Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
1.
Clin Endocrinol (Oxf) ; 99(1): 58-63, 2023 07.
Article in English | MEDLINE | ID: mdl-36905105

ABSTRACT

OBJECTIVE: Individuals with 45,X/46,XY or 46,XY gonadal dysgenesis are at increased risk of germ cell malignancies. Therefore, prophylactic bilateral gonadectomy is advised in girls and considered in boys with atypical genitalia for undescended, macroscopically abnormal gonads. However, severely dysgenetic gonads may not contain germ cells rendering gonadectomy unnecessary. Therefore, we investigate if undetectable preoperative serum anti-Müllerian hormone (AMH) and inhibin B can predict the absence of germ cells, (pre)malignant or otherwise. DESIGN, PATIENTS AND MEASUREMENTS: Individuals who had undergone bilateral gonadal biopsy and/or gonadectomy because of suspected gonadal dysgenesis in 1999-2019 were included in this retrospective study if preoperative AMH and/or inhibin B were available. Histological material was reviewed by an experienced pathologist. Haematoxylin and eosin and immunohistochemical stainings for SOX9, OCT4, TSPY and SCF (KITL) were used. RESULTS: Thirteen males and 16 females were included, 20 with 46,XY and 9 with 45,X/46,XY DSD. Three females had dysgerminoma alongside gonadoblastoma; two gonadoblastoma, one germ cell neoplasia in situ (GCNIS) and three males had pre-GCNIS and/or pre-gonadoblastoma. Gonadoblastoma and/or dysgerminoma were present in 3/11 individuals with undetectable AMH and inhibin B, one of whom also had non-(pre)malignant germ cells. Of the other 18, in whom AMH and/or inhibin B were detectable, only one had no germ cells. CONCLUSIONS: Undetectable serum AMH and inhibin B cannot reliably predict the absence of germ cells and germ cell tumours in individuals with 45,X/46,XY or 46,XY gonadal dysgenesis. This information should help in counselling about prophylactic gonadectomy, taking into account both the germ cell cancer risk and potential for gonadal function.


Subject(s)
Dysgerminoma , Gonadal Dysgenesis, 46,XY , Gonadal Dysgenesis , Gonadoblastoma , Neoplasms, Germ Cell and Embryonal , Ovarian Neoplasms , Male , Female , Humans , Gonadoblastoma/genetics , Gonadoblastoma/surgery , Anti-Mullerian Hormone , Dysgerminoma/surgery , Retrospective Studies
2.
BMC Pregnancy Childbirth ; 23(1): 58, 2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36694125

ABSTRACT

BACKGROUND: Approximately 10-15% of 46,XY disorders of sex development (DSDs) have an SRY mutation residing in the high mobility group (HMG) domain. Here, we present a case of 46,XY DSD caused by a novel missense mutation in the HMG region of SRY rapidly progressing to germ cell tumors (GCTs). CASE PRESENTATION: An adolescent female (15 years old) exhibiting primary amenorrhea was later diagnosed as a 46,XY female with bilateral gonadal dysplasia on the basis of peripheral lymphocyte karyotype 46,XY and a novel missense mutation in SRY (c.281 T > G, p.L94R). The novel missense mutation (c.281 T > G, p.L94R) and its adjacent region were conserved. Protein structure analysis showed that the mutant site was located in the middle of the HMG domain, and the mutant protein had a diminished ability to bind to DNA. Imaging examination revealed an adolescent female with a naive uterus. Laparoscopy and initial pathological examination revealed left gonadal dysplasia and right gonadal dysplasia with gonadoblastoma (GB). Right gonadectomy by laparoscopy was performed upon consent from the patient's parents. Less than 1 year postoperatively, the left gonadal gland deteriorated as observed by the findings of a mass in the left adnexal region by pelvic MRI and serum AFP > 1000 ng/ml by serological tests, and then total hysterectomy and adnexal and left gonadectomy by laparoscopy were performed. The GCT stage was classified as stage Ic according to FIGO. At this time, pathologic examination showed that the left gonad had progressed to yolk sac tumor and dysgerminoma. The patient underwent chemotherapy post-operatively but developed type III myelosuppression and tumor recurrence several months later. CONCLUSIONS: The patient initially presented with right gonadoblastoma but chose only right gonadectomy by laparoscopy to preserve the female sex characteristics, which resulted in rapid deterioration of the left gonad and poor treatment outcomes. This case demonstrates the importance of early genetic diagnosis and treatment of 46,XY female DSD.


Subject(s)
Dysgerminoma , Endodermal Sinus Tumor , Gonadoblastoma , Ovarian Neoplasms , Sex-Determining Region Y Protein , Adolescent , Female , Humans , Dysgerminoma/diagnosis , Dysgerminoma/genetics , Dysgerminoma/surgery , Gonadoblastoma/genetics , Gonadoblastoma/surgery , Gonadoblastoma/pathology , Gonads/pathology , Gonads/surgery , Mutation, Missense , Neoplasm Recurrence, Local , Ovarian Neoplasms/complications , Ovarian Neoplasms/genetics , Ovarian Neoplasms/surgery
3.
Fetal Pediatr Pathol ; 42(1): 63-71, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35199613

ABSTRACT

Background: WT1 deletions are associated with nephroblastomas, WT mutations are associated with 46, XY sex reversal. It is unclear why only a few WT1 deletions are associated with sex reversal. Case report. This 46, XY female had a 15.2 MB interstitial deletion of 11p14.1p11.2, which included WT1 and FSHB. No pathogenic abnormalities were identified in 156 other genes associated with disorders of sexual development. Bilateral gonadoblastomas were incidentally diagnosed at 17 months of age at the time of prophylactic gonadectomies. She was treated without biopsy for bilateral nephroblastomas radiologically identified at 18 months of age. Bilateral partial nephrectomies contained treated intralobular nephrogenic rests. Conclusion: It is unclear why WT1 deletions are less associated with 46, XY sex reversal than WT1 mutations. Treating suspected nephroblastomas without biopsy, even in patients with syndromes associated with bilateral nephroblastomas, may still lead to diagnostic and therapeutic uncertainties.


Subject(s)
Gonadoblastoma , Kidney Neoplasms , Ovarian Neoplasms , Wilms Tumor , Humans , Female , Gonadoblastoma/genetics , Gonadoblastoma/pathology , Rest , Wilms Tumor/drug therapy , Wilms Tumor/genetics , Syndrome , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology
4.
J Pediatr Hematol Oncol ; 44(8): 471-473, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35700406

ABSTRACT

Frasier syndrome (FS) is a rare condition, caused by splice-site mutations of intron 9 in the Wilms' tumor suppressor gene 1 (WT1 gene). The WT1 protein is essential for urogenital development and patients with 46XY karyotype present with female (FS type 1) or male phenotype, gonadal dysgenesis, progressive glomerulopathy, and high risk of gonadoblastoma. We describe a female patient with an IVS9+4C>T donor splice-site mutation, who underwent a preventive gonadectomy at the age of 6 years due to imaging findings of dysplastic gonads. The biopsy revealed bilateral gonadoblastoma, emphasizing the need for early gonadectomy in 46XY FS patients.


Subject(s)
Gonadoblastoma , Ovarian Neoplasms , Male , Female , Humans , Frasier Syndrome/genetics , Frasier Syndrome/complications , Gonadoblastoma/genetics , Gonadoblastoma/pathology , Mutation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/surgery , Ovarian Neoplasms/complications , Castration/adverse effects
5.
Zhonghua Bing Li Xue Za Zhi ; 50(10): 1145-1150, 2021 Oct 08.
Article in Zh | MEDLINE | ID: mdl-34619868

ABSTRACT

Objective: To investigate the clinicopathological features of gonadal neoplastic related lesions in children with disorders of sexual development (DsD). Methods: The clinical manifestations, chromosomal karyotype, histology and immunophenotype of 12 cases of neoplastic related lesions from Guangzhou Women and Children's Medical Center, Guangzhou were analyzed during Jan 2015 to May 2020. Results: Twelve cases of neoplastic related lesions were screened in 205 cases of DsD, and 6 patients with gonadal germ cell neoplasia aged 3-13 years with an average age of 8.3 years. There were 2 males and 4 females. Clinical features showed malformation of external genitalia in 2 cases, short stature in 2 cases, clitoral enlargement in 1 case, lower abdominal pain and a huge pelvic mass in 1 case. Chromosomal karyotyping of peripheral blood showed 2 cases of 46XY and 4 cases of 45X/46XY. Fourteen gonadal specimens were examined. Microscopically, 1 case showed dysgerminoma in left ovary, and malignant mixed germ cell tumors in right ovary, as well as gonadoblastoma (GB) and undifferentiated gonadal tissue (UGT). The remaining 5 cases were all precursor lesions of germ cell tumor. Six specimens showed GB, 3 of UGT, and 3 specimens showed germ cell neoplasia in situ (GCNIS), one of which was accompanied by intratubular seminoma and 1 was GB with GCNIS. The other 6 patients with DsD were aged from 8 months to 2 years and 5 months, including 5 males and 1 females. Clinical manifestations showed 5 cases of hypospadias and 1 case of bilateral indirect inguinal hernia. Microscopically, 6 cases showed maturation delay of gonocytes in seminiferous tubules. Immunohistochemically, the primordial germ cells/gonocytes expressed OCT3/4, PLAP and c-KIT in the 12 cases. Conclusion: Gonadal neoplasia in children with DsD is mainly precursor lesions of germ cell tumor and improved understanding of these lesions is of great significance.


Subject(s)
Disorders of Sex Development , Gonadoblastoma , Neoplasms, Germ Cell and Embryonal , Ovarian Neoplasms , Testicular Neoplasms , Child , Female , Gonadoblastoma/genetics , Gonadoblastoma/surgery , Humans , Male
6.
Hum Mol Genet ; 26(5): 901-912, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28169398

ABSTRACT

The gonadoblastoma gene, testis-specific protein Y-encoded (TSPY), on the Y chromosome and its X-homologue, TSPX, are cell cycle regulators and function as a proto-oncogene and a tumor suppressor respectively in human oncogenesis. TSPY and TSPX competitively bind to the androgen receptor (AR) and AR variants, such as AR-V7, at their conserved SET/NAP domain, and exacerbate and repress the transactivation of the AR/AR-V7 target genes in ligand dependent and independent manners respectively. The inhibitory domain has been mapped to the carboxyl acidic domain of TSPX, truncation of which renders TSPX to be stimulatory while its transposition to the C-terminus of TSPY results in an inhibitory hybrid protein. TSPY and TSPX co-localize with the endogenous AR, in the presence of ligand, on the promoters and differentially regulate the expression of the endogenous AR target genes in the androgen-responsive LNCaP prostate cancer cells. Transcriptome analysis shows that TSPY and TSPX expressions differentially affect significant numbers of canonical pathways, upstream regulators and cellular functions. Significantly, among the common ones, TSPY activates and TSPX inhibits numerous growth-related and oncogenic canonical pathways and cellular functions in the respective cell populations. Hence, TSPY and TSPX exert opposing effects on the transactivation functions of AR and AR-Vs important for various physiological and disease processes sensitive to male sex hormone actions, thereby not only affecting the pathogenesis of male-specific prostate cancer but also likely contributing to sex differences in the health and diseases of man.


Subject(s)
Cell Cycle Proteins/genetics , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Transcriptome/genetics , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Chromosomes, Human, X/genetics , Chromosomes, Human, Y/genetics , DNA-Binding Proteins , Gene Expression Regulation, Developmental , Gonadoblastoma/genetics , Humans , Male , Nuclear Proteins/biosynthesis , Prostatic Neoplasms/pathology , Protein Domains/genetics , Proto-Oncogene Mas , Receptors, Androgen/biosynthesis , Testis/growth & development , Testis/pathology , Transcriptional Activation/genetics
7.
Hum Reprod ; 34(4): 770-779, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30753444

ABSTRACT

STUDY QUESTION: Which Y genes mapped to the 'Gonadoblastoma Y (GBY)' locus on human Y chromosome are expressed in germ cells of individuals with some Differences of Sexual Development (DSD) and a Y chromosome in their karyotype (DSD-XY groups)? SUMMARY ANSWER: The GBY candidate genes DDX3Y and TSPY are expressed in the germ cells of DSD-XY patients from distinct etiologies: patients with mixed gonadal dysgenesis (MGD) and sex chromosome mosaics (45,X0/46,XY; 46,XX/46,XY); patients with complete androgen insensitivity (CAIS), patients with complete gonadal dysgenesis (CGD; e.g. Swyer syndrome). WHAT IS KNOWN ALREADY: A GBY locus was proposed to be present on the human Y chromosome because only DSD patients with a Y chromosome in their karyotype have a high-although variable-risk (up to 55%) for germ cell tumour development. GBY was mapped to the proximal part of the short and long Y arm. TSPY located in the proximal part of the short Y arm (Yp11.1) was found to be a strong GBY candidate gene. It is expressed in the germ cells of DSD-XY patients with distinct etiologies but also in foetal and pre-meiotic male spermatogonia. However, the GBY region extends to proximal Yq11 and therefore includes probably more than one candidate gene. STUDY DESIGN, SIZE, DURATION: Protein expression of the putative GBY candidate gene in proximal Yq11, DDX3Y, is compared with that of TSPY in serial gonadal tissue sections of 40 DSD-XY individuals from the three DSD patient groups (MGD, Complete Androgen Insensitivity Syndrome [CAIS], CGD) with and without displaying malignancy. Expression of OCT3/4 in the same tissue samples marks the rate of pluripotent germ cells. PARTICIPANTS/MATERIALS, SETTING, METHOD: A total of 145 DSD individuals were analysed for the Y chromosome to select the DSD-XY subgroup. PCR multiplex assays with Y gene specific marker set score for putative microdeletions in GBY Locus. Immunohistochemical experiments with specific antisera mark expression of the GBY candidate proteins, DDX3Y, TSPY, in serial sections of the gonadal tissue samples; OCT3/4 expression analyses in parallel reveal the pluripotent germ cell fraction. MAIN RESULTS AND THE ROLE OF CHANCE: Similar DDX3Y and TSPY protein expression patterns were found in the germ cells of DSD-XY patients from each subgroup, independent of age. In CAIS patients OCT3/4 expression was often found only in a fraction of these germ cells. This suggest that GBY candidate proteins are also expressed in the non-malignant germ cells of DSD-XY individuals like in male spermatogonia. LIMITATIONS, REASONS FOR CAUTION: Variation of the expression profiles of GBY candidate genes in the germ cells of some DSD-XY individuals suggests distinct transcriptional and translational control mechanisms which are functioning during expression of these Y genes in the DSD-XY germ cells. Their proposed GBY tumour susceptibility function to transform these germ cells to pre-malignant GB/Germ Cell Neoplasia in Situ (GB/GCNIS) cells seems therefore to be limited and depending on their state of pluripotency. WIDER IMPLICATIONS OF THE FINDINGS: These experimental findings are of general importance for each individual identified in the clinic with DSD and a Y chromosome in the karyotype. To judge their risk of germ cell tumour development, OCT3/4 expression analyses on their gonadal tissue section is mandatory to reveal the fraction of germ cells still being pluripotent. Comparative expression analysis of the GBY candidate genes can be helpful to reveal the fraction of germ cells with genetically still activated Y chromosomes contributing to further development of malignancy if at high expression level. STUDY FUNDING/COMPETING INTEREST(S): This research project was supported by a grant (01GM0627) from the BMBF (Bundesministerium für Bildung und Forschung), Germany to P.H.V. and B.B. The authors have no competing interests.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomes, Human, Y/metabolism , DEAD-box RNA Helicases/metabolism , Genetic Loci , Germ Cells/metabolism , Gonadoblastoma/genetics , Karyotype , Minor Histocompatibility Antigens/metabolism , Ovarian Neoplasms/genetics , Testicular Neoplasms/genetics , Adolescent , Adult , Biopsy , Cell Cycle Proteins/genetics , Child , Child, Preschool , DEAD-box RNA Helicases/genetics , Female , Gene Expression Regulation, Neoplastic , Gonadoblastoma/blood , Gonadoblastoma/pathology , Gonads/pathology , Humans , Infant , Male , Minor Histocompatibility Antigens/genetics , Ovarian Neoplasms/blood , Ovarian Neoplasms/pathology , Testicular Neoplasms/blood , Testicular Neoplasms/pathology , Young Adult
8.
Int J Gynecol Pathol ; 38(6): 581-587, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30252730

ABSTRACT

Herein, we report a case of a 9-yr-old girl who had a 46, XX peripheral karyotype and apparent developmentally normal ovaries. She presented with abdominal pain and a right adnexal mass. No clinical or pathologic evidence of gonadal dysgenesis or undifferentiated gonadal tissue was detected. She underwent right salpingo-oophorectomy with rupture of the tumor at the time of operation due to recent adnexal torsion. The original pathologic diagnosis was gonadoblastoma and mixed germ cell tumor. Most significantly in our study, we identified a rare and novel pathway for the development of malignant mixed germ cell tumor from gonadoblastoma in the absence of identifiable dysgerminoma. The histologically identifiable steps of progression in our case were as follows: (1) residual islands of classic gonadoblastoma, (2) overgrowth by "dissecting" gonadoblastoma composed of transformed germ cells with clear cytoplasm and sex cord elements surrounded by a basement membrane, (3) stromal infiltration by dedifferentiated germ cells with loss of basement membrane, (4) formation of malignant mixed germ cell tumor. The dedifferentiated areas were composed of anaplastic germ cells with amphophilic cytoplasm that gradually replaced the sex cord elements by clonal expansion. Both the original transformed and the anaplastic germ cell components strongly expressed OCT4. We believe that the mixed germ cell tumor arose from the dedifferentiated germ cell component through neoplastic progression. This premise suggests that the germ cell component of "dissecting" gonadoblastoma rarely undergoes anaplastic change in the absence of transition to germinoma and can be the direct precursor of mixed germ cell tumor.


Subject(s)
Gonadoblastoma/diagnosis , Neoplasms, Germ Cell and Embryonal/diagnosis , Ovarian Neoplasms/diagnosis , Child , Female , Gonadoblastoma/genetics , Gonadoblastoma/pathology , Humans , Karyotype , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phenotype , Sexual Development/genetics
9.
Mol Biol Rep ; 46(3): 2971-2978, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30879272

ABSTRACT

Two sisters phenotypically normal females, presenting with tumor abdominal mass with histopathological findings of teratoma and gonadoblastoma associated to 46,XY male-to-female sex reversal syndrome, secondary to a duplication in DAX-1, possibly inherited of maternal gonadal mosaicism. Copy number variation and functional effects of the duplication were done by MLPA multiplex ligation-dependent probe amplification and real time PCR. DAX-1, also known as dosage sensitive sex reversal gene (DSS), is considered the most likely candidate gene involved in XY gonadal dysgenesis when overexpressed. The excess of DAX-1 gene disturbs testicular development by down regulation of SF-1, WT1, and SOX9. This is the first report of 46,XY sex reversal in two siblings who have a maternally inherited duplication of DAX-1 associated with reduced levels of expression of downstream genes as SOX9-SF1.


Subject(s)
DAX-1 Orphan Nuclear Receptor/genetics , Gonadal Dysgenesis/genetics , Sex Determination Processes/genetics , Adolescent , Child , DAX-1 Orphan Nuclear Receptor/metabolism , DNA Copy Number Variations , DNA-Binding Proteins/genetics , Female , Gene Dosage/genetics , Gene Duplication , Gonadal Dysgenesis, 46,XY/genetics , Gonadoblastoma/genetics , Humans , Pedigree , Sex Determination Analysis/methods , Sex Differentiation , Sexual Maturation/genetics , Siblings , Teratoma , Testis/abnormalities
10.
Pediatr Dev Pathol ; 22(4): 380-385, 2019.
Article in English | MEDLINE | ID: mdl-30646821

ABSTRACT

45,X/46,XY mosaicism is one of a heterogenous group of congenital conditions known as differences (disorders) of sex development (DSD) that results in abnormal development of internal and external genitalia. Patients with DSD, particularly those with segments of the Y chromosome, are at increased risk for germ cell tumors including gonadoblastoma. Gonadoblastoma is a neoplasm comprised of a mixture of germ cells and elements resembling immature granulosa or Sertoli cells with or without Leydig cells or lutein-type cells in an ovarian type stroma. Gonadoblastoma has an increased prevalence of 15% to 40% in patients with 45,X/46,XY mosaicism and has been previously reported in patients as young as 5 months of age with that karyotype. Herein, we describe a 3-month-old child with 45,X/46,XY karyotype who was referred for the evaluation of asymmetric labia majora. Additional evaluation revealed left streak gonad and right dysplastic/dysgenetic testis. Both gonads contained foci of cells typical for gonadoblastoma as well as undifferentiated gonadal tissue, underscoring the potential for very early infantile gonadoblastoma and the spectrum of developmental anomalies associated with this karyotype.


Subject(s)
Disorders of Sex Development/diagnosis , Gonadal Dysgenesis, Mixed/diagnosis , Gonadoblastoma/diagnosis , Disorders of Sex Development/genetics , Disorders of Sex Development/pathology , Female , Gonadal Dysgenesis, Mixed/genetics , Gonadal Dysgenesis, Mixed/pathology , Gonadoblastoma/genetics , Gonadoblastoma/pathology , Humans , Infant, Newborn , Karyotype , Male , Mosaicism
11.
J Formos Med Assoc ; 118(1 Pt 3): 450-456, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30017534

ABSTRACT

BACKGROUND/PURPOSE: 45,X/46,XY mosaicism is a rare sex chromosome abnormality. Here, we present our experience in the management of 45,X/46,XY Taiwanese children. PATIENTS AND METHODS: We enrolled 19 patients from January 1981 to September 2016. The diagnosis of 45,X/46,XY mosaicism was made by karyotyping peripheral blood lymphocytes. All medical records were thoroughly reviewed. RESULTS: Of the 19 patients, 16 were reared as females and 3 as males. The age at diagnosis ranged from 1 month to 15 years and 9 months. Atypical genitalia, short stature, and Turner stigmata were common manifestations. No patient exhibited a cardiac malformation but 29% had renal malformations and 12.5% had autoimmune thyroid disease who developed thyroid dysfunction later. Nine girls with short stature received growth hormone therapy and their height standard deviation score rose from -3.4 ± 1.1 to -1.4 ± 0.9 in adulthood (P < 0.01). The gonadal phenotypes included bilateral streak gonads in nine patients, a streak gonad with contralateral gonadal agenesis in one, mixed gonadal dysgenesis in five, bilateral dysgenetic testes in two, and bilateral gonadoblastomas in one. CONCLUSION: The 45,X/46,XY phenotype varies widely and a high index of suspicion is important to ensure early diagnosis. Cardiac and renal malformations should be screened ultrasonically at diagnosis and thyroid status should be monitored annually. Growth hormone effectively improves adult height in short girls. Prophylactic gonadectomy is indicated for those with intra-abdominal streaks or dysgenetic gonads to prevent the development of a malignancy.


Subject(s)
Disorders of Sex Development/drug therapy , Disorders of Sex Development/genetics , Growth Hormone/therapeutic use , Mosaicism , Adolescent , Body Height/genetics , Child , Child, Preschool , Female , Gonadal Dysgenesis, Mixed/genetics , Gonadoblastoma/genetics , Humans , Infant , Karyotyping , Male , Taiwan , Turner Syndrome/genetics
12.
Pediatr Nephrol ; 32(1): 81-89, 2017 01.
Article in English | MEDLINE | ID: mdl-27300205

ABSTRACT

BACKGROUND: WT1 is one of the genes commonly reported as mutated in children with steroid-resistant nephrotic syndrome (SRNS). We analyzed genotype-phenotype correlations in pediatric SRNS patients with WT1 mutations. METHODS: From 2001 to 2015, WT1 mutations were detected in 21 out of 354 children with SRNS by genetic screening (5.9 %). The patients were grouped into missense (n = 11) and KTS splicing (n = 10) mutation groups. RESULTS: Nine (82 %) patients with missense mutations presented with congenital/infantile nephrotic syndrome, while 8 (80 %) with KTS splicing mutations presented with childhood-onset SRNS. Progression to end-stage renal disease (ESRD) was noted in all patients with missense mutations (median age, 2.6 months; interquartile range [IQR], 0.8 months to 1.7 years) and in 5 patients with KTS splicing mutations (median, 9.3 years; IQR, 3.3-16.5 years). Disorders of sexual development (DSDs) were noted in all 12 patients with a 46, XY karyotype and in only 1 of the 8 patients with a 46, XX karyotype. One patient developed a Wilms tumor and another developed gonadoblastoma. Three patients had a diaphragmatic defect or hernia. CONCLUSIONS: WT1 mutations manifest as a wide spectrum of renal and extra-renal phenotypes. Genetic diagnosis is essential for overall management and to predict the genotype-specific risk of DSDs and the development of malignancies.


Subject(s)
Glomerulonephritis/genetics , Nephrotic Syndrome/genetics , WT1 Proteins/genetics , Adolescent , Child , Child, Preschool , Disorders of Sex Development/genetics , Disorders of Sex Development/pathology , Drug Resistance , Female , Genotype , Glomerulonephritis/pathology , Gonadoblastoma/genetics , Gonadoblastoma/pathology , Humans , Infant , Infant, Newborn , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/pathology , Male , Mutation , Mutation, Missense , Nephrotic Syndrome/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phenotype , Republic of Korea , Survival Analysis , Treatment Outcome , Wilms Tumor/genetics , Wilms Tumor/pathology
13.
Pol J Pathol ; 67(4): 411-414, 2016.
Article in English | MEDLINE | ID: mdl-28547971

ABSTRACT

We present a case of a woman with primary amenorrhea. Ultrasound imaging showed a uterus of normal size but bands of connective tissues at the site of ovaries. A genetic test was done which revealed the XY karyotype. Swyer syndrome was diagnosed. The patient did not report for the follow-up visits. Three years later, the woman reported back because of increasing abdominal circumference. The patient underwent an operation. Radical hysterectomy was performed. Histopathological examination showed dysgerminoma and gonadoblastoma on the left gonad and dysgerminoma on the right one. This case report presents the natural history of Swyer syndrome.


Subject(s)
Dysgerminoma/pathology , Gonadal Dysgenesis, 46,XY/complications , Gonadoblastoma/pathology , Gonads/pathology , Adolescent , Dysgerminoma/genetics , Female , Gonadoblastoma/genetics , Humans
14.
Yi Chuan ; 37(11): 1105-15, 2015 11.
Article in Zh | MEDLINE | ID: mdl-26582524

ABSTRACT

Gonadoblastoma (GB), a rare in situ germ cell tumor derived from sex cord and germ cells, is closely associated with gonadal dysgenesis. About 80% of GB individuals exhibit 46, XY female phenotype while the others are 45, XY and 46, XX with disorders of sex development. Moreover, 35% of GB can eventually develop into malignant tumors, such as seminoma and dysgerminoma tumors. The molecular genetic mechanism of GB remains to be fully uncovered due to phenotypic and genetic heterogeneity. Increasing studies show that the formation of GB is closely related to genes regulating sexual differentiation and determination (e.g., SRY, WT1, SOX9, Foxl2, TSPY, etc), and is affected by the interaction of genetic and epigenetic regulation. Here we describe the clinical and pathological features, diagnosis and treatment of GB, and also summarize the molecular genetic and epigenetic mechanisms underlying the gonadal abnormalities that lead to GB. We analyze and construct the common gene regulatory networks related to the development of GB, and describe some obstacles and deficiencies in current studies to provide innovative perspectives on further studying the pathological and molecular mechanisms of GB.


Subject(s)
Gonadoblastoma/genetics , Epigenesis, Genetic , Female , Gene Regulatory Networks , Genes, Wilms Tumor , Humans , Male , Mutation , Sex-Determining Region Y Protein/genetics
15.
Biochem Biophys Res Commun ; 446(1): 206-11, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24583132

ABSTRACT

The testis-specific protein Y-encoded (TSPY) is a repetitive gene located on the gonadoblastoma region of the Y chromosome, and has been considered to be the putative gene for this oncogenic locus on the male-only chromosome. It is expressed in spermatogonial cells and spermatocytes in normal human testis, but abundantly in gonadoblastoma, testicular germ cell tumors and a variety of somatic cancers, including melanoma, hepatocellular carcinoma and prostate cancer. Various studies suggest that TSPY accelerates cell proliferation and growth, and promotes tumorigenesis. In this report, we show that TSPY could bind directly to the chromatin/DNA at exon 1 of its own gene, and greatly enhance the transcriptional activities of the endogenous gene in the LNCaP prostate cancer cells. Domain mapping analyses of TSPY have localized the critical and sufficient domain to the SET/NAP-domain. These results suggest that TSPY could efficiently amplify its expression and oncogenic functions through a positive feedback loop, and contribute to the overall tumorigenic processes when it is expressed in various human cancers.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomes, Human, Y/genetics , Prostatic Neoplasms/genetics , Binding Sites/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Exons , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Gonadoblastoma/genetics , Gonadoblastoma/metabolism , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Interaction Mapping , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Up-Regulation
16.
Hum Reprod ; 29(7): 1413-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24826988

ABSTRACT

STUDY QUESTION: What is the optimal protocol of management for phenotypic female patients with Y chromosome or Y-derived sequences, in particular for adult patients? SUMMARY ANSWER: Immediate gonadectomy, long-term hormone therapy and psychological care are suggested to be the optimal management for older phenotypic female patients with Y chromosome or Y-derived sequences. WHAT IS KNOWN ALREADY: Phenotypic female patients with Y chromosome or Y-derived sequences are at increasing risk of developing gonadal tumors with age. Early diagnosis and safe guidelines of management for these patients are needed. STUDY DESIGN, SIZE, DURATION: One hundred and two phenotypic women with Y chromosome or Y-derived sequences were included in a straightforward, retrospective-observational study conducted over a period of 26 years from January 1985 to November 2010. PARTICIPANTS/MATERIALS, SETTING AND METHODS: Patients aged 16-34 years presenting to our Academic Department of Gynecology with symptoms of disorders of sex development were subjected to history taking, hormonal evaluation, conventional cytogenetic analysis, PCR, histopathology and immunohistochemistry. Features of the gonads were examined and the outcome of prophylactic gonadectomy evaluated. MAIN RESULTS AND THE ROLE OF CHANCE: Among the patients recruited in our study, 48 patients (47.1%) were diagnosed with complete/partial androgen insensitivity syndrome (CAIS/PAIS) (46XY), 33 cases (32.4%) with gonadal dysgenesis (46XY) and the remaining subjects (20.1%) with mixed gonadal dysgenesis (with sex chromosome structural abnormalities). The total incidence of malignancy was 17.6%. Seventeen patients (16.7%) had gonadoblastoma, while one patient (1.0%) with gonadal dysgenesis had dysgerminoma. Gonadoblastoma were observed in 2/21 patients with sex chromosome structural abnormalities (9.5%), 3/33 patients with gonadal dysgenesis (9.1%), 9/30 patients with CAIS (30.0%) and 3/18 patients with PAIS (16.7%). LIMITATIONS, REASONS FOR CAUTION: Selection bias in this cohort study may affect data interpretation due to the low incidence of disorders of sex development in the general population. WIDER IMPLICATIONS OF THE FINDINGS: The risk for malignant transformation may occur in early life and highly increase with age in patients with Y chromosome or Y-derived sequences. Optimal timing of gonadectomy should be decided by multiple factors including the subgroup of disorder, age and degree of patient's maturity. In addition, gonadal biopsy is suggested when the disease is diagnosed and any evidence of premalignancy warranties gonadectomy. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the National Key Scientific Research Project (2013CB967404), Natural Science Funds of Zhejiang Province (Y13H04005), Zhejiang Qianjiang talent plan (2013R10027), the Fundamental Research Funds for the Central Universities and Key Projects in the National Science & Technology Pillar Program during the Eleventh Five-Year Plan Period (2012BAI32B04). The authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER None.


Subject(s)
Chromosomes, Human, Y/ultrastructure , Gonadal Disorders/genetics , Gonadoblastoma/genetics , Adolescent , Adult , Androgen-Insensitivity Syndrome/diagnosis , Androgen-Insensitivity Syndrome/genetics , Chromosome Aberrations , Cytogenetics , Female , Genitalia/pathology , Gonadal Disorders/diagnosis , Gonadal Disorders/surgery , Gonadal Dysgenesis/diagnosis , Gonadal Dysgenesis/genetics , Gonadoblastoma/diagnosis , Gonadoblastoma/surgery , Humans , Immunohistochemistry , Male , Phenotype , Retrospective Studies , Risk , Sex Factors , Young Adult
17.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 31(5): 600-3, 2014 Oct.
Article in Zh | MEDLINE | ID: mdl-25297590

ABSTRACT

TSPY1 (testis-specific protein, Y-linked 1) gene family, located in male-specific region of Y-chromosome (MSY), has the maximum number of copies organized as a long tandem repeat array in protein-coding gene families of human genome. TSPY1 is identified to be the most important candidate gene for gonadoblastoma, and its coding protein can promote the proliferation and differentiation of tumor cells. Recently, TSPY1 gene family is also proposed to play an important role in spermatogenesis. In this review, the structure characteristics of the gene family were illustrated, and the functional studies of TSPY1 in the process of tumorigenesis and spermatogenesis were discussed.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomes, Human, Y/genetics , Genetic Predisposition to Disease/genetics , Gonadoblastoma/genetics , Testicular Neoplasms/genetics , Gene Dosage , Humans , Male , Spermatogenesis/genetics
18.
Medicine (Baltimore) ; 103(25): e38297, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905377

ABSTRACT

BACKGROUND: Disorders of sex development (DSD) are congenital conditions characterized by atypical development of chromosomal, gonadal, and phenotypic sex. 46, XY DSD can result from disorders of testicular development or androgen synthesis. METHODS: We present 2 rare cases of 46, XY DSD, specifically XY pure gonadal dysgenesis and complete androgen insensitivity syndrome. RESULTS: Both cases underwent prophylactic gonadectomy due to the elevated risk of gonadal malignancy. Bilateral gonadoblastoma and dysgerminoma were diagnosed on one side, while Leydig cell hyperplasia and only Sertoli cells were diagnosed in the seminiferous tubules on both sides. The normal menstruation for the pure gonadal dysgenesis patient only as CAIS patients never menstruate. Estrogen replacement therapy was administered periodically to promote the development of secondary sexual characteristics and menstruation in pure gonadal dysgenesis case, as well as to prevent osteoporosis. Follow-up examinations revealed no tumor recurrence, and the patient with Swyer syndrome had regular menstrual cycles. CONCLUSION: Laparoscopic bilateral prophylactic gonadectomy and long-term hormone therapy with patient counseling and support are recommended.


Subject(s)
Androgen-Insensitivity Syndrome , Gonadal Dysgenesis, 46,XY , Humans , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/diagnosis , Androgen-Insensitivity Syndrome/complications , Male , Gonadal Dysgenesis, 46,XY/genetics , Gonadal Dysgenesis, 46,XY/diagnosis , Female , Gonadoblastoma/genetics , Gonadoblastoma/diagnosis , Gonadoblastoma/surgery
19.
Am J Med Genet A ; 161A(8): 1882-96, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23824832

ABSTRACT

Deletion of the distal segment of 9p causes a syndrome comprising trigonocephaly, minor anomalies, and intellectual disability. Patients with this condition also frequently present with genitourinary abnormalities including cryptorchidism, hypospadias, ambiguous genitalia, or 46,XY testicular dysgenesis. The region responsible for the gonadal dysgenesis has been localized to 9p24.3 with the likely responsible gene identified as DMRT1. Similar to patients with other molecular causes of 46,XY gonadal dysgenesis, patients with partial del 9p have an increased risk of gonadoblastoma. We present two patients with 46,XY gonadal dysgenesis due to partial 9p monosomy. Both patients were also diagnosed with gonadoblastoma following gonadectomy at an early age. Chromosomal microarray analyses refined the cytogenetic abnormalities and allowed potential genotype-phenotype relationships to be determined. We also review the literature as it pertains to partial 9p monosomy, genital abnormalities and gonadoblastoma and note that a large percentage of affected patients present with two copy number variations. We propose that a two-hit mechanism may be involved in the incomplete penetrance and variable expressivity of partial 9p monosomy and an abnormal genital phenotype. The significant percentage of gonadoblastoma in patients with 46,XY complete gonadal dysgenesis due to partial 9p monosomy also continues to support the necessity of gonadectomy in this patient population.


Subject(s)
Gonadal Dysgenesis, 46,XY/genetics , Gonadoblastoma/genetics , Chromosome Deletion , Chromosomes, Human, Pair 9/genetics , Female , Gonadal Dysgenesis, 46,XY/pathology , Gonadoblastoma/pathology , Humans , Infant , Infant, Newborn , Karyotyping , Review Literature as Topic
20.
Nat Genet ; 17(4): 467-70, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9398852

ABSTRACT

Frasier syndrome (FS) is a rare disease defined by male pseudo-hermaphroditism and progressive glomerulopathy. Patients present with normal female external genitalia, streak gonads and XY karyotype and frequently develop gonadoblastoma. Glomerular symptoms consist of childhood proteinuria and nephrotic syndrome, characterized by unspecific focal and segmental glomerular sclerosis, progressing to end-stage renal failure in adolescence or early adulthood. No case of Wilms' tumour has been reported, even in patients with extended follow-up. In contrast with FS patients, most individuals with Denys-Drash syndrome (DDS; refs 6,7) have ambiguous genitalia or a female phenotype, an XY karyotype and dysgenetic gonads. Renal symptoms are characterized by diffuse mesangial sclerosis, usually before the age of one year, and patients frequently develop Wilms' tumour. Mutations of the Wilms'-tumour gene, WT1, cause different pathologies of the urogenital system, including DDS. WT1 is composed of ten exons and encodes a protein with four zinc-finger motifs and transcriptional and tumour-suppressor activities. Alternative splicing generates four isoforms: the fifth exon may or may not be present, and an alternative splice site in intron 9 allows the addition of three amino acids (KTS) between the third and fourth zinc fingers of WT1 (ref. 17). Here we demonstrate that FS is caused by mutations in the donor splice site in intron 9 of WT1, with the predicted loss of the +KTS isoform. Examination of WT1 transcripts indeed showed a diminution of the +KTS/-KTS isoform ratio in patients with FS.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Wilms Tumor , Mutation , RNA Splicing , Transcription Factors/genetics , Urogenital Abnormalities/genetics , Adult , Disorders of Sex Development/genetics , Female , Gonadal Dysgenesis/genetics , Gonadoblastoma/genetics , Humans , Ovarian Neoplasms/genetics , Syndrome , WT1 Proteins
SELECTION OF CITATIONS
SEARCH DETAIL