/genetics , Genetic Variation , Heterozygote , Multifactorial Inheritance , Phosphoproteins/genetics , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Steroidogenic Factor 1/genetics , Transcription Factors/genetics , Adolescent , Child , Child, Preschool , Disorder of Sex Development, 46,XY/pathology , Female , Humans , Infant , Infant, Newborn , Male
8.
Endocr J ; 66(9): 837-842, 2019 Sep 28.
Article in English | MEDLINE | ID: mdl-31178538

ABSTRACT

Steroid 5α-reductase type 2 deficiency (5αRD2) is a congenital disorder of sex development caused by impairment of conversion from testosterone (T) to 5α-dihydrotestosterone (DHT). DHT deficiency leads to various degrees of undervirilized external genitalia including micropenis, primarily correlated with mutations of the SRD5A2 gene that encodes 5α-reductase type 2. Four Japanese boys with isolated micropenis were diagnosed as 5αRD2 by elevated ratios of serum T/DHT, and decreased ratios of urinary 5α/5ß-reduced steroid metabolites. Genetic analyses for SRD5A2 identified that the four patients shared a hypomorphic mutation R227Q that has a residual activity related to the mild-form of 5αRD2. For prepubertal micropenis, DHT was transdermally applied to the four patients at the ages of 4-11 year, increasing a median of stretched penile lengths (SPLs) from 2.6 cm (-2.5 SD) to 4.4 cm (-0.2 SD). Nevertheless, the post-pubertal penile growth was apparently retarded, despite normal levels of T secreted from well-developed testes. The second course of DHT treatment underwent at ages of 12-18 year, but unable to normalize SPLs at a range of 6.0 to 7.0 cm (-3.4 to -2.4 SD). The prostate volumes of two patients were variable at 8.1 and 21 cm3, and a sperm cell count of one patient was normal as young adult. DHT treatment contributes to development of the penis and prostate, which are favorable for the potential fertility of 5αRD2 adults. Meanwhile, the retarded penile growth and a risk of prostate overgrowth may complicate the post-pubertal management with DHT for 5αRD2 males.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/deficiency , Dihydrotestosterone/administration & dosage , Disorder of Sex Development, 46,XY/drug therapy , Genital Diseases, Male/drug therapy , Hypospadias/drug therapy , Penis/abnormalities , Penis/drug effects , Puberty/drug effects , Steroid Metabolism, Inborn Errors/drug therapy , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/blood , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Child , Child, Preschool , Disorder of Sex Development, 46,XY/blood , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/pathology , Drug Administration Schedule , Genital Diseases, Male/blood , Genital Diseases, Male/genetics , Humans , Hypospadias/blood , Hypospadias/genetics , Hypospadias/pathology , Longitudinal Studies , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mutation , Penis/growth & development , Penis/pathology , Puberty/physiology , Sexual Maturation/drug effects , Steroid Metabolism, Inborn Errors/blood , Steroid Metabolism, Inborn Errors/genetics , Steroid Metabolism, Inborn Errors/pathology , Testosterone/blood , Time Factors , Treatment Outcome
9.
Hum Mol Genet ; 25(16): 3446-3453, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27378692

ABSTRACT

Cell lineages of the early human gonad commit to one of the two mutually antagonistic organogenetic fates, the testis or the ovary. Some individuals with a 46,XX karyotype develop testes or ovotestes (testicular or ovotesticular disorder of sex development; TDSD/OTDSD), due to the presence of the testis-determining gene, SRY Other rare complex syndromic forms of TDSD/OTDSD are associated with mutations in pro-ovarian genes that repress testis development (e.g. WNT4); however, the genetic cause of the more common non-syndromic forms is unknown. Steroidogenic factor-1 (known as NR5A1) is a key regulator of reproductive development and function. Loss-of-function changes in NR5A1 in 46,XY individuals are associated with a spectrum of phenotypes in humans ranging from a lack of testis formation to male infertility. Mutations in NR5A1 in 46,XX women are associated with primary ovarian insufficiency, which includes a lack of ovary formation, primary and secondary amenorrhoea as well as early menopause. Here, we show that a specific recurrent heterozygous missense mutation (p.Arg92Trp) in the accessory DNA-binding region of NR5A1 is associated with variable degree of testis development in 46,XX children and adults from four unrelated families. Remarkably, in one family a sibling raised as a girl and carrying this NR5A1 mutation was found to have a 46,XY karyotype with partial testicular dysgenesis. These unique findings highlight how a specific variant in a developmental transcription factor can switch organ fate from the ovary to testis in mammals and represents the first missense mutation causing isolated, non-syndromic 46,XX testicular/ovotesticular DSD in humans.


Subject(s)
DNA-Binding Proteins/genetics , Disorder of Sex Development, 46,XY/genetics , Primary Ovarian Insufficiency/genetics , Sexual Development/genetics , Steroidogenic Factor 1/genetics , Adult , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/pathology , Cell Lineage/genetics , Child , Disorder of Sex Development, 46,XY/pathology , Female , Gonads/growth & development , Gonads/pathology , Humans , Karyotype , Male , Mutation, Missense , Ovary/growth & development , Ovary/pathology , Pedigree , Primary Ovarian Insufficiency/pathology , Sex Determination Processes , Testis/growth & development , Testis/pathology
10.
Hum Mol Genet ; 23(14): 3657-65, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24549039

ABSTRACT

In recent years, considerable advances have been made in our understanding of genetics of mammalian gonad development; however, the underlying genetic aetiology in the majority of patients with 46,XY disorders of sex development (DSD) still remains unknown. Based on mouse models, it has been hypothesized that haploinsufficiency of the Friend of GATA 2 (FOG2) gene could lead to 46,XY gonadal dysgenesis on specific inbred genetic backgrounds. Using whole exome sequencing, we identified independent missense mutations in FOG2 in two patients with 46,XY gonadal dysgenesis. One patient carried a non-synonymous heterozygous mutation (p.S402R), while the other patient carried a heterozygous p.R260Q mutation and a homozygous p.M544I mutation. Functional studies indicated that the failure of testis development in these cases could be explained by the impaired ability of the mutant FOG2 proteins to interact with a known regulator of early testis development, GATA4. This is the first example of mutations in the coding sequence of FOG2 associated with 46,XY DSD in human and adds to the list of genes in the human known to be associated with DSD.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/pathology , GATA4 Transcription Factor/metabolism , Testis/abnormalities , Transcription Factors/genetics , Transcription Factors/metabolism , Exome , Female , Genetic Association Studies , HEK293 Cells , Heterozygote , Homozygote , Humans , Male , Models, Molecular , Mutation, Missense , Pedigree , Sequence Analysis, DNA , Testis/metabolism
11.
Am J Med Genet A ; 167(6): 1360-4, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25898814

ABSTRACT

The association of 46,XY disorder of sex development (DSD) with congenital diaphragmatic hernia (CDH) is rare, but has been previously described with and without other congenital anomalies. Literature review identified five cases of 46,XY DSD associated with CDH and other congenital anomalies. These five cases share characteristics including CDH, 46,XY karyotype with external female appearing or ambiguous genitalia, cardiac anomalies, and decreased life span. The present case had novel features including truncus arteriosus, bifid thymus, gut malrotation, and limb anomalies consisting of rhizomelia and adactyly. With this case report, we present a review of the literature of cases of 46,XY DSD and CDH in association with multiple congenital abnormalities. This case may represent a unique syndrome of 46,XY DSD and diaphragmatic hernia or a more severe presentation of a syndrome represented in the previously reported cases.


Subject(s)
Abnormalities, Multiple/genetics , Digestive System Abnormalities/genetics , Disorder of Sex Development, 46,XY/genetics , Hand Deformities, Congenital/genetics , Heart Defects, Congenital/genetics , Hernias, Diaphragmatic, Congenital/genetics , Intestinal Volvulus/genetics , Abnormalities, Multiple/pathology , Digestive System Abnormalities/pathology , Disorder of Sex Development, 46,XY/pathology , Facies , Fatal Outcome , Female , Hand Deformities, Congenital/pathology , Heart Defects, Congenital/pathology , Hernias, Diaphragmatic, Congenital/pathology , Humans , Infant , Infant, Newborn , Intestinal Volvulus/pathology , Male , Thymus Gland/metabolism , Thymus Gland/pathology , Truncus Arteriosus/metabolism , Truncus Arteriosus/pathology
12.
Theor Biol Med Model ; 12: 26, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26573569

ABSTRACT

BACKGROUND: Gonadal sex determination (GSD) in humans is a complex biological process that takes place in early stages of embryonic development when the bipotential gonadal primordium (BGP) differentiates towards testes or ovaries. This decision is directed by one of two distinct pathways embedded in a GSD network activated in a population of coelomic epithelial cells, the Sertoli progenitor cells (SPC) and the granulosa progenitor cells (GPC). In males, the pathway is activated when the Sex-Determining Region Y (SRY) gene starts to be expressed, whereas in females the WNT4/ ß-catenin pathway promotes the differentiation of the GPCs towards ovaries. The interactions and dynamics of the elements that constitute the GSD network are poorly understood, thus our group is interested in inferring the general architecture of this network as well as modeling the dynamic behavior of a set of genes associated to this process under wild-type and mutant conditions. METHODS: We reconstructed the regulatory network of GSD with a set of genes directly associated with the process of differentiation from SPC and GPC towards Sertoli and granulosa cells, respectively. These genes are experimentally well-characterized and the effects of their deficiency have been clinically reported. We modeled this GSD network as a synchronous Boolean network model (BNM) and characterized its attractors under wild-type and mutant conditions. RESULTS: Three attractors with a clear biological meaning were found; one of them corresponding to the currently known gene expression pattern of Sertoli cells, the second correlating to the granulosa cells and, the third resembling a disgenetic gonad. CONCLUSIONS: The BNM of GSD that we present summarizes the experimental data on the pathways for Sertoli and granulosa establishment and sheds light on the overall behavior of a population of cells that differentiate within the developing gonad. With this model we propose a set of regulatory interactions needed to activate either the SRY or the WNT4/ ß-catenin pathway as well as their downstream targets, which are critical for further sex differentiation. In addition, we observed a pattern of altered regulatory interactions and their dynamics that lead to some disorders of sex development (DSD).


Subject(s)
Models, Biological , Sex Determination Processes , Cell Differentiation , Cell Lineage , Disorder of Sex Development, 46,XY/pathology , Female , GATA4 Transcription Factor/metabolism , Gonads , Granulosa Cells/cytology , Humans , Male , Sertoli Cells/cytology
13.
Hum Hered ; 77(1-4): 108-17, 2014.
Article in English | MEDLINE | ID: mdl-25060274

ABSTRACT

Disorders of sex development (DSD) are defined as 'congenital conditions in which the development of chromosomal, gonadal, or anatomical sex is atypical' [Lee et al., Pediatrics 2006;118:e488-e500]. Studies conducted in Western countries, with low rates of consanguinity, show that truly ambiguous genitalia have an estimated incidence of 1:5,000 births. There are indications that the prevalence of DSD is higher in endogamous communities. The incidence of ambiguous genitalia in Saudi Arabia has been estimated at 1:2,500 live births; whilst in Egypt, it has been estimated at 1:3,000 live births. This may be due in part to an increase in disorders of androgen synthesis associated with 46,XX DSD. There is clearly a need for further studies to address the frequency of DSD in communities with high levels of consanguinity. This will be challenging, as an accurate diagnosis is difficult and expensive even in specialized centres. In developing countries with high levels of consanguinity, these limitations can be compounded by cultural, social and religious factors. Overall there is an indication that consanguinity may lead to an increase in incidences of both 46,XY and 46,XX DSD, and a co-ordinated study of populations with higher incidences of consanguinity/endogamy is needed to resolve this.


Subject(s)
Androgens/biosynthesis , Consanguinity , Disorders of Sex Development/genetics , Disorders of Sex Development/pathology , Adrenal Hyperplasia, Congenital/genetics , Adrenal Hyperplasia, Congenital/pathology , Cholesterol/biosynthesis , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/pathology , Female , Humans , Male , Testis/abnormalities , Testis/pathology
14.
Clin Endocrinol (Oxf) ; 80(2): 191-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23859637

ABSTRACT

OBJECTIVE: The steroidogenic acute regulatory protein (StAR) transports cholesterol to the mitochondria for steroidogenesis. Loss of StAR function causes lipoid congenital adrenal hyperplasia (LCAH) which is characterized by impaired synthesis of adrenal and gonadal steroids causing adrenal insufficiency, 46,XY disorder of sex development (DSD) and failure of pubertal development. Partial loss of StAR activity may cause adrenal insufficiency only. PATIENT: A newborn girl was admitted for mild dehydration, hyponatremia, hyperkalemia and hypoglycaemia and had normal external female genitalia without hyperpigmentation. Plasma cortisol, 17OH-progesterone, DHEA-S, androstendione and aldosterone were low, while ACTH and plasma renin activity were elevated, consistent with the diagnosis of primary adrenal insufficiency. Imaging showed normal adrenals, and cytogenetics revealed a 46,XX karyotype. She was treated with fluids, hydrocortisone and fludrocortisone. DESIGN, METHODS AND RESULTS: Genetic studies revealed a novel homozygous STAR mutation in the 3' acceptor splice site of intron 4, c.466-1G>A (IVS4-1G>A). To test whether this mutation would affect splicing, we performed a minigene experiment with a plasmid construct containing wild-type or mutant StAR gDNA of exons-introns 4-6 in COS-1 cells. The splicing was assessed on total RNA using RT-PCR for STAR cDNAs. The mutant STAR minigene skipped exon 5 completely and changed the reading frame. Thus, it is predicted to produce an aberrant and shorter protein (p.V156GfsX19). Computational analysis revealed that this mutant protein lacks wild-type exons 5-7 which are essential for StAR-cholesterol interaction. CONCLUSIONS: STAR c.466-1A skips exon 5 and causes a dramatic change in the C-terminal sequence of the protein, which is essential for StAR-cholesterol interaction. This splicing mutation is a loss-of-function mutation explaining the severe phenotype of our patient. Thus far, all reported splicing mutations of STAR cause a severe impairment of protein function and phenotype.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , Alternative Splicing/genetics , Disorder of Sex Development, 46,XY/genetics , Mutation , Phosphoproteins/genetics , Adrenal Hyperplasia, Congenital/pathology , Adrenal Insufficiency/diagnosis , Adrenal Insufficiency/genetics , Animals , Binding Sites/genetics , COS Cells , Chlorocebus aethiops , Cholesterol/chemistry , Cholesterol/metabolism , Disorder of Sex Development, 46,XY/pathology , Exons/genetics , Female , Humans , Infant, Newborn , Models, Molecular , Phenotype , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Binding , Protein Structure, Tertiary
15.
Reprod Biomed Online ; 29(4): 499-508, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25154015

ABSTRACT

Complex chromosomal rearrangements (CCR) represent rare structural chromosome abnormalities frequently associated with infertility. In this study, meiotic segregation in spermatozoa of an infertile normospermic carrier of a 4-breakpoint t(1;3;6) CCR was analysed. A newly developed array comparative genomic hybridization protocol was used, and all chromosomes in 50 single sperm cells were simultaneously examined. Three-colour FISH was used to analyse chromosome segregation in 1557 other single sperm cells. It was also used to measure an interchromosomal effect; sperm chromatin structure assay was used to measure chromatin integrity. A high-frequency of unbalanced spermatozoa (84%) was observed, mostly arising from the 3:3 symmetrical segregation mode. Array comparative genomic hybridization was used to detect additional aneuploidies in two out of 50 spermatozoa (4%) in chromosomes not involved in the complex chromosome rearrangement. Significantly increased rates of diploidy and XY disomy were found in the CCR carrier compared with the control group (P < 0.001). Defective condensation of sperm chromatin was also found in 22.7% of spermatozoa by sperm chromatin structure assay. The results indicate that the infertility in the man with CCR and normal spermatozoa was caused by a production of chromosomally unbalanced, XY disomic and diploid spermatozoa and spermatozoa with defective chromatin condensation.


Subject(s)
Chromosome Breakpoints , Chromosome Segregation , Disorder of Sex Development, 46,XY/diagnosis , Gene Rearrangement , Spermatozoa/pathology , Translocation, Genetic , Adult , Comparative Genomic Hybridization , Czech Republic , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/pathology , Disorder of Sex Development, 46,XY/physiopathology , Heterozygote , Humans , In Situ Hybridization, Fluorescence , Infertility, Male/etiology , Male , Meiotic Prophase I , Single-Cell Analysis
16.
Reprod Biomed Online ; 29(4): 490-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25131559

ABSTRACT

The effect of quadrivalent geometry on meiotic behaviour was evaluated. Segregation patterns of 404 cleavage stage embryos from 40 reciprocal translocation carriers undergoing 75 PGD cycles were analysed according to the asymmetric degree of quadrivalent. The percentage of alternate products with severe asymmetric quadrivalents was significantly lower than patients with mild asymmetric quadrivalents (22.5% versus 38.7%, P = 0.001). The incidence of 3:1 products was significantly higher in patients with severe compared with mild asymmetric quadrivalents (23.1% versus 12.2%, P = 0.004). The incidence of adjacent 1 (25.8% versus 24.3%), 2 (11.5% versus 12.6%) and 4:0/other segregation products (17.0% versus 12.2%) were not statistically significantly different between embryos from patients with severe or mild asymmetric quadrivalents. After adjusting for the confounder of sex using a logistic regression model, the odds of alternate embryos is about one-half for carriers classified as severe (OR 0.456, 95% CI 0.291 to 0.705), and the odds of 3:1 embryos is 2.2 times higher for carriers with severe asymmetric quadrivalents (OR 2.235, 95% CI 1.318 to 3.846). Our results suggest that the meiotic segregation pattern is related to the degree of asymmetry of specific quadrivalents. Severe asymmetric quadrivalents increases the risk of abnormal embryos.


Subject(s)
46, XX Disorders of Sex Development/diagnosis , Asymmetric Cell Division , Blastomeres/pathology , Chromosome Segregation , Disorder of Sex Development, 46,XY/diagnosis , Preimplantation Diagnosis , Translocation, Genetic , 46, XX Disorders of Sex Development/genetics , 46, XX Disorders of Sex Development/pathology , 46, XX Disorders of Sex Development/prevention & control , Adult , Blastomeres/cytology , China , Cohort Studies , Cryopreservation , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/pathology , Disorder of Sex Development, 46,XY/prevention & control , Ectogenesis , Embryo Transfer , Family Characteristics , Female , Heterozygote , Humans , Male , Sperm Injections, Intracytoplasmic , Vitrification
17.
Mol Genet Genomic Med ; 12(5): e2453, 2024 May.
Article in English | MEDLINE | ID: mdl-38769888

ABSTRACT

BACKGROUND: 46,XY sex reversal 11 (SRXY11) [OMIM#273250] is characterized by genital ambiguity that may range from mild male genital defects to gonadal sex reversal in severe cases. DHX37 is an RNA helicase that has recently been reported as a cause of SRXY11. So far, a total of 21 variants in DHX37 have been reported in 58 cases with 46,XY disorders of sex development (DSD). METHODS: Whole exome sequencing (WES) was conducted to screen for variations in patients with 46,XY DSD. The subcellular localization of mutant DHX37 proteins was detected by immunofluorescence. And the levels of mutant DHX37 proteins were detected via Western blotting. RESULTS: A novel pathogenic variant of DHX37 was identified in a patient with 46,XY DSD c.2012G > C (p.Arg671Thr). Bioinformatics analysis showed that the protein function of the variant was impaired. Compared with the structure of the wild-type DHX37 protein, the number of hydrogen bonds and interacting amino acids of the variant protein were changed to varying degrees. In vitro assays revealed that the variant had no significant effect on the intracellular localization of the protein but significantly reduced the expression level of the protein. CONCLUSIONS: Our finding further expands the spectrum of the DHX37 variant and could assist in the molecular diagnosis of 46,XY DSD patients.


Subject(s)
DEAD-box RNA Helicases , Disorder of Sex Development, 46,XY , Humans , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/pathology , Male , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , HEK293 Cells
18.
Andrology ; 12(1): 98-108, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37147882

ABSTRACT

PURPOSE: The etiology of 46, XY disorders of sex development (46, XY DSD) is complex, and studies have shown that different series of patients with 46, XY DSD has different genetic spectrum. In this study, we aimed to investigate the underlying genetic etiology in a Chinese series of patients with 46, XY DSD by whole exome sequencing (WES). METHODS: Seventy patients with 46, XY DSD were enrolled from the Peking Union Medical College Hospital (Beijing, China). The detailed clinical characteristics were evaluated, and peripheral blood was collected for WES to find the patients' rare variants (RVs) of genes related to 46, XY DSD. The clinical significance of the RVs was annotated according to American College of Medical Genetics and Genomics (ACMG) guidelines. RESULTS: A total of 57 RVs from nine genes were identified in 56 patients with 46, XY DSD, which include 21 novel RVs and 36 recurrent RVs. Based on the American ACMG guidelines, 43 variants were classified as pathogenic(P) or likely pathogenic (LP) variants and 14 variants were defined as variants of uncertain significance (VUS). P or LP variants were identified in 64.3% (45/70) patients of the series. Thirty-nine, 14, and 4 RVs were involved in the process of androgen synthesis and action, testicular determination and developmental process, and syndromic 46, XY DSD, respectively. The top three genes most frequently affected to cause 46, XY DSD were AR, SRD5A2, and NR5A1. Seven patients were found harboring RVs of the 46, XY DSD pathogenic genes identified in recent years, namely DHX37 in four patients, MYRF in two patients, and PPP2R3C in one patient. CONCLUSION: We identified 21 novel RVs of nine genes, which extended the genetic spectrum of 46, XY DSD pathogenic variants. Our study showed that 60% of the patients were caused by AR, SRD5A2 or NR5A1 P/LP variants. Therefore, polymerase chain reaction (PCR) amplification and Sanger sequencing of these three genes could be performed first to identify the pathogeny of the patients. For those patients whose pathogenic variants had not been found, whole-exome sequencing could be helpful in determining the etiology.


Subject(s)
Disorder of Sex Development, 46,XY , Humans , Male , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , China , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/pathology , Membrane Proteins/genetics , Mutation , Sexual Development , Testis/pathology , East Asian People/genetics , Steroidogenic Factor 1/genetics , Receptors, Antigen/genetics
19.
Chromosome Res ; 20(1): 215-38, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22290220

ABSTRACT

Disorders of sex development (DSD) are congenital conditions in which the development of chromosomal, gonadal, or anatomical sex is atypical. Many of the genes required for gonad development have been identified by analysis of DSD patients. However, the use of knockout and transgenic mouse strains have contributed enormously to the study of gonad gene function and interactions within the development network. Although the genetic basis of mammalian sex determination and differentiation has advanced considerably in recent years, a majority of 46,XY gonadal dysgenesis patients still cannot be provided with an accurate diagnosis. Some of these unexplained DSD cases may be due to mutations in novel DSD genes or genomic rearrangements affecting regulatory regions that lead to atypical gene expression. Here, we review our current knowledge of mammalian sex determination drawing on insights from human DSD patients and mouse models.


Subject(s)
Disorder of Sex Development, 46,XY/genetics , Gonads/pathology , Sex Determination Processes , 46, XX Testicular Disorders of Sex Development/genetics , 46, XX Testicular Disorders of Sex Development/pathology , Animals , Disorder of Sex Development, 46,XY/pathology , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gonads/growth & development , Humans , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Signaling System , Mice , Mice, Knockout , Mice, Transgenic , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Sex Chromosomes/genetics , Sex Chromosomes/metabolism , Sex Differentiation , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Wilms Tumor/genetics , Wilms Tumor/pathology
20.
Sex Dev ; 17(1): 8-15, 2023.
Article in English | MEDLINE | ID: mdl-36724755

ABSTRACT

INTRODUCTION: Steroid 5-alpha reductase deficiency (5α-R2D) is a rare condition caused by genetic variants that reduce the activity of the enzyme that converts testosterone into dihydrotestosterone. The clinical spectrum of 5α-R2D is known to overlap with other 46,XY differences of sex development (DSD) such as androgen insensitivity or gonadal dysgenesis. However, the clinical trajectories of the aetiologies can differ, with 5α-R2D presenting its own challenges. METHODS: In this study, we have collated clinical information for five individuals with variants in SRD5A2 identified using research genetic testing in an Australian paediatric setting. RESULTS: We describe how a genetic finding resolved or confirmed a diagnosis for these individuals and how it guided clinical management and family counselling. CONCLUSION: This work highlights the importance of early genetic testing in children born with 46,XY DSD where it complements traditional first-line testing.


Subject(s)
Disorder of Sex Development, 46,XY , Genetic Testing , Male , Humans , Child , Mutation , Australia , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/pathology , Testosterone , Membrane Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL