Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 303
Filter
Add more filters

Publication year range
1.
Nature ; 617(7961): 616-622, 2023 May.
Article in English | MEDLINE | ID: mdl-36972684

ABSTRACT

Steroid hormone receptors are ligand-binding transcription factors essential for mammalian physiology. The androgen receptor (AR) binds androgens mediating gene expression for sexual, somatic and behavioural functions, and is involved in various conditions including androgen insensitivity syndrome and prostate cancer1. Here we identified functional mutations in the formin and actin nucleator DAAM2 in patients with androgen insensitivity syndrome. DAAM2 was enriched in the nucleus, where its localization correlated with that of the AR to form actin-dependent transcriptional droplets in response to dihydrotestosterone. DAAM2 AR droplets ranged from 0.02 to 0.06 µm3 in size and associated with active RNA polymerase II. DAAM2 polymerized actin directly at the AR to promote droplet coalescence in a highly dynamic manner, and nuclear actin polymerization is required for prostate-specific antigen expression in cancer cells. Our data uncover signal-regulated nuclear actin assembly at a steroid hormone receptor necessary for transcription.


Subject(s)
Actins , Formins , Nuclear Proteins , Receptors, Androgen , Transcription, Genetic , Humans , Actins/metabolism , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/metabolism , Androgens/pharmacology , Androgens/metabolism , Formins/metabolism , Gene Expression Regulation/drug effects , Nuclear Proteins/metabolism , Polymerization/drug effects , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , RNA Polymerase II/metabolism , Signal Transduction/drug effects , Steroids/metabolism , Steroids/pharmacology , Testosterone/analogs & derivatives , Transcription, Genetic/drug effects
2.
Mol Genet Genomics ; 295(5): 1103-1111, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32435981

ABSTRACT

The androgen insensitivity syndrome (AIS) is a congenital disease characterized by androgen resistance due to androgen receptor (AR) gene mutations, resulting in disorders of sex differentiation in 46,XY individuals. However, the underlying mechanisms in the majority of AR variants and the phenotype-genotype correlations are unclear. Here, we identified a p.Y764H variant of the AR gene that results in different phenotypes in a family. Structural analyses revealed that amino acid substitution affected protein properties and spatial conformation, and in vitro, functional studies showed impaired nuclear translocation ability of the mutated protein. Moreover, the extent to which this variant reduced nuclear translocation depends on the dihydrotestosterone (DHT) concentrations. Our results, for the first time, demonstrated a pathogenesis of the p.Y764H mutations in AR resulting in AIS phenotype, and indicated that AIS patients with p.Y764H mutation and preserved gonad might have residual AR activity at high androgen levels, putting patients at risk for pubertal virilization in the future. We provide an in-depth insight into the pathogenesis in AIS based on the amino acid substitution, which may help aid its precise diagnosis, personalized treatment, and organized follow-up to avoid gender dysphoria.


Subject(s)
Amino Acid Substitution , Androgen-Insensitivity Syndrome/genetics , Cell Nucleus/metabolism , Exome Sequencing/methods , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Androgen-Insensitivity Syndrome/metabolism , Animals , COS Cells , China , Chlorocebus aethiops , Female , Hemizygote , Humans , Infant , Male , Pedigree , Phenotype , Protein Transport , Receptors, Androgen/chemistry , Siblings
3.
Endocr J ; 66(4): 387-393, 2019 Apr 25.
Article in English | MEDLINE | ID: mdl-30787207

ABSTRACT

Endogenous and exogenous androgens induce masculinization of external genitalia through binding to the androgen receptor (AR). The target genes of androgens in external genitalia remain to be determined, although previous studies have shown that the apolipoprotein D gene (APOD) was significantly upregulated by dihydrotestosterone (DHT), the most potent androgen in humans. In the present study, we performed microarray analysis for genital skin fibroblasts obtained from four boys with buried penis (the control individuals) and a patient with partial androgen insensitivity syndrome (PAIS) due to a hypomorphic mutation in AR (the PAIS patient). We identified 24 transcripts that were upregulated or downregulated by DHT in all samples of control individuals and, to a lesser extent, in the sample of the PAIS patient. Differences between DHT-treated and -untreated samples were small; the results of 24 transcripts did not reach statistical significance. The 24 transcripts included CYP1B1, a gene possibly involved in the development of genital tubercle in mice, and APOD, as well as several genes that have been reported as androgen targets in prostate or other tissues. The results of this study indicate that androgen-mediated masculinization of external genitalia is unlikely to depend on massive transcriptional changes in specific AR target genes. Rather, minor transcriptional changes of several genes, and/or a complex molecular network may play a major role in penile development. Importantly, our data suggest the possible involvement of CYP1B1 in human genital development and confirm the clinical importance of APOD as a biomarker for AR function.


Subject(s)
Androgens/pharmacology , Dihydrotestosterone/pharmacology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Penis/drug effects , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Infant , Male , Penis/cytology , Penis/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Tissue Array Analysis
4.
Int J Mol Sci ; 20(21)2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31671693

ABSTRACT

We analyzed three cases of Complete Androgen Insensitivity Syndrome (CAIS) and report three hitherto undisclosed causes of the disease. RNA-Seq, Real-timePCR, Western immunoblotting, and immunohistochemistry were performed with the aim of characterizing the disease-causing variants. In case No.1, we have identified a novel androgen receptor (AR) mutation (c.840delT) within the first exon in the N-terminal transactivation domain. This thymine deletion resulted in a frameshift and thus introduced a premature stop codon at amino acid 282. In case No.2, we observed a nonsynonymous mutation in the ligand-binding domain (c.2491C>T). Case No.3 did not reveal AR mutation; however, we have found a heterozygous mutation in CYP11A1 gene, which has a role in steroid hormone biosynthesis. Comparative RNA-Seq analysis of CAIS and control revealed 4293 significantly deregulated genes. In patients with CAIS, we observed a significant increase in the expression levels of PLCXD3, TM4SF18, CFI, GPX8, and SFRP4, and a significant decrease in the expression of SPATA16, TSACC, TCP10L, and DPY19L2 genes (more than 10-fold, p < 0.05). Our findings will be helpful in molecular diagnostics of patients with CAIS, as well as the identified genes could be also potential biomarkers for the germ cells differentiation process.


Subject(s)
Androgen-Insensitivity Syndrome/genetics , Cholesterol Side-Chain Cleavage Enzyme/genetics , Mutation , Receptors, Androgen/genetics , Sequence Analysis, DNA/methods , Adolescent , Adult , Androgen-Insensitivity Syndrome/metabolism , Case-Control Studies , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Exons , Female , Frameshift Mutation , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Protein Domains , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Young Adult
5.
Arch Sex Behav ; 47(8): 2287-2298, 2018 11.
Article in English | MEDLINE | ID: mdl-30128981

ABSTRACT

Disorders of sex development (DSD) are congenital conditions in which the typical genetic and hormonal profiles are affected and thereby the usual process of sexual differentiation. Most of these studies, however, have been conducted in Western countries. In the present study, preschool sex-typed activities of Iranian individuals with DSD and their age-matched non-affected male and female relatives were assessed using the Pre-School Activities Inventory (PSAI) modified for retrospective self-report. A total of 192 individuals participated in our study, including 33 46,XX individuals with congenital adrenal hyperplasia (CAH; M age = 10.36, SD = 5.52), 15 46,XY individuals with complete androgen insensitivity syndrome (CAIS; M age = 19.8, SD = 7.14), and 16 46,XY individuals with 5-alpha reductase deficiency type-2 (5α-RD-2; M age = 17.31, SD = 7.28), as well as one age-matched non-affected male and female relative for each patient. With regard to PSAI scores, male-identifying participants with 5α-RD-2 and male controls reported similar levels of male-typical childhood play. Female-identifying participants with 5α-RD-2 and CAH showed comparable scores: significantly less masculine and more feminine than male controls, but significantly more masculine and less feminine than females with CAIS and female controls. These findings support the role of androgens in the development of sex-typical childhood play behavior, with those being exposed to higher levels of fetal functional androgens expressing more masculine behavior at preschool ages.


Subject(s)
Child Behavior , Gender Identity , Sex Characteristics , Sexual Development , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/deficiency , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Adolescent , Adrenal Hyperplasia, Congenital/genetics , Adrenal Hyperplasia, Congenital/metabolism , Adrenal Hyperplasia, Congenital/physiopathology , Adult , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/metabolism , Androgen-Insensitivity Syndrome/physiopathology , Androgens/metabolism , Child , Child, Preschool , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/metabolism , Disorder of Sex Development, 46,XY/physiopathology , Female , Humans , Hypospadias/genetics , Hypospadias/metabolism , Hypospadias/physiopathology , Iran , Male , Retrospective Studies , Self Report , Sex Differentiation , Steroid Metabolism, Inborn Errors/genetics , Steroid Metabolism, Inborn Errors/metabolism , Steroid Metabolism, Inborn Errors/physiopathology
6.
FASEB J ; 29(3): 894-910, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25404712

ABSTRACT

Leydig cell number and function decline as men age, and low testosterone is associated with all "Western" cardio-metabolic disorders. However, whether perturbed androgen action within the adult Leydig cell lineage predisposes individuals to this late-onset degeneration remains unknown. To address this, we generated a novel mouse model in which androgen receptor (AR) is ablated from ∼75% of adult Leydig stem cell/cell progenitors, from fetal life onward (Leydig cell AR knockout mice), permitting interrogation of the specific roles of autocrine Leydig cell AR signaling through comparison to adjacent AR-retaining Leydig cells, testes from littermate controls, and to human testes, including from patients with complete androgen insensitivity syndrome (CAIS). This revealed that autocrine AR signaling is dispensable for the attainment of final Leydig cell number but is essential for Leydig cell maturation and regulation of steroidogenic enzymes in adulthood. Furthermore, these studies reveal that autocrine AR signaling in Leydig cells protects against late-onset degeneration of the seminiferous epithelium in mice and inhibits Leydig cell apoptosis in both adult mice and patients with CAIS, possibly via opposing aberrant estrogen signaling. We conclude that autocrine androgen action within Leydig cells is essential for the lifelong support of spermatogenesis and the development and lifelong health of Leydig cells.


Subject(s)
Androgen-Insensitivity Syndrome/pathology , Androgens/pharmacology , Apoptosis/drug effects , Leydig Cells/pathology , Receptors, Androgen/physiology , Testis/pathology , Adolescent , Adult , Androgen-Insensitivity Syndrome/drug therapy , Androgen-Insensitivity Syndrome/metabolism , Animals , Autocrine Communication , Blotting, Western , Cells, Cultured , Child , Humans , Immunoenzyme Techniques , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protective Agents/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spermatogenesis/drug effects , Testis/drug effects , Testis/metabolism , Young Adult
7.
Am J Physiol Regul Integr Comp Physiol ; 309(2): R189-95, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25947172

ABSTRACT

Testosterone (TES) and other androgens exert a direct vasorelaxing action on the vasculature in vitro that is structurally specific and independent of cytosolic androgen receptor (AR). The effects of intravenous androgen infusions on mean arterial blood pressure (BP) and heart rate (HR) were determined in conscious, unrestrained, chronically catheterized, ganglionically blocked (hexamethonium, HEX; 30 mg/kg ip) male Sprague-Dawley (SD) and testicular-feminized male (Tfm; AR-deficient) rats, 16-20 wk of age. BP and HR were recorded at baseline and with increasing doses of androgens (0.375-6.00 µmol·kg(-1)·min(-1) iv; 10 min/dose). Data are expressed as means ± SE (n = 5-8 rats/group). In SD rats, baseline BP and HR averaged 103 ± 4 mmHg and 353 ± 12 beats/min (bpm). TES produced a dose-dependent reduction in BP to a low of 87 ± 4 mmHg (Δ16%), while HR was unchanged (354 ± 14 bpm). Neither BP (109 ± 3 mmHg) nor HR (395 ± 13 bpm) were altered by vehicle (10% EtOH in 0.9% saline; 0.15 ml·kg(-1)·min(-1), iv). In Tfm, TES produced a similar reduction in BP (99 ± 3 to 86 ± 3 mmHg, Δ13%); HR was unchanged (369 ± 18 bpm). In SD, 5ß-dihydrotestosterone (genomically inactive metabolite) produced a greater reduction in BP than TES (102 ± 2 to 79 ± 2 mmHg, Δ23%); HR was unchanged (361 ± 9). A 20-µg iv bolus of sodium nitroprusside in both SD and Tfm rats reduced BP 30-40 mmHg, while HR was unchanged, confirming blockade by HEX. Pretreatment of SD rats with neuronal nitric oxide synthase (nNOS) inhibitor (S-methyl-thiocitrulline, SMTC; 20 µg·kg(-1)·min(-1) × 30 min) abolished the hypotensive effects of TES infusion on BP (104 ± 2 vs. 101 ± 2 mmHg) and HR (326 ± 11 vs. 324 ± 8 bpm). These data suggest the systemic hypotensive effect of TES and other androgens involves a direct vasodilatory action on the peripheral vasculature which, like the effect observed in isolated arteries, is structurally specific and AR-independent, and involves activation of nNOS.


Subject(s)
Androgens/administration & dosage , Arterial Pressure/drug effects , Arteries/drug effects , Hypotension/chemically induced , Nitric Oxide Synthase Type I/metabolism , Testosterone/administration & dosage , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/metabolism , Androgen-Insensitivity Syndrome/physiopathology , Androgens/chemistry , Animals , Arteries/enzymology , Arteries/physiopathology , Dihydrotestosterone/administration & dosage , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Heart Rate/drug effects , Hypotension/enzymology , Hypotension/physiopathology , Infusions, Intravenous , Male , Molecular Structure , Nitric Oxide Synthase Type I/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Structure-Activity Relationship , Testosterone/analogs & derivatives , Testosterone/chemistry , Time Factors , Vasodilation/drug effects
8.
Sci Rep ; 14(1): 16302, 2024 07 15.
Article in English | MEDLINE | ID: mdl-39009627

ABSTRACT

Androgen insensitivity syndrome (AIS) is a difference of sex development (DSD) characterized by different degrees of undervirilization in individuals with a 46,XY karyotype despite normal to high gonadal testosterone production. Classically, AIS is explained by hemizygous mutations in the X-chromosomal androgen receptor (AR) gene. Nevertheless, the majority of individuals with clinically diagnosed AIS do not carry an AR gene mutation. Here, we present a patient with a 46,XY karyotype, born with undervirilized genitalia, age-appropriate testosterone levels and no uterus, characteristic for AIS. Diagnostic whole exome sequencing (WES) showed a maternally inherited LINE1 (L1) retrotransposon insertion in the 5' untranslated region (5'UTR) of the AR gene. Long-read nanopore sequencing confirmed this as an insertion of a truncated L1 element of ≈ 2.7 kb and showed an increased DNA methylation at the L1 insertion site in patient-derived genital skin fibroblasts (GSFs) compared to healthy controls. The insertion coincided with reduced AR transcript and protein levels in patient-derived GSFs confirming the clinical diagnosis AIS. Our results underline the relevance of retrotransposons in human disease, and expand the growing list of human diseases associated with them.


Subject(s)
Androgen-Insensitivity Syndrome , DNA Methylation , Epigenesis, Genetic , Long Interspersed Nucleotide Elements , Receptors, Androgen , Humans , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Male , Long Interspersed Nucleotide Elements/genetics , Female , Exome Sequencing , Transcription, Genetic
9.
J Biol Chem ; 287(37): 31195-206, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22829593

ABSTRACT

The androgen receptor (AR) mediates the effects of male sexual hormones on development and physiology. Alterations in AR function are central to reproductive disorders, prostate cancer, and Kennedy disease. AR activity is influenced by post-translational modifications, but their role in AR-based diseases is poorly understood. Conjugation by small ubiquitin-like modifier (SUMO) proteins at two synergy control (SC) motifs in AR exerts a promoter context-dependent inhibitory role. SC motifs are composed of a four-amino acid core that is often preceded and/or followed by nearby proline or glycine residues. The function of these flanking residues, however, has not been examined directly. Remarkably, several AR mutations associated with oligospermia and androgen insensitivity syndrome map to Pro-390, the conserved proline downstream of the first SC motif in AR. Similarly, mutations at Gly-524, downstream of the second SC motif, were recovered in recurrent prostate cancer samples. We now provide evidence that these clinically isolated substitutions lead to a partial loss of SC motif function and AR SUMOylation that affects multiple endogenous genes. Consistent with a structural role as terminators of secondary structure elements, substitution of Pro-390 by Gly fully supports both SC motif function and SUMOylation. As predicted from the functional properties of SC motifs, the clinically isolated mutations preferentially enhance transcription driven by genomic regions harboring multiple AR binding sites. The data support the view that alterations in AR SUMOylation play significant roles in AR-based diseases and offer novel SUMO-based therapeutic opportunities.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Mutation , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Sumoylation , Amino Acid Motifs , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/therapy , HEK293 Cells , Humans , Male , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Receptors, Androgen/genetics
10.
J Biol Chem ; 287(14): 10905-15, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22334658

ABSTRACT

Naturally occurring germ line mutations in the X-linked human androgen receptor (AR) gene cause incomplete masculinization of the external genitalia by disrupting AR function in males with androgen insensitivity syndrome. Almost all AR missense mutations that cause androgen insensitivity syndrome are located in the highly structured DNA and ligand binding domains. In this report we investigate the functional defect associated with an AR exon 1 missense mutation, R405S, that caused partial androgen insensitivity. The 46,XX heterozygous maternal carrier had a wild-type Arg-405 CGC allele but transmitted an AGC mutant allele coding for Ser-405. At birth, the 46,XY proband had a bifid scrotum, hypospadias, and micropenis consistent with clinical stage 3 partial androgen insensitivity. Androgen-dependent transcriptional activity of AR-R405S expressed in CV1 cells was less than wild-type AR and refractory in androgen-dependent AR NH(2)- and carboxyl interaction transcription assays that depend on the coregulator effects of melanoma antigen-A11. This mutation created a Ser-405 phosphorylation site evident by the gel migration of an AR-R405S NH(2)-terminal fragment as a double band that converted to the wild-type single band after treatment with λ-phosphatase. Detrimental effects of the R405S mutation were related to the proximity of the AR WXXLF motif (433)WHTLF(437) required for melanoma antigen-A11 and p300 to stimulate transcriptional activity associated with the AR NH(2)- and carboxyl-terminal interaction. We conclude that the coregulator effects of melanoma antigen-A11 on the AR NH(2)- and carboxyl-terminal interaction amplify the androgen-dependent transcriptional response to p300 required for normal human male sex development in utero.


Subject(s)
Androgen-Insensitivity Syndrome/genetics , Antigens, Neoplasm/metabolism , Exons/genetics , Mutation, Missense , Neoplasm Proteins/metabolism , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Transcriptional Activation/genetics , Amino Acid Motifs , Androgen-Insensitivity Syndrome/metabolism , Base Sequence , Binding Sites , E1A-Associated p300 Protein/metabolism , Female , Humans , Male , Phenotype , Phosphorylation/genetics , Receptors, Androgen/genetics
11.
Chin Med Sci J ; 28(2): 88-94, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23806370

ABSTRACT

OBJECTIVE: To determine whether testosterone modulates markers of cardiomyocytes aging via its classic androgen receptor (AR)-dependent pathway or conversion to estradiol. METHODS: Male littermates and testicular feminized (Tfm) mice were randomly separated into 4 experimental groups littermate controls (n=8), Tfm mice (n=7), testosterone-treated Tfm mice (n=8), and Tfm mice treated with testosterone in combination with the aromatase inhibitor anastrazole (n=7). Cardiomyocytes were isolated from mouse left ventricles, the activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), and the amount of malondialdehyde (MDA) were measured using colorimetry method, and expression of p16(INK4α) and retinoblastoma (Rb) proteins were detected by Western blotting. RESULTS: The SOD and GSH-Px enzyme activities of cardiomyocytes were decreased, and the MDA levels and the expression of p16(INK4α) and Rb proteins were increased in Tfm mice compared with control mice. An increase was observed in the activities of SOD and GSH-Px enzyme as well as a decrease in MDA levels and the expression of p16(INK4α) and Rb proteins in the testosterone-treated Tfm mice. After co-treatment with anastrazole in Tfm mice, these improvement were partly inhibited. CONCLUSION: Physiological testosterone replacement can delay cardiomyocyte aging in Tfm mice, an effect that is independent of the AR pathway and in part conversion to estradiol.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Cellular Senescence , Myocytes, Cardiac/physiology , Receptors, Androgen/physiology , Testosterone/physiology , Animals , Cyclin-Dependent Kinase Inhibitor p16/analysis , Female , Glutathione Peroxidase/metabolism , Male , Mice , Superoxide Dismutase/metabolism
12.
Genes Genomics ; 45(4): 467-474, 2023 04.
Article in English | MEDLINE | ID: mdl-35445939

ABSTRACT

BACKGROUND: Androgens and androgen receptor (AR) are critical regulators of the masculinization process in male sexual development. The absence of a functioning AR results in the development of the androgen insensitivity syndrome (AIS), a rare disorder of sexual development (DSD) characterized by the external genitalia feminization, gynecomastia, and impaired spermatogenesis. OBJECTIVE: To determine the AR gene mutations associated with male DSD in four unrelated Vietnamese patients. METHODS: To detect the disease-causing mutations, whole exome sequencing (WES) was performed on four patients diagnosed with AIS. Sanger sequencing was then used for validation of the identified mutations. Finally, 12 web-based tools, three-dimensional protein modeling software, and the guidelines issued by the American College of Medical Genetics and Genomics were used to assess the potential pathogenicity of these mutations. RESULTS: Four distinct novel mutations, namely c.1834T > A (p.Cys612Ser), c.2122 C > G (p.Leu708Val), c.2630T > G (p.Phe877Cys), and c.2641 C > A (p.Leu881Met) in the AR gene, were identified in four AIS patients using WES. The in silico analysis results revealed that the Cys612, Leu708, Phe877, and Leu881 sites are important for an appropriate response to androgens of the AR, and mutation at these sites can have adverse effects on the AR functions, androgen-AR interaction, and AR signaling pathway. CONCLUSIONS: WES and in silico analyses strongly suggested that four novel AR mutations are pathogenic and have led to the development of AIS in the four Vietnamese patients under consideration.


Subject(s)
Androgen-Insensitivity Syndrome , Humans , Male , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/diagnosis , Androgen-Insensitivity Syndrome/metabolism , Androgens , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Southeast Asian People , Mutation
13.
Eur J Endocrinol ; 188(1)2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36721956

ABSTRACT

OBJECTIVE: Congenital defects of androgen synthesis or action in 46,XY individuals can result in impaired virilisation, despite the apparent testicular development. In a recent case, report of a young adult with complete androgen insensitivity syndrome (CAIS), tumourous gonadal tissue was shown to express HSD17B3 in Sertoli cells (SCs) and not in Leydig cells (LCs). This expression pattern differs from the typical adult human testis and resembles a foetal mouse testis, suggesting an underlying testicular development and function defect. Here, we investigate the effect of altered androgen signalling in gonads from five 46,XY individuals with defects in androgen synthesis or action. METHODS: Gonadal tissue sections from four patients with CAIS, one with CYP17A1 deficiency, and one control were immunostained for LC developmental and steroidogenic markers. The expression of some of these markers during development was investigated by reanalysing previously published single-cell RNA sequencing (scRNA-seq) data from normal human testicular tissues. RESULTS: All gonadal tissues from the patients show an exclusive expression of HSD17B3 in SCs and an expression of the foetal/immature LC marker DLK1 in a subset of LCs, suggesting an androgen-dependent differentiation defect of adult SCs and LCs. Furthermore, reanalysis of scRNA-seq data reveals an expression of HSD17B3 in foetal and neonatal SCs that is downregulated in adult SCs. CONCLUSIONS: Androgen signalling may affect the differentiation of adults, but possibly not foetal SCs or LCs, and may induce a shift of testosterone production from the tubular compartment in the foetal phase to the interstitial compartment in the adult phase.


Subject(s)
Androgen-Insensitivity Syndrome , Androgens , Animals , Humans , Male , Mice , Young Adult , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/metabolism , Androgens/metabolism , Gonads , Leydig Cells/metabolism , Testis/metabolism , Testosterone/metabolism
14.
Gynecol Endocrinol ; 27(11): 940-3, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21501000

ABSTRACT

Androgen receptor (AR) is one of the large superfamily of nuclear hormone receptors. AR consists of distinct domains including an N-terminal DNA-binding domain and a C-terminal ligand-binding domain (LBD). Regulation of AR nuclear import and subsequent transactivation activity represent essential steps in androgen action. Mutations in the AR gene are known to cause different degrees of androgen insensitivity syndrome (AIS). This study aimed to identify the possible contribution of LBD of AR to cellular distribution, ligand binding, and transactivation activities using mutant AR clone lacking the entire LBD that we previously observed in an AIS patient. Subcellular distribution was assessed by green fluorescence protein-tagged vector and transcriptional activity was analyzed by luciferase assay. Wild-type AR had ligand-dependent transcriptional activation and nuclear import activities. On the other hand, mutant AR had no transcriptional activity regardless of the presence of ligand, 5-α-dihydroxytestosterone (DHT). These mutants were presented predominantly in the nucleus even without DHT. The observation of no transactivation in the mutant receptor must be due to the loss of complex formation between androgen and AR protein. The C-terminal domain has the critical role in the cellular localization and transactivation as well as on the ligand binding.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Receptors, Androgen/metabolism , Cell Nucleus/metabolism , Humans , Ligands , Male , Protein Binding/physiology , Receptors, Androgen/genetics , Receptors, Androgen/physiology , Transcriptional Activation
15.
Dev Dyn ; 239(2): 373-85, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19941349

ABSTRACT

The ventral urogenital sinus (UGS) of control male mice has two rows of 3-4 prostatic buds at birth, but how androgens regulate ventral bud (VB) number and patterning is unclear. VBs in both sexes appeared to be a mixture of prostatic and urethral buds. UGSs from Tfm male and antiandrogen (flutamide)-exposed mice had small VBs, suggesting that initiation of some VBs is androgen independent. Tfm male mice are widely considered completely androgen insensitive yet their UGSs were 5alpha-dihydrotestosterone (DHT)- responsive. VBs (6-8) were generally distributed bimodally on the left-right axis at both minimal and normal male androgen signaling. Yet control females and DHT-exposed Tfm males had 13-14 VBs, whose left-right distribution was fairly uniform. These results suggest that VB number and distribution respond biphasically as androgen signaling increases from minimal, and that androgens regulate bud specification. Complete VB agenesis by the selective budding inhibitor 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) required high androgen signaling.


Subject(s)
Androgens/metabolism , Body Patterning , Fetus/metabolism , Prostate/embryology , Receptors, Androgen/metabolism , Androgen Antagonists , Androgen-Insensitivity Syndrome/metabolism , Animals , Animals, Newborn , Dihydrotestosterone , Female , Flutamide , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Organ Culture Techniques , Polychlorinated Dibenzodioxins , Sex Characteristics , Signal Transduction , Teratogens , Terminology as Topic , Transcription Factors/metabolism , Urethra/embryology
16.
Front Endocrinol (Lausanne) ; 12: 727131, 2021.
Article in English | MEDLINE | ID: mdl-34526969

ABSTRACT

Introduction: Osteopenia and osteoporosis have been reported in adults with Complete Androgen Insensitivity Syndrome (CAIS). Little is known about changes in bone mineral density (BMD) in adolescents with CAIS and whether it is affected by early gonadectomy. Body composition data have not been reported. Methods: Single-center, retrospective study of CAIS adolescents who underwent dual-energy x-ray absorptiometry (DXA) (Hologic, Horizon A). Body composition is presented as lean and fat mass indices (LMI, FMI). Z-scores for lumbar spine areal BMD (LBMD), total body less head (TBLH), bone mineral content (BMC), LMI, and FMI were calculated using female normative data. Results are expressed as median and min, max. Results: Six females with genetically confirmed CAIS were identified-one with intact gonads and five with history of gonadectomy at 2-11 months. In the subject with intact gonads, LBMD-Z and TBLH BMC-Z were -1.56 and -1.26, respectively, at age 16 years. Among those with gonadectomy, LBMD-Z was -1.8 (-3.59 to 0.49) at age 15.6 years (12-16.8) and decreased in all three subjects who had longitudinal follow-up despite hormone replacement therapy (HRT). Adherence to HRT was intermittent. LMI-Z and FMI-Z were 0.1 (-1.39 to 0.7) and 1.0 (0.22 to 1.49), respectively. Conclusions: These limited data indicate that adolescents with CAIS have bone mass deficit. Further studies are needed to understand the extent of BMD abnormalities and the effect of gonadectomy, especially early in childhood, and to establish the optimal HRT regimen for bone accrual. Data on lean mass are reassuring.


Subject(s)
Androgen-Insensitivity Syndrome/complications , Body Composition/physiology , Bone Diseases, Metabolic/etiology , Absorptiometry, Photon , Adolescent , Androgen-Insensitivity Syndrome/metabolism , Androgen-Insensitivity Syndrome/pathology , Androgen-Insensitivity Syndrome/surgery , Bone Density , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Castration , Female , Humans , Ideal Body Weight/physiology , Infant , Male , Muscles/pathology , Organ Size , Retrospective Studies
17.
Int J Exp Pathol ; 91(5): 394-407, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20353424

ABSTRACT

The present study examined the response of the prostate epithelium of senescent gerbils submitted to orchiectomy and with or without steroidal blockade. Animals were divided into five groups, all surgically castrated except the control group composed of intact animals. In the experimental groups, doses of flutamide and/or tamoxifen were applied for 1, 3, 7 and 30 days postcastration. The structural methods applied reveal that castration, whether associated or not with anti-steroidal drugs, promoted short- and long-term decrease in wet and relative weights of the prostate. The quantitative decline of epithelial compartment proportion observed at the end of treatment was due to the sum of slight changes in the epithelium and lumen. The apoptotic index had risen significantly at 1 day and declined at 7 days postcastration. Androgen receptor (AR) expression decreased after 3 days of hormonal ablation, coinciding with the highest levels of apoptosis and cell proliferation observed in all treated groups. The majority of cells remained differentiated in all groups due to CK 8/18 expression. Some animals remained with injuries such as carcinomas and adenocarcinomas after hormonal ablation. In the latter a mixture of AR-positive and AR-negative cells was identified. Microinvasive carcinomas found in the group treated for 30 days consisted of PCNA-positive, inflammatory and non-proliferating cells. Low apoptosis incidence and bcl-2 positive cells were observed in these lesions. The treatments promoted a reduction of lesions in older gerbils, but treatment-resistant tumours will improve understanding of the events that lead to hormone resistance.


Subject(s)
Aging/metabolism , Aging/pathology , Androgen-Insensitivity Syndrome/metabolism , Androgen-Insensitivity Syndrome/pathology , Prostate/metabolism , Prostate/pathology , Androgen Antagonists/pharmacology , Androgens/blood , Androgens/deficiency , Animals , Apoptosis/physiology , Body Weight/physiology , Estradiol/blood , Estradiol/deficiency , Estrogen Antagonists/pharmacology , Flutamide/pharmacology , Gerbillinae , Male , Orchiectomy , Organ Size/physiology , Prostate/drug effects , Tamoxifen/pharmacology , Testosterone/blood , Testosterone/deficiency
18.
Sci Rep ; 10(1): 15062, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934281

ABSTRACT

Anti-Müllerian hormone (AMH) is secreted by Sertoli cells of the testes from early fetal life until puberty, when it is downregulated by androgens. In conditions like complete androgen insensitivity syndrome (CAIS), AMH downregulation does not occur and AMH increases at puberty, due in part to follicle-stimulating hormone (FSH) effect. However, other conditions like Peutz-Jeghers syndrome (PJS), characterised by low FSH, also have increased AMH. Because both CAIS and PJS may present as hyperoestrogenic states, we tested the hypothesis that oestradiol (E2) upregulates AMH expression in peripubertal Sertoli cells and explored the molecular mechanisms potentially involved. The results showed that E2 is capable of inducing an upregulation of endogenous AMH and of the AMH promoter activity in the prepubertal Sertoli cell line SMAT1, signalling through ERα binding to a specific ERE sequence present on the hAMH promoter. A modest action was also mediated through the membrane oestrogen receptor GPER. Additionally, the existence of ERα expression in Sertoli cells in patients with CAIS was confirmed by immunohistochemistry. The evidence presented here provides biological plausibility to the hypothesis that testicular AMH production increases in clinical conditions in response to elevated oestrogen levels.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Anti-Mullerian Hormone/metabolism , Estrogen Receptor alpha/biosynthesis , Neoplasm Proteins/biosynthesis , Peutz-Jeghers Syndrome/metabolism , Response Elements , Sertoli Cells/metabolism , Androgen-Insensitivity Syndrome/pathology , Animals , Cell Line , Child , Child, Preschool , Estradiol/metabolism , Female , Humans , Male , Mice , Peutz-Jeghers Syndrome/pathology , Sertoli Cells/pathology
19.
Clin Endocrinol (Oxf) ; 71(2): 253-60, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19178528

ABSTRACT

OBJECTIVE: Androgen insensitivity syndrome (AIS) is associated with mutations throughout the androgen receptor (AR) gene. Different mutations at the same codon have been identified in individuals with various phenotypes suggesting the nature of the codon substituted may influence the degree of AIS. We investigated if phenotype could be predicted by comparing the functionality of AR mutations with those at the same codon of known phenotype. PATIENTS: We identified patients from the Cambridge Disorders of Sex Development Database with the AR substitutions: Phe754Ser with microphallus without hypospadias and Asp690Val with complete AIS. Mutations Phe754Leu, Phe754Val and Asp690deletion (Asp690del) have previously been reported to be associated with different degrees of AIS. DESIGN: We characterized the functional properties of Phe754Ser, Phe754Leu, Phe754Val, Asp690Val and Asp690del receptor mutants in vitro and used the crystal structure of the AR ligand binding domain to model the mutations. RESULTS: The receptor mutants Phe754Ser, Phe754Leu and Phe754Val bound androgen with decreasing affinity, while Asp690Val showed reduced affinity compared to Asp690del. A similar pattern of reduced activation was seen on androgen responsive elements. We suggest how the mutations could affect AR structure, resulting in the observed phenotypes. CONCLUSIONS: The relative functional properties of Phe754 and Asp690 mutant AR receptors correlate broadly with their specific phenotypes. Therefore, comparing the molecular consequences of novel mutations with others at the same codon may be a useful aid to AIS patient management, particularly for sex of rearing decisions when prediction of functionality is important.


Subject(s)
Androgen-Insensitivity Syndrome/genetics , Mutation, Missense , Receptors, Androgen/genetics , Adolescent , Amino Acid Sequence , Androgen-Insensitivity Syndrome/metabolism , Androgens/metabolism , Child, Preschool , Female , Humans , Infant , Male , Molecular Sequence Data , Phenotype , Protein Binding , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Sequence Alignment
20.
Science ; 211(4488): 1285-94, 1981 Mar 20.
Article in English | MEDLINE | ID: mdl-7010603

ABSTRACT

Sexual dimorphism in selected extragenital tissues is described with emphasis on the molecular basis of the differences. Testosterone rather than 5 alpha-dihydrotestosterone appears to be the major intracellular androgen in organs other than skin and reproductive tract, but other steroid metabolites and their receptors are required to produce the diverse tissue differences observed in males and females. There is also evidence that multiple hormones from several endocrine glands are required to act in concert with androgens to produce and maintain their effects. Although many of the consequences of sexual dimorphism, such as body size and strength, have been evident for centuries, other differences between males and females such as disease incidence, response to drugs and toxins, and the metabolism and assimilation of dietary constituents have only recently been discovered.


Subject(s)
Sex Differentiation , Testosterone/physiology , Androgen-Insensitivity Syndrome/metabolism , Androgens/metabolism , Androgens/physiology , Animals , Erythropoiesis , Estradiol/physiology , Humans , Kidney/metabolism , Liver/metabolism , Male , Mice , Muscles/metabolism , Progestins/physiology , Proteins/metabolism , Rats , Receptors, Androgen/metabolism , Testosterone/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL