Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Hum Reprod ; 32(12): 2561-2573, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29121256

ABSTRACT

STUDY QUESTION: What is the prevalence of malignant testicular germ cell tumors (TGCT) and its precursors, (pre-) germ cell neoplasia in situ (GCNIS), in late teenagers and adults who have androgen insensitivity syndrome (AIS) and the impact of an individual's genetic susceptibility to development of TGCT? SUMMARY ANSWER: No GCNIS or TGCT was diagnosed, but pre-GCNIS was identified in 14 and 10% of complete and partial AIS patients, respectively, and was associated with a higher genetic susceptibility score (GSS), with special attention for KITLG (rs995030) and ATFZIP (rs2900333). WHAT IS KNOWN ALREADY: Many adult women with AIS decline prophylactic gonadectomy, while data regarding the incidence, pathophysiology and outcomes of TGCT in postpubertal individuals with AIS are lacking. The relevance of genetic factors, such as single nucleotide polymorphisms (SNPs), in predisposing AIS individuals to TGCT is unknown. STUDY DESIGN, SIZE, DURATION: This multicenter collaborative study on prophylactically removed gonadal tissue was conducted in a pathology lab specialized in germ cell tumor biology. PARTICIPANTS/MATERIALS, SETTING, METHODS: Material from 52 postpubertal individuals with molecularly confirmed AIS (97 gonadal samples) was included; the median age at surgery was 17.5 (14-54) years. Immunohistochemical studies and high-throughput profiling of 14 TGCT-associated SNPs were performed. The main outcome measures were the prevalence of pre-GCNIS, GCNIS and TGCT, and its correlation with a GSS, developed based on the results of recent genome-wide association studies. MAIN RESULTS AND ROLE OF CHANCE: The earliest recognizable change preceding GCNIS, referred to as pre-GCNIS, was present in 14% of individuals with complete and 10% of those with partial AIS at a median age of 16 years. No GCNIS or invasive TGCT were found. The median GSS was significantly greater for those with, compared to those without, pre-GCNIS (P = 0.01), with an overlap between groups. Our data suggest important roles for risk alleles G at KITLG (rs995030) and C at ATFZIP (rs2900333), among the 14 studied TGCT-associated SNPs. LARGE SCALE DATA: N/A. LIMITATIONS REASONS FOR CAUTION: A limited number of cases were included. WIDER IMPLICATIONS OF THE FINDINGS: Our data suggest that the prevalence of pre-GCNIS in individuals with AIS beyond puberty is around 15%. Genetic susceptibility likely contributes to pre-GCNIS development in AIS but factors related to malignant progression remain unclear. Although data in older patients remain scarce, malignant progression appears to be a rare event, although the natural history of the premalignant lesion remains unknown. Therefore, the practice of routine prophylactic gonadectomy in adults with AIS appears questionable and the patient's preference, after having been fully informed, should be decisive in this matter. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by research grants from the Research Foundation Flanders (FWO) (to M.C.), the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq G0D6713N) (to B.B.M. and M.C.) and the European Society for Pediatric Endocrinology (ESPE), granted by Novo Nordisk AB (to J.K.). There are no competing interests.


Subject(s)
Androgen-Insensitivity Syndrome/diagnosis , Androgen-Insensitivity Syndrome/genetics , Neoplasms, Germ Cell and Embryonal/diagnosis , Neoplasms, Germ Cell and Embryonal/genetics , Polymorphism, Single Nucleotide , Testicular Neoplasms/diagnosis , Testicular Neoplasms/genetics , Adolescent , Adult , Alleles , Androgen-Insensitivity Syndrome/complications , Androgen-Insensitivity Syndrome/epidemiology , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Male , Middle Aged , Neoplasms, Germ Cell and Embryonal/complications , Neoplasms, Germ Cell and Embryonal/epidemiology , Phenotype , Prevalence , Sexual Maturation , Stem Cell Factor/genetics , Testicular Neoplasms/complications , Testicular Neoplasms/epidemiology , Young Adult
2.
J Clin Endocrinol Metab ; 101(11): 4468-4477, 2016 11.
Article in English | MEDLINE | ID: mdl-27583472

ABSTRACT

CONTEXT: Only approximately 85% of patients with a clinical diagnosis complete androgen insensitivity syndrome and less than 30% with partial androgen insensitivity syndrome can be explained by inactivating mutations in the androgen receptor (AR) gene. OBJECTIVE: The objective of the study was to clarify this discrepancy by in vitro determination of AR transcriptional activity in individuals with disorders of sex development (DSD) and male controls. DESIGN: Quantification of DHT-dependent transcriptional induction of the AR target gene apolipoprotein D (APOD) in cultured genital fibroblasts (GFs) (APOD assay) and next-generation sequencing of the complete coding and noncoding AR locus. SETTING: The study was conducted at a university hospital endocrine research laboratory. PATIENTS: GFs from 169 individuals were studied encompassing control males (n = 68), molecular defined DSD other than androgen insensitivity syndrome (AIS; n = 18), AR mutation-positive AIS (n = 37), and previously undiagnosed DSD including patients with a clinical suspicion of AIS (n = 46). INTERVENTION(S): There were no interventions. MAIN OUTCOME MEASURE(S): DHT-dependent APOD expression in cultured GF and AR mutation status in 169 individuals was measured. RESULTS: The APOD assay clearly separated control individuals (healthy males and molecular defined DSD patients other than AIS) from genetically proven AIS (cutoff < 2.3-fold APOD-induction; 100% sensitivity, 93.3% specificity, P < .0001). Of 46 DSD individuals with no AR mutation, 17 (37%) fell below the cutoff, indicating disrupted androgen signaling. CONCLUSIONS: AR mutation-positive AIS can be reliably identified by the APOD assay. Its combination with next-generation sequencing of the AR locus uncovered an AR mutation-negative, new class of androgen resistance, which we propose to name AIS type II. Our data support the existence of cellular components outside the AR affecting androgen signaling during sexual differentiation with high clinical relevance.


Subject(s)
Androgen-Insensitivity Syndrome/diagnosis , Apolipoproteins D , Biological Assay/standards , Disorders of Sex Development/diagnosis , Receptors, Androgen/metabolism , Testosterone/analogs & derivatives , Adult , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/metabolism , Cells, Cultured , Disorders of Sex Development/genetics , Disorders of Sex Development/metabolism , Fibroblasts , High-Throughput Nucleotide Sequencing , Humans , Male , Mutation , Receptors, Androgen/genetics , Sensitivity and Specificity , Testosterone/metabolism , Transcription, Genetic
3.
Sex Dev ; 7(6): 282-8, 2013.
Article in English | MEDLINE | ID: mdl-24247294

ABSTRACT

A child born with ambiguous genitalia (Prader III) was found to have a 45,X[92.2%]/46,X,psu dic(Y)(p12)[7.8%] karyotype in peripheral blood lymphocytes. The testosterone level was consistent with that of a normal male; however, gonadotropins were elevated. Ultrasound and endoscopy of the urogenital sinus revealed well-developed Müllerian structures. At 3.5 months, the child was operated for right-sided incarcerated hernia, and the gonad situated at the inguinal region was biopsied and classified as primitive testis. Based on the presence of Müllerian structures, anatomy of external genitalia and wish of the parents, the child was assigned female gender. She underwent removal of the left gonad at 4 months during another acute surgery; histology was similar to the right gonad. The rest of the right gonad was removed at 16 months, and feminizing genitoplasty took place at 3 years. The right and left gonad contained 28 and 22% of cells with a Y chromosome, respectively. During further histological examination, dysgenetic features of the gonads were discovered. Some germ cells displayed abnormal development based on the specific expression of immunohistochemical markers (OCT3/4, TSPY, KITLG), indicating a possible risk for future malignant germ cell tumor development. Contribution of the 45,X cell line to the phenotype was also observed: the patient developed celiac disease, and her growth pattern resembled that of Turner syndrome responding to growth hormone treatment.


Subject(s)
Disorders of Sex Development/genetics , Gonadal Dysgenesis, Mixed/genetics , Gonads/pathology , Body Height , Body Weight , Celiac Disease/complications , Chromosomes, Human, Y/genetics , Disorders of Sex Development/pathology , Disorders of Sex Development/surgery , Female , Gonadal Dysgenesis, Mixed/pathology , Gonadal Dysgenesis, Mixed/surgery , Gonads/chemistry , Gonads/surgery , Human Growth Hormone/therapeutic use , Humans , Immunohistochemistry , Male , Mosaicism , Octamer Transcription Factor-3/analysis , Phenotype , Sex Chromosome Aberrations , Testis/pathology , Turner Syndrome/genetics , Uterus/pathology
4.
Sex Dev ; 7(5): 223-34, 2013.
Article in English | MEDLINE | ID: mdl-23774508

ABSTRACT

Androgen receptor (AR) mutations in androgen insensitivity syndrome (AIS) are associated with a variety of clinical phenotypes. The aim of the present study was to compare the molecular properties and potential pathogenic nature of 8 novel and 3 recurrent AR variants with a broad variety of functional assays. Eleven AR variants (p.Cys177Gly, p.Arg609Met, p.Asp691del, p.Leu701Phe, p.Leu723Phe, p.Ser741Tyr, p.Ala766Ser, p.Arg775Leu, p.Phe814Cys, p.Lys913X, p.Ile915Thr) were analyzed for hormone binding, transcriptional activation, cofactor binding, translocation to the nucleus, nuclear dynamics, and structural conformation. Ligand-binding domain variants with low to intermediate transcriptional activation displayed aberrant Kd values for hormone binding and decreased nuclear translocation. Transcriptional activation data, FxxFF-like peptide binding and DNA binding correlated well for all variants, except for p.Arg609Met, p.Leu723Phe and p.Arg775Leu, which displayed a relatively higher peptide binding activity. Variants p.Cys177Gly, p.Asp691del, p.Ala766Ser, p.Phe814Cys, and p.Ile915Thr had intermediate or wild type values in all assays and showed a predominantly nuclear localization in living cells. All transcriptionally inactive variants (p.Arg609Met, p.Leu701Phe, p.Ser741Tyr, p.Arg775Leu, p.Lys913X) were unable to bind to DNA and were associated with complete AIS. Three variants (p.Asp691del, p.Arg775Leu, p.Ile915Thr) still displayed significant functional activities in in vitro assays, although the clinical phenotype was associated with complete AIS. The data show that molecular phenotyping based on 5 different functional assays matched in most (70%) but not all cases.


Subject(s)
Androgen-Insensitivity Syndrome/genetics , Receptors, Androgen/genetics , Humans , Male , Mutation
5.
Sex Dev ; 6(6): 279-83, 2012.
Article in English | MEDLINE | ID: mdl-22797409

ABSTRACT

Persistent Müllerian duct syndrome (PMDS) is characterized by the presence of a uterus, fallopian tubes, and the upper part of the vagina in phenotypic normal male patients. Here, we report a patient diagnosed with PMDS with a novel homozygous missense mutation in the anti-Müllerian hormone (AMH) gene (single nucleotide insertion (C) at position 208 (c.208dup, p.Leu70fs)) leading to a frameshift and the introduction of a premature stop codon. Biopsy of both gonads revealed that germ cells were present in an irregular distribution. However, the absence of OCT3/4, PLAP and c-KIT expression indicated physiological maturation.


Subject(s)
Anti-Mullerian Hormone/genetics , Disorder of Sex Development, 46,XY/genetics , Mutation, Missense/genetics , Amino Acid Sequence , Anti-Mullerian Hormone/blood , Anti-Mullerian Hormone/chemistry , Base Sequence , DNA Mutational Analysis , Disorder of Sex Development, 46,XY/blood , Disorder of Sex Development, 46,XY/pathology , Homozygote , Humans , Infant, Newborn , Inhibins/blood , Male , Pedigree , Spermatogonia/pathology , Testis/pathology , Testosterone/blood
6.
J Clin Endocrinol Metab ; 97(9): 3107-14, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22723330

ABSTRACT

CONTEXT: High-dose estrogen treatment to reduce final height of tall girls increases their risk for infertility in later life. OBJECTIVE: The aim was to study the effect of estrogen dose on fertility outcome of these women. DESIGN/SETTING: We conducted a retrospective cohort study of university hospital patients. PATIENTS: We studied 125 tall women aged 20-42 yr, of whom 52 women had been treated with 100 µg and 43 with 200 µg of ethinyl estradiol (EE) in adolescence. MAIN OUTCOMES: Time to first pregnancy, treatment for infertility, and live birth rate were measured. RESULTS: The time to first pregnancy was increased in treated women. Of untreated women, 80% conceived within 1 yr vs. 69% of women treated with 100 µg EE and 59% of women treated with 200 µg EE. This trend of increased time to pregnancy with increasing estrogen dose was significant (log rank trend test, P = 0.01). Compared with untreated women, fecundability was reduced in women treated with both 100 µg EE [hazard ratio = 0.42; 95% confidence interval (CI), 0.19-0.95] and 200 µg EE (hazard ratio = 0.30; 95% CI, 0.13-0.72). We also observed a significant trend in the incidence of treatment for infertility with increased estrogen dose (P = 0.04). Fecundity was affected in women treated with 200 µg EE who had reduced odds of achieving at least one live birth (odds ratio = 0.13; 95% CI, 0.02-0.81), but not in women treated with 100 µg EE. CONCLUSIONS: We report a dose-response relationship between fertility in later life and estrogen dose used for the treatment of tall stature in adolescent girls; a higher estrogen dose is associated with increased infertility.


Subject(s)
Estrogens/pharmacology , Fertility/drug effects , Adolescent , Adult , Analysis of Variance , Body Height , Confidence Intervals , Dose-Response Relationship, Drug , Ethinyl Estradiol/pharmacology , Female , Humans , Infertility, Female/drug therapy , Infertility, Female/epidemiology , Kaplan-Meier Estimate , Live Birth , Pregnancy , Proportional Hazards Models , Young Adult
7.
Eur J Endocrinol ; 166(2): 341-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22080441

ABSTRACT

OBJECTIVE: Most patients with NR5A1 (SF-1) mutations and poor virilization at birth are sex-assigned female and receive early gonadectomy. Although studies in pituitary-specific Sf-1 knockout mice suggest hypogonadotropic hypogonadism, little is known about endocrine function at puberty and on germ cell tumor risk in patients with SF-1 mutations. This study reports on the natural course during puberty and on gonadal histology in two adolescents with SF-1 mutations and predominantly female phenotype at birth. DESIGN AND METHODS: Clinical and hormonal data and histopathological studies are reported in one male and one female adolescent with, respectively, a nonsense mutation (c.9T>A, p.Tyr3X) and a deletion of the first two coding exons (NCBI36/hg18 Chr9:g.(126306276-126307705)_(126303229-126302828)del) of NR5A1, both predicted to fully disrupt gene function. RESULTS: LH and testosterone concentrations were in the normal male range, virilization was disproportionate to the neonatal phenotype. In the girl, gonadectomy at 13 years revealed incomplete spermatogenesis and bilateral precursor lesions of testicular carcinoma in situ. In the boy, at the age of 12, numerous germ cells without signs of malignancy were present in bilateral testicular biopsy specimen. CONCLUSIONS: In SF-1 mutations, the neonatal phenotype poorly predicts virilization at puberty. Even in poorly virilized cases at birth, male gender assignment may allow spontaneous puberty without signs of hypogonadotropic hypogonadism, and possibly fertility. Patients with SF-1 mutations are at increased risk for malignant germ cell tumors. In case of preserved gonads, early orchidopexy and germ cell tumor screening is warranted. The finding of premalignant and/or malignant changes should prompt gonadectomy or possibly irradiation.


Subject(s)
Gonadal Dysgenesis, 46,XY/genetics , Gonads/pathology , Parturition/physiology , Steroidogenic Factor 1/genetics , Virilism/genetics , Virilism/pathology , Adolescent , Base Sequence , Female , Gonadal Dysgenesis, 46,XY/pathology , Humans , Infant, Newborn , Male , Mutation/physiology , Phenotype , Puberty/genetics , Puberty/physiology
8.
Singapore Med J ; 52(11): 810-3, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22173250

ABSTRACT

INTRODUCTION: Congenital adrenal hyperplasia (CAH) or adrenogenital syndrome is the most common cause of female ambiguous genitalia. Management of such patients involves medical treatment using glucocorticoids such as hydrocortisone, prednisone or dexamethasone. Monitoring is done by measurement of 17-hydroxyprogesterone (17-OHP) or androstenedione in serum, plasma or saliva. The aim of this study was to develop a system of monitoring steroid treatment in CAH patients using only saliva. METHODS: We studied the saliva of 24 CAH patients who received glucocorticoid replacement therapy. The patients were asked to collect saliva upon awakening, and in the afternoon and evening. The levels of 17-OHP and androstenedione in the saliva as well as in serum were then measured by immunoassay. RESULTS: There was a significant positive correlation between 17-OHP in serum and in saliva (R equals 0.929, p-value less than 0.01). A significant positive correlation between androstenedione level in saliva and serum was also found (R equals 0.611, p-value less than 0.01). This study also revealed a significant positive correlation between androstenedione and 17-OHP in serum (R equals 0.647, p-value less than 0.01) and saliva (R equals 0.799, p-value less than 0.01). All patients showed increased level of 17-OHP and androstenedione in the sample collected upon awakening. CONCLUSION: Determination of salivary androstenedione and 17-OHP in CAH patients could be a useful alternative to the measurement of these hormones in serum.


Subject(s)
17-alpha-Hydroxyprogesterone/metabolism , Adrenal Hyperplasia, Congenital/blood , Adrenal Hyperplasia, Congenital/metabolism , Androstenedione/biosynthesis , Androstenedione/blood , Disorders of Sex Development/blood , Adolescent , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Glucocorticoids/therapeutic use , Humans , Hydrocortisone/therapeutic use , Infant , Plasma/metabolism , Saliva/metabolism , Time Factors
9.
Horm Res Paediatr ; 76(5): 307-13, 2011.
Article in English | MEDLINE | ID: mdl-21921580

ABSTRACT

BACKGROUND/AIMS: Genetic variation in several candidate genes has been associated with short stature. Recently, a high-mobility group A2 (HMGA2) gene SNP has been robustly associated with height in the general population. Only few have attempted to study these genes in extremely tall stature. We therefore studied common genetic variation in candidate genes for height in extremely tall Dutch. METHODS: We included 116 constitutionally tall cases with height >2 SD and 103 controls with normally distributed height <2 SD. We genotyped 10 common polymorphisms previously associated with height variation. RESULTS: The HMGA2 gene SNP was significantly associated with tall stature. Using a logistic regression model, we calculated that carrying the HMGA2 (rs1042725) C allele significantly increased the odds of being tall (OR = 1.53, 95% CI 1.02-2.28; p = 0.03). In addition, controls with one or two copies of the C allele were significantly taller than controls carrying the TT genotype [TC: mean (SD) +0.61 (0.21) SDS; p = 0.004, and CC: +0.77 (0.25) SDS; p = 0.003]. CONCLUSION: Our study shows that a common polymorphism in the HMGA2 gene is not only associated with height variation in the general population but also plays an important role in one of the extremes of the height distribution.


Subject(s)
Body Height/genetics , Genetic Variation , Growth Disorders/genetics , HMGA2 Protein/genetics , Adolescent , Adult , Case-Control Studies , DNA Mutational Analysis , Female , Genetic Association Studies , HMGA2 Protein/metabolism , Humans , Male , Middle Aged , Netherlands , Young Adult
10.
Growth Horm IGF Res ; 21(6): 318-24, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21944866

ABSTRACT

CONTEXT/OBJECTIVE: The growth hormone (GH)/insulin-like growth factor-1(IGF-1) axis is the key regulator of somatic growth in humans and its genes are plausible candidates to study the genetics of height variation. Here, we studied polymorphic variation in the GH/IGF-1 axis in the extremely tall Dutch. METHODS: Case-control study of 166 tall cases with height >2 SDS and 206 controls with normally distributed height <2 SDS. Excluded were subjects with endocrine disorders or growth syndromes. We analyzed genomic DNA at 7 common polymorphisms in the GH-1, GH receptor (GHR), IGF-1 and IGFBP-3 genes. RESULTS: The association of the GH-1 1663 SNP with tall stature approached statistical significance, with the T-allele more present in the tall (allele frequency (AF): 0.44 vs. 0.36; p=0.084). Moreover, haplotype frequencies at this locus were significantly different between cases and controls, with the GGT haplotype most commonly seen in cases (p=0.01). Allele frequencies of GHR polymorphisms were not different. For the IGF-1 CA-repeat we observed a higher frequency of homozygous 192-bp carriers among tall males compared to control males (AF: 0.62 vs. 0.55; p=0.02). The IGFBP-3 -202 C-allele occurred more frequently in cases than in controls (AF: 0.58 vs. 0.50; p=0.002). Within cases, those carrying one or two copies of the -202 C-allele were significantly taller than AA genotype carriers (AC, p=0.028 and CC, p=0.009). Serum IGFBP-3 levels were highest in AA genotype carriers, the -202 SNP explained 5.8% of the variation. CONCLUSION: Polymorphic variation in the GH-1, IGF-1 and IGFBP-3 genes is associated with extremely tall stature. In particular, the IGFBP-3 -202 SNP is associated not only with being very tall but also with height variation within the tall.


Subject(s)
Body Height/genetics , DNA/genetics , Human Growth Hormone/genetics , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/genetics , Polymorphism, Single Nucleotide/genetics , Case-Control Studies , Child , Child, Preschool , DNA/blood , Female , Gene Frequency , Genotype , Human Growth Hormone/blood , Humans , Infant , Infant, Newborn , Insulin-Like Growth Factor Binding Protein 3/blood , Male , Polymerase Chain Reaction , Prognosis
11.
Sex Dev ; 5(4): 167-80, 2011.
Article in English | MEDLINE | ID: mdl-21791949

ABSTRACT

Malignant germ cell tumor (GCT) formation is a well-known complication in the management of patients with a disorder of sex development (DSD). DSDs are defined as congenital conditions in which development of chromosomal, gonadal, or anatomical sex is atypical. DSD patients in whom the karyotype - at least at the gonadal level - contains (a part of) the Y chromosome are at increased risk for neoplastic transformation of germ cells, leading to the development of the so-called 'type II germ cell tumors'. However, tumor risk in the various forms of DSD varies considerably between the different diagnostic groups. This contribution integrates our actual knowledge on the pathophysiology of tumor development in DSDs, recent findings on gonadal (mal)development in DSD patients, and possible correlations between the patient's phenotype and his/her risk for germ cell tumor development.


Subject(s)
Disorders of Sex Development , Neoplasms, Germ Cell and Embryonal , Ovary/embryology , Sex Determination Processes , Testis/embryology , Adolescent , Adult , Animals , Child , Child, Preschool , Chromosomes, Human, Y/genetics , Disorders of Sex Development/complications , Disorders of Sex Development/genetics , Female , Forkhead Box Protein L2 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Humans , Infant , Infant, Newborn , Male , Middle Aged , Neoplasms, Germ Cell and Embryonal/complications , Neoplasms, Germ Cell and Embryonal/genetics , Risk Factors , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/physiology , SOXB1 Transcription Factors , Sex Determination Processes/genetics , Sex Differentiation/genetics
12.
Sex Dev ; 5(4): 181-7, 2011.
Article in English | MEDLINE | ID: mdl-21654157

ABSTRACT

Steroidogenic factor 1 (SF1, officially NR5A1) is a nuclear receptor involved in adrenal and gonadal development. NR5A1 mutations have been identified in patients with various forms of 46,XY disorders of sex development (DSD), including complete gonadal dysgenesis with or without adrenal insufficiency, mild testicular dysgenesis with ambiguous external genitalia or female external genitalia with clitoromegaly, and penoscrotal hypospadias. We developed a synthetic probe set for MLPA analysis of the NR5A1 gene covering its 7 exons and analyzed 20 patients with 46,XY gonadal DSD in whom analyses failed to identify a genetic cause. We identified a partial NR5A1 deletion affecting exons 2 and 3, leading to NR5A1 haploinsufficiency in 1 patient presenting with female external genitalia with clitoromegaly, absence of a uterus, and mildly dysgenetic testes. This is the first partial NR5A1 gene deletion identified by MLPA in a patient with 46,XY gonadal DSD. This finding stresses the importance of investigating copy number changes, even at the exon level, in genes involved in gonadal DSD. As NR5A1 mutations can cause a wide spectrum of DSD with relatively high frequency, the analysis of the NR5A1 gene by MLPA is quite important and should be extended to larger groups of patients.


Subject(s)
Disorder of Sex Development, 46,XY/genetics , Gene Deletion , Molecular Probe Techniques , Steroidogenic Factor 1/genetics , Humans , Male , Molecular Probes , Polymerase Chain Reaction
13.
J Clin Endocrinol Metab ; 96(7): E1171-80, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21508138

ABSTRACT

CONTEXT: Gonadectomy is avoided whenever possible in boys with 45,X/46,XY. However, no clinical markers are currently available to guide clinicians in predicting gonadal tumor risk or hormone production. OBJECTIVE: The objective of the study was to test the hypothesis that gonadal histology and risk for development of a malignant germ cell tumor are reflected by the clinical presentation of a 45,X/46,XY individual. DESIGN: The design of the study was the correlation of clinical data [external masculinization score (EMS), pubertal outcome] with pathology data (gonadal phenotype, tumor risk). SETTING: This was a multicenter study involving two multidisciplinary disorder of sex development teams. PATIENTS: Patients included genetically proven 45,X/46,XY (and variants) cases, of whom at least one gonadal biopsy or gonadectomy specimen was available, together with clinical details. INTERVENTIONS: Patients (n = 48) were divided into three groups, based on the EMS. Gonadal histology and tumor risk were assessed on paraffin-embedded samples (n = 87) by morphology and immunohistochemistry on the basis of established criteria. MAIN OUTCOME MEASURES: Gonadal differentiation and tumor risk in the three clinical groups were measured. Clinical outcome in patients with at least one preserved gonad was also measured. RESULTS: Tumor risk in the three groups was significantly related to the gonadal differentiation pattern (P < 0.001). In boys, hormone production was sufficient and was not predicted by the EMS. CONCLUSIONS: The EMS reflects gonadal differentiation and tumor risk in patients with 45,X/46,XY. In boys, testosterone production is often sufficient, but strict follow-up is warranted because of malignancy risk, which appears inversely related to EMS. In girls, tumor risk is limited but gonads are not functional, making gonadectomy the most reasonable option.


Subject(s)
Genetic Predisposition to Disease , Gonadal Dysgenesis, 46,XY/genetics , Gonads/pathology , Mosaicism , Neoplasms/genetics , Sex Chromosome Aberrations , Turner Syndrome/genetics , Child , Child, Preschool , Female , Gonadal Dysgenesis, 46,XY/pathology , Humans , Male , Phenotype , Risk , Turner Syndrome/pathology
14.
J Clin Endocrinol Metab ; 96(4): 1098-105, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21289262

ABSTRACT

BACKGROUND/OBJECTIVE: High-dose estrogen treatment to reduce final height of tall girls has been shown to interfere with fertility. Ovarian function has not been studied. We therefore evaluated fertility and ovarian function in tall women who did or did not receive such treatment in adolescence. METHODS: This was a retrospective cohort study of 413 tall women aged 23-48 yr, of whom 239 women had been treated. A separate group of 126 fertile, normoovulatory volunteers aged 22-47 yr served as controls. RESULTS: Fertility was assessed in 285 tall women (157 treated, 128 untreated) who had attempted to conceive. After adjustment for age, treated women were at increased risk of experiencing subfertility [odds ratio (OR) 2.29, 95% confidence interval (CI) 1.38-3.81] and receiving infertility treatments (OR 3.44, 95% CI 1.76-6.73). Moreover, fecundity was notably affected because treated women had significantly reduced odds of achieving at least one live birth (OR 0.26, 95% CI 0.13-0.52). Remarkably, duration of treatment was correlated with time to pregnancy (r = 0.23, P = 0.008). Ovarian function was assessed in 174 tall women (119 treated, 55 untreated). Thirty-nine women (23%) exhibited a hypergonadotropic profile. After adjusting for age category, treated women had significantly higher odds of being diagnosed with imminent ovarian failure (OR 2.83, 95% CI 1.04-7.68). Serum FSH levels in these women were significantly increased, whereas antral follicle counts and serum anti-Müllerian hormone levels were decreased. CONCLUSION: High-dose estrogen-treated tall women are at risk of subfertility in later life. Their fecundity is significantly reduced. Treated women exhibit signs of accelerated ovarian aging with concomitant follicle pool depletion, which may be the basis of the observed subfertility.


Subject(s)
Estrogens/pharmacology , Fertility/drug effects , Growth Disorders/physiopathology , Ovary/drug effects , Adolescent , Adult , Body Height/drug effects , Body Height/physiology , Cohort Studies , Dose-Response Relationship, Drug , Estrogens/adverse effects , Estrogens/therapeutic use , Female , Fertility/physiology , Growth Disorders/complications , Growth Disorders/drug therapy , Humans , Infertility, Female/chemically induced , Infertility, Female/physiopathology , Middle Aged , Ovary/physiology , Pregnancy , Retrospective Studies , Young Adult
15.
J Clin Endocrinol Metab ; 95(12): 5233-40, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20826589

ABSTRACT

BACKGROUND/OBJECTIVE: Sex steroid treatment to reduce final height of tall boys has been available since the 1950s. In women, it has been shown to interfere with fertility. In men, no such data are available. We therefore evaluated fertility and gonadal function in tall men who did or did not receive high-dose androgen treatment in adolescence. METHODS: We conducted a retrospective cohort study of 116 tall men, of whom 60 had been treated. Reproductive and gonadal function was assessed by standardized interview, semen analysis, endocrine parameters, ultrasound imaging, and fatherhood. Mean age at treatment commencement was 14.2 yr, and mean follow-up was 21.2 yr. RESULTS: Sixty-six men (36 treated and 30 untreated) had attempted to achieve fatherhood. The probability of conceiving their first pregnancy within 1 yr was similar in treated and untreated men (26 vs. 24; Breslow P=0.8). Eleven treated and 13 untreated men presented with a left-sided varicocele (P=0.5). Testicular volume, sperm quality, and serum LH, FSH, and inhibin B levels were comparable between treated and untreated men. However, treated men had significantly reduced serum T levels, adjusted for known confounders [mean (sd) 13.3 (1.8) vs. 15.2 (1.9) nmol/liter; P=0.005). In addition, testicular volume and serum inhibin B and FSH levels in treated men were significantly correlated with age at treatment commencement. CONCLUSION: At a mean follow-up of 21 yr after high-dose androgen treatment, we conclude that fatherhood and semen quality in tall treated men are not affected. Serum testosterone levels, however, are reduced in androgen-treated men. Future research is required to determine whether declining testosterone levels may become clinically relevant for these men as they age.


Subject(s)
Androgens/therapeutic use , Body Height , Fathers , Fertility/physiology , Adolescent , Adult , Body Mass Index , Body Weight , Educational Status , Female , Follow-Up Studies , Humans , Male , Pregnancy , Registries , Retrospective Studies , Semen/physiology , Testosterone/blood
16.
Sex Dev ; 4(4-5): 259-69, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20558977

ABSTRACT

Certain patients with disorders of sex development (DSD), who bear Y chromosome material in their karyotype, are at increased risk for the development of type II germ cell tumors (GCT), which arise from early fetal germ cells. DSD gonads frequently harbor immature germ cells which express early fetal germ cell markers. Some of them (e.g. OCT3/4 and NANOG) seem to be of pathogenetic relevance in GCT development providing cells with the ability of pluripotency, proliferation and apoptosis suppression. Also TSPY (testis-specific protein Y-encoded), the main candidate for the so-called gonadoblastoma locus on Y chromosome, is overexpressed in germ cells of DSD patients and possibly contributes to their survival and proliferation. Nowadays, the use of immunohistochemical methods is highly relevant in identifying DSD gonads at risk. The risk for GCT development varies. While the prevalence of GCT is 15% in patients with partial androgen insensitivity, it may reach more than 30% in patients with gonadal dysgenesis. Patients with complete androgen insensitivity and ovotesticular DSD develop malignancies in 0.8% and 2.6% of cases, respectively. However, these data may be biased for various reasons. To better estimate the risk in individual groups of DSD, further investigations on large patient series are needed.


Subject(s)
Disorders of Sex Development/complications , Disorders of Sex Development/pathology , Ovarian Neoplasms/complications , Ovarian Neoplasms/pathology , Testicular Neoplasms/complications , Testicular Neoplasms/pathology , Female , Germ Cells/pathology , Humans , Male , Neoplasms, Germ Cell and Embryonal/etiology , Neoplasms, Germ Cell and Embryonal/pathology , Risk Factors
17.
Horm Res Paediatr ; 73(3): 223-30, 2010.
Article in English | MEDLINE | ID: mdl-20197677

ABSTRACT

BACKGROUND: Global accessibility and dissemination of developments in pediatric endocrinology prompted to examine how to develop an educational interactive e-SPE web portal. METHODS: A systematic approach was used to identify the relevant aspects of accessibility and dissemination. An orientation at the big idea was made, executed by an analysis of the needs of student and teacher pediatric endocrinologists, a definition of the learning objectives, a research in educational literature and an exploration of ICT design specifications. RESULTS: The intensive collaboration between medical, educational and information technology disciplines resulted in a portal design. The portal meets requirements of adult learning, stresses interaction between partners in learning and offers direct feedback during the learning process. The portal supports the development of not only knowledge but also competences both at junior and advanced levels. CONCLUSION: When the e-SPE portal is completed, the options for summative assessment will be examined as a medium for international certification in conjunction with local and national requirements (http://espe.elearning.nl).


Subject(s)
Education, Medical, Undergraduate/methods , Endocrinology/education , Internet , Pediatrics/education , Clinical Competence , Computer-Assisted Instruction , Education, Medical, Graduate/methods , Educational Measurement/methods , Feedback , Humans , Internship and Residency , Learning , Teaching
18.
Sex Dev ; 3(5): 237-44, 2009.
Article in English | MEDLINE | ID: mdl-19851057

ABSTRACT

Mutations in the androgen receptor (AR) gene, rendering the AR protein partially or completely inactive, cause androgen insensitivity syndrome, which is a form of a 46,XY disorder of sex development (DSD). We present 3 novel AR variants found in a cohort of Indonesian DSD patients: p.I603N, p.P671S, and p.Q738R. The aim of this study was to determine the possible pathogenic nature of these newly found unclassified variants. To investigate the effect of these variants on AR function, we studied their impact on transcription activation, AR ligand-binding domain interaction with an FxxLF motif containing peptide, AR subcellular localization, and AR nuclear dynamics and DNA-binding. AR-I603N had completely lost its transcriptional activity due to disturbed DNA-binding capacity and did not show the 114-kDa hyperphosphorylated AR protein band normally detectable after hormone binding. The patient with AR-I603N displays a partial androgen insensitivity syndrome phenotype, which is explained by somatic mosaicism. A strongly reduced transcriptional activity was observed for AR-Q738R, together with diminished interaction with an FxxLF motif containing peptide. AR-P671S also showed reduced transactivation ability, but no change in DNA- or FxxLF-binding capacity and interferes with transcriptional activity for as yet unclear reasons.


Subject(s)
Disorders of Sex Development/genetics , Mutation/genetics , Receptors, Androgen/genetics , Androgen-Insensitivity Syndrome/genetics , Child , Child, Preschool , Humans , Indonesia , Male
19.
J Pathol ; 216(1): 43-54, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18566970

ABSTRACT

Carcinoma in situ (CIS) of the testis is the pre-invasive stage of type II testicular germ cell tumours (TGCTs) of adolescents and adults. These tumours are the most frequently diagnosed cancer in Caucasian adolescents and young adults. In dysgenetic gonads, the precursor of type II GCTs can be either CIS or a lesion known as gonadoblastoma (GB). CIS/GB originates from a primordial germ cell (PGC)/gonocyte, ie an embryonic cell. CIS can be cured by local low-dose irradiation, with limited side effects on hormonal function. Therefore, strategies for early diagnosis of CIS are essential. Various markers are informative to diagnose CIS in adult testis by immunohistochemistry, including c-KIT, PLAP, AP-2gamma, NANOG, and POU5F1 (OCT3/4). OCT3/4 is the most informative and consistent in presence and expression level, resulting in intense nuclear staining. In the case of maturational delay of germ cells, frequently present in gonads of individuals at risk for type II (T)GCTs, use of these markers can result in overdiagnosis of malignant germ cells. This demonstrates the need for a more specific diagnostic marker to distinguish malignant germ cells from germ cells showing maturation delay. Here we report the novel finding that immunohistochemical detection of stem cell factor (SCF), the c-KIT ligand, is informative in this context. This was demonstrated in over 400 cases of normal (fetal, neonatal, infantile, and adult) and pathological gonads, as well as TGCT-derived cell lines, specifically in cases of CIS and GB. Both membrane-bound and soluble SCF were expressed, suggestive of an autocrine loop. SCF immunohistochemistry can be a valuable diagnostic tool, in addition to OCT3/4, to screen for precursor lesions of TGCTs, especially in patients with germ cell maturation delay.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma in Situ/diagnosis , Gonadoblastoma/diagnosis , Stem Cell Factor/analysis , Testicular Neoplasms/diagnosis , Adolescent , Adult , Biomarkers, Tumor/genetics , Gonadoblastoma/genetics , Gonadoblastoma/metabolism , Humans , Immunohistochemistry , Infant , Male , Reverse Transcriptase Polymerase Chain Reaction , Testicular Neoplasms/metabolism
20.
J Pathol ; 215(1): 31-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18348162

ABSTRACT

The transcription factors SOX9 and FOXL2 are required for male and female mammalian gonadal development. We have used specific antibodies to investigate the role of these key proteins in disorders of sex development (DSD), specifically inter-sex states. In normal gonads, SOX9 was found to be restricted to the presence of (pre-)Sertoli cells, while FOXL2 was found in granulosa cells, and in stromal cells interpreted as early ovarian stroma. Both proteins were found within a single patient, when testicular and ovarian development was present; and within the same gonad, when both differentiation lineages were identified, as in ovotesticular DSD (ie hermaphrodite). Especially SOX9 was informative to support the presence of early testicular development (ie seminiferous tubules), expected based on morphological criteria only. In a limited number of DSD cases, FOXL2 was found within reasonably well-developed seminiferous tubules, but double staining demonstrated that it was never strongly co-expressed with SOX9 in the same cell. All seminiferous tubules containing carcinoma in situ (CIS), the malignant counterpart of a primordial germ cell, ie the precursor of type II germ cell tumours of the testis, seminomas and non-seminomas, showed the presence of SOX9 and not FOXL2. In contrast, gonadoblastomas (GBs), the precursor of the same type of cancer, in a dysgenetic gonad, showed expression of FOXL2 and no, or only very low, SOX9 expression. These findings indicate that gonadal differentiation, ie testicular or ovarian, determines the morphology of the precursor of type II germ cell tumours, CIS or GB, respectively. We show that in DSD patients, the formation of either ovarian or/and testicular development can be visualized using FOXL2 and SOX9 expression, respectively. In addition, it initiates a novel way to study the role of the supportive cells in the development of either CIS or GB.


Subject(s)
Disorders of Sex Development/embryology , Forkhead Transcription Factors/analysis , Gene Expression Regulation, Developmental , Gonads/embryology , High Mobility Group Proteins/analysis , Transcription Factors/analysis , Adult , Biomarkers, Tumor/analysis , Carcinoma in Situ/chemistry , Female , Forkhead Box Protein L2 , Gonadoblastoma/chemistry , Gonadoblastoma/embryology , Gonads/chemistry , Humans , Immunohistochemistry , Male , Neoplasms, Germ Cell and Embryonal/chemistry , Neoplasms, Germ Cell and Embryonal/embryology , Ovary/chemistry , Ovary/embryology , SOX9 Transcription Factor , Testicular Neoplasms/chemistry , Testis/chemistry , Testis/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...