Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Sex Dev ; 11(5-6): 225-237, 2017.
Article in English | MEDLINE | ID: mdl-29393262

ABSTRACT

Androgen insensitivity syndrome (AIS) is a hereditary condition in patients with a 46,XY karyotype in which loss-of-function mutations of the androgen receptor (AR) gene are responsible for defects in virilization. The aim of this study was to investigate the consequences of the lack of AR activity on germ cell survival and the degree of testicular development reached by these patients by analyzing gonadal tissue from patients with AIS prior to Sertoli cell maturation at puberty. Twenty-three gonads from 13 patients with AIS were assessed and compared to 18 testes from 17 subjects without endocrine disorders. The study of the gonadal structure using conventional microscopy and the ultrastructural characteristics of remnant germ cells using electron microscopy, combined with the immunohistochemical analysis of specific germ cell markers (MAGE-A4 for premeiotic germ cells and of OCT3/4 for gonocytes), enabled us to carry out a thorough investigation of germ cell life in an androgen-insensitive microenvironment throughout prepuberty until young adulthood. Here, we show that germ cell degeneration starts very early, with a marked decrease in number after only 2 years of life, and we demonstrate the permanence of gonocytes in AIS testis samples until puberty, describing 2 different populations. Additionally, our results provide further evidence for the importance of AR signaling in peritubular myoid cells during prepuberty to maintain Sertoli and spermatogonial cell health and survival.


Subject(s)
Androgen-Insensitivity Syndrome/pathology , Puberty/metabolism , Puberty/physiology , Androgen-Insensitivity Syndrome/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Child , Child, Preschool , Germ Cells/metabolism , Humans , Immunohistochemistry , Infant , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Spermatogonia/metabolism , Spermatogonia/pathology , Testis/metabolism , Testis/pathology
2.
Clin Endocrinol (Oxf) ; 82(5): 704-11, 2015 May.
Article in English | MEDLINE | ID: mdl-25040157

ABSTRACT

BACKGROUND: IGF1R gene mutations have been associated with varying degrees of intrauterine and postnatal growth retardation, and microcephaly. OBJECTIVE: To identify and characterize IGF1R gene variations in a cohort of 28 Argentinean children suspected of having IGF-1 insensitivity, who were selected on the basis of the association of pre/postnatal growth failure and microcephaly. METHODS: The coding sequence and flanking intronic regions of IGF1R gene were amplified and directly sequenced. Functional characterization was performed by two in vitro assays: 1) [Methyl-(3) H] thymidine incorporation into DNA in fibroblast cell primary cultures from patients and controls treated with IGF-1 for 16-24 h. 2) PI3K/Akt pathway was evaluated with phospho-Akt (Ser473) STAR ELISA Kit (Millipore) in fibroblast cultures from patients and controls stimulated with IGF-1 for 10 min. Prepubertal clinical and GH-IGF-1 axis evaluation was followed up. RESULTS: We identified three novel heterozygous missense mutations in three unrelated patients, de novo p.Arg1256Ser, de novo p.Asn359Tyr and p.Tyr865Cys. In control cells, proliferation assay showed that IGF-1 significantly induced DNA synthesis at 20 h and Akt phosphorylation assay that it significantly stimulated phosphorylation after 10 min (P < 0·05 by anova and Bonferroni Tests). However, no significant increase was observed in any of the three patient fibroblasts in both functional studies. GH therapy growth response in two patients was inconsistent. CONCLUSION: These variations led to failure of the IGF1R function causing pre- and postnatal growth retardation and microcephaly. Microcephaly should be considered in the evaluation of SGA patients, because it seems to favour the frequency of detection of IGF1R mutations.


Subject(s)
Growth Disorders/genetics , Microcephaly/genetics , Mutation , Receptors, Somatomedin/genetics , Adult , Argentina , Biopsy , Child , Child, Preschool , Cohort Studies , DNA/genetics , DNA Mutational Analysis , Female , Fetal Growth Retardation/genetics , Fibroblasts/metabolism , Genetic Variation , Heterozygote , Human Growth Hormone/metabolism , Humans , Infant , Insulin-Like Growth Factor I/metabolism , Introns , Male , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1
3.
Horm Res Paediatr ; 78(2): 119-26, 2012.
Article in English | MEDLINE | ID: mdl-22907560

ABSTRACT

In humans, steroidogenic factor 1 (NR5A1/SF-1) mutations have been reported to cause gonadal dysgenesis, with or without adrenal failure, in both 46,XY and 46,XX individuals. We have previously reported extreme within-family variability in affected 46,XY patients. Even though low ovarian reserve with preserved fertility has been reported in females harboring NR5A1 gene mutations, fertility has only been observed in one reported case in affected 46,XY individuals. A kindred with multiple affected members presenting gonadal dysgenesis was studied. Four 46,XY individuals presented severe hypospadias at birth, one of them associated with micropenis and cryptorchidism. The other 3 developed spontaneous male puberty, and 1 has fathered 5 children. Four 46,XX patients presented premature ovarian failure (one of them was not available for the study) or high follicle-stimulating hormone levels. Mutational analysis of the NR5A1 gene revealed a novel heterozygous mutation, c.938G→A, predicted to cause a p.Arg313Hys amino acid change. A highly conserved amino acid of the ligand-binding domain of the mature protein is affected, predicting abnormal protein function. We confirm that preserved fertility can be observed in patients with a 46,XY disorder of sex development due to heterozygous mutations in the NR5A1 gene.


Subject(s)
Disorder of Sex Development, 46,XY/genetics , Fertility , Gonadal Dysgenesis, 46,XX/genetics , Mutation , Steroidogenic Factor 1/genetics , Adult , Child, Preschool , Female , Humans , Male , Pedigree
4.
Horm Res Paediatr ; 75(1): 70-7, 2011.
Article in English | MEDLINE | ID: mdl-20861607

ABSTRACT

BACKGROUND: Three novel heterozygous SF-1 gene mutations affecting multiple members of two unrelated families with a history of 46,XY disorders of sex development (DSD) and 46,XX ovarian insufficiency are described. METHODS: clinical and mutational analysis of the SF-1 gene in 9 subjects of two families. RESULTS: family 1 had 2 affected 46,XY DSD subjects. One, born with severe perineal hypospadias, was raised as a male, and presented normal adolescence. The other, born with ambiguous genitalia, uterus, and mild testicular dysgenesis, was raised as a female. A W279X heterozygous mutation and an intronic deletion (g3314-3317delTCTC (IVS 4 + 8) was found in the SF-1 gene. In family 2, 4/6 affected siblings had 46,XY DSD or hypospadias. An affected 46,XX sister had normal sexual development but increased FSH levels. The 37-year-old affected mother had entered menopause. An Y183X heterozygous mutation was detected. CONCLUSION: an extreme within-family phenotypic variability, ranging from severe prenatal undervirilization to normal pubertal development, was observed in 46,XY-affected siblings, indicating that other unknown factors might be involved in the phenotype. Low ovarian reserve and preserved fertility in 46,XX subjects can be observed in heterozygous SF-1 gene mutations.


Subject(s)
46, XX Disorders of Sex Development/genetics , Disorder of Sex Development, 46,XY/genetics , Genetic Variation , Primary Ovarian Insufficiency/genetics , Steroidogenic Factor 1/genetics , 46, XX Disorders of Sex Development/pathology , Adolescent , Adult , Child , Child, Preschool , Disorder of Sex Development, 46,XY/pathology , Female , Genetic Association Studies , Gonadal Dysgenesis/genetics , Humans , Hyperplasia , Hypospadias/genetics , Infant , Male , Mutation , Pedigree , Testis/pathology , Young Adult
5.
J Clin Endocrinol Metab ; 95(9): E98-103, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20534762

ABSTRACT

CONTEXT: Patients with TSH-beta subunit defects and congenital hypothyroidism are missed by TSH-based neonatal screening. OBJECTIVE: Our objective was to report the molecular consequences of a novel splice-junction mutation and a novel missense mutation in the TSH-beta subunit gene found in two patients with congenital central hypothyroidism and conventional treatment-resistant anemia. RESULTS: Patient 1 had a homozygous G to A nucleotide change at the 5' donor splice site of exon/intron 2. This resulted in a silent change at codon 34 of the mature protein. In vitro splicing assays showed that the mutant minigene dramatically affected pre-mRNA processing, causing exon 2 to be completely skipped. The putative product from a new out-of-frame translational start point in exon 3 is expected to yield a nonsense 25-amino-acid peptide. In patient 2, sequence analysis revealed a compound heterozygosis for the already reported 313delT (C105Vfs114X) mutation and for a second novel mutation in exon 3, substituting G for A at cDNA nucleotide position 323, resulting in a C88Y change. This cysteine residue is conserved among all dimeric pituitary and placental glycoprotein hormone-beta subunits. Data from in silico analysis confirmed that the C88Y mutation would affect subunit conformation. Indeed, two different bioinformatics approaches, PolyPhen and SIFT analysis, predicted C88Y to be a damaging substitution. CONCLUSIONS: In isolated TSH deficiency, the exact molecular diagnosis is mandatory for diagnosis of isolated pituitary deficiency, delineation of prognosis, and genetic counseling. Moreover, diagnosis of central hypothyroidism should be considered in the face of severe infant anemia of uncertain etiology.


Subject(s)
Congenital Hypothyroidism/genetics , Mutation , Neonatal Screening , Thyrotropin, beta Subunit/genetics , Amino Acid Substitution/physiology , Child , Child, Preschool , False Negative Reactions , Humans , Infant, Newborn , Mutation/physiology , Neonatal Screening/standards , Pedigree , Polymorphism, Single Nucleotide/physiology , Protein Conformation , Thyrotropin, beta Subunit/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...