Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Sex Dev ; 16(4): 252-260, 2022.
Article in English | MEDLINE | ID: mdl-35764069

ABSTRACT

INTRODUCTION: NR5A1 is an essential transcription factor that regulates several target genes involved in reproduction and endocrine function. Pathogenic variants in this gene are responsible for a wide spectrum of disorders/differences of sex development (DSD). METHODS: The molecular study involved Sanger sequencing, in vitro assays, and whole exome sequencing (WES). RESULTS: Four variants were identified within the NR5A1 non-coding region in 3 patients with 46,XY DSD. In vitro analyses showed that promoter activity was affected in all cases. WES revealed variants in SRA1, WWOX, and WDR11 genes. DISCUSSION/CONCLUSION: Evaluation of clinical and phenotypic significance of variants located in a non-coding region of a gene can be complex, and little is known regarding their association with DSD. Nevertheless, based on the important region for interaction with cofactors essential to promote appropriated sex development and on our in vitro results, it is feasible to say that an impact on gene expression can be expected and that this may be correlated with the DSD pathophysiology presented in our patients. Considering the number of cases that remain elusive after screening for the well-known DSD related genes, we emphasize the importance of a careful molecular analysis of NR5A1 non-coding region which is commonly neglected and might explain some idiopathic DSD cases.


Subject(s)
Disorder of Sex Development, 46,XY , Disorders of Sex Development , Humans , Mutation , Disorder of Sex Development, 46,XY/genetics , Phenotype , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Sexual Development/genetics , Disorders of Sex Development/genetics
2.
Genes (Basel) ; 13(4)2022 03 29.
Article in English | MEDLINE | ID: mdl-35456418

ABSTRACT

Premature ovarian insufficiency (POI) is one of the main causes of female premature infertility. POI is a genetically heterogeneous disorder with a complex molecular etiology; as such, the genetic causes remain unknown in the majority of patients. Therefore, this study aimed to identify mutations and characterize the associated molecular contribution of gonadogenesis-determinant genes to POI. Genomic assays, including PCR-SSCP and Sanger sequencing, followed by in silico analyses were used to investigate the underpinnings of ovarian deficiency in 11 women affected by POI. Large deletions and nucleotide insertions and duplications were excluded by PCR. Thirteen genetic variants were identified in the WT1 (c.213G>T, c.609T>C, c.873A>G, c.1122G>A), NR0B1 (c.353C>T, c.425G>A), NR5A1 (c.437G>C, IVS4-20C>T), LHX9 (IVS2-12G>C, IVS3+13C>T, c.741T>C), ZNF275 (c.969C>T), and NRIP1 (c.3403C>T) genes. Seven novel genetic variants and five unpublished substitutions were identified. No genetic aberrations were detected in the ZFP92 and INSL3 genes. Each variant was genotyped using PCR-SSCP in 100 POI-free subjects, and their allelic frequencies were similar to the patients. These analyses indicated that allelic variation in the WT1, NR0B1, NR5A1, LHX9, ZFP92, ZNF275, INSL3, and NRIP1 genes may be a non-disease-causing change or may not contribute significantly to the genetics underlying POI disorders. Findings support the polygenic nature of this clinical disorder, with the SNVs identified representing only a probable contribution to the variability of the human genome.


Subject(s)
Infertility, Female , Primary Ovarian Insufficiency , Alleles , Cohort Studies , DAX-1 Orphan Nuclear Receptor/genetics , Female , Humans , Infertility, Female/genetics , LIM-Homeodomain Proteins/genetics , Male , Mutation , Mutation, Missense , Primary Ovarian Insufficiency/genetics , Steroidogenic Factor 1/genetics , Transcription Factors/genetics , WT1 Proteins/genetics
3.
Sex Dev ; 16(4): 242-251, 2022.
Article in English | MEDLINE | ID: mdl-36657429

ABSTRACT

INTRODUCTION: Ovotesticular disorder of sex development (OT-DSD) is a rare condition defined by concomitance of testicular tissue and ovarian tissue (containing follicles) in the same individual. In SRY-negative 46,XX OT-DSD, the presence of testicular tissue may be due to variations in NR5A1. Our aims were to search for NR5A1 variants in SRY-negative 46,XX OT-DSD patients and to perform a systematic review on the contribution of NR5A1 variations to 46,XX OT-DSD. METHODS: Sanger sequencing of NR5A1 was performed in seven SRY-negative 46,XX OT-DSD patients: five simplex cases and two with another sibling with a 46,XX DSD. Systematic review of original studies on NR5A1 sequencing of 46,XX OT-DSD patients was performed according to PRISMA-P guideline. Case reports were selected for analysis of clinical features. Individuals with NR5A1-associated testicular DSD were not included. RESULTS: Sanger sequencing of NR5A1 did not reveal pathogenic variants among our patients. Our cohort was included in this systematic review with seven other articles, totalizing fifty-six 46,XX OT-DSD patients investigated by Sanger or whole-exome sequencing. From them, three NR5A1 pathogenic variants were identified (5% of the cases). Clinical analysis of these 3 cases and 5 case reports revealed: predominance of ovotestis (13/16 gonads) and bilateral OT-DSD (5/8 cases). CONCLUSION: The etiology of most 46,XX OT-DSD cases remains elusive, highlighting the importance of a deeper molecular investigation.


Subject(s)
Disorders of Sex Development , Ovotesticular Disorders of Sex Development , Steroidogenic Factor 1 , Humans , Male , Disorders of Sex Development/genetics , Disorders of Sex Development/pathology , Gonads , Meta-Analysis as Topic , Ovotesticular Disorders of Sex Development/genetics , Ovotesticular Disorders of Sex Development/pathology , Steroidogenic Factor 1/genetics , Testis/pathology
4.
Anim. Reprod. (Online) ; 19(3): e20220011, set. 2022. ilus
Article in English | VETINDEX | ID: biblio-1393197

ABSTRACT

Sex steroid hormones are critical in gonadal differentiation in turtles. The gonads are not the only organs responsible for producing these hormones during this phase. Mesonephros play an important role in steroidogenesis. The present study aimed to investigate the presence of steroidogenic cells in mesonephros of Podocnemis expansa during gonadal differentiation and to evaluate their morphology and ultrastructure. Ten embryos of P. expansa were collected from 5 nests on day 36 of incubation, during spawning period on an artificial beach. Embryos were extracted from eggs by slicing the shell and euthanized. They were dissected under a stereoscopic microscope to collect the gonad-mesonephro complex, in which were fixed and subsequently processed for light microscopy, immunohistochemistry and transmission electron microscopy analysis. During histological analysis was observed mesonephros has typical morphological structure. Immunohistochemistry showed immunoreaction to aromatase in cells of intertubular space. Confirming these findings, it was possible to observe a type of intertubular cell in several regions of mesonephro, being more predominant in region close to blood vessels, distal and proximal tubules. In ultrastructural analysis these cells were characterized by having a clear, large, and rounded nucleus with evident nucleolus and cytoplasm rich in electron-dense droplets. This study demonstrated for the first time the presence of cells with morphological, immunohistochemical and ultrastructural characteristics similar to steroid-producing cells in P. expansa mesonephrons, suggesting that this organ may contribute to gonadal differentiation in this species.(AU)


Subject(s)
Animals , Turtles/physiology , Sex Determination Processes/physiology , Steroidogenic Factor 1/analysis , Aromatase/immunology
5.
Diabetes ; 70(7): 1498-1507, 2021 07.
Article in English | MEDLINE | ID: mdl-33883215

ABSTRACT

Leptin plays an important role in the protection against diet-induced obesity (DIO) by its actions in ventromedial hypothalamic (VMH) neurons. However, little is known about the intracellular mechanisms involved in these effects. To assess the role of the STAT3 and ERK2 signaling in neurons that express the steroidogenic factor 1 (SF1) in the VMH in energy homeostasis, we used cre-lox technology to generate male and female mice with specific disruption of STAT3 or ERK2 in SF1 neurons of the VMH. We demonstrated that the conditional knockout of STAT3 in SF1 neurons of the VMH did not affect body weight, food intake, energy expenditure, or glucose homeostasis in animals on regular chow. However, with high-fat diet (HFD) challenge, loss of STAT3 in SF1 neurons caused a significant increase in body weight, food intake, and energy efficiency that was more remarkable in females, which also showed a decrease in energy expenditure. In contrast, deletion of ERK2 in SF1 neurons of VMH did not have any impact on energy homeostasis in both regular diet and HFD conditions. In conclusion, STAT3 but not ERK2 signaling in SF1 neurons of VMH plays a crucial role in protection against DIO in a sex-specific pattern.


Subject(s)
Diet, High-Fat , Mitogen-Activated Protein Kinase 1/physiology , Obesity/prevention & control , STAT3 Transcription Factor/physiology , Ventromedial Hypothalamic Nucleus/physiology , Animals , Energy Metabolism , Female , Male , Mice , Mice, Inbred C57BL , RNA Splicing Factors/physiology , Sex Characteristics , Steroidogenic Factor 1/physiology
6.
Peptides ; 135: 170426, 2021 01.
Article in English | MEDLINE | ID: mdl-33069692

ABSTRACT

The hypothalamus mediates important exercise-induced metabolic adaptations, possibly via hormonal signals. Hypothalamic leptin receptor (LepR)- and steroidogenic factor 1 (SF1)-expressing neurons are directly responsive to growth hormone (GH) and deletion of GH receptor (GHR) in these cells impairs neuroendocrine responses during situations of metabolic stress. In the present study, we determined whether GHR ablation in LepR- or SF1-expressing cells modifies acute and chronic metabolic adaptations to exercise. Male mice carrying deletion of GHR in LepR- or SF1-expressing cells were submitted to 8 weeks of treadmill running training. Changes in aerobic performance and exercise-induced metabolic adaptations were determined. Mice carrying GHR deletion in LepR cells showed increased aerobic performance after 8 weeks of treadmill training, whereas GHR ablation in SF1 cells prevented improvement in running capacity. Trained mice carrying GHR ablation in SF1 cells exhibited increased fat mass and reduced cross-sectional area of the gastrocnemius muscle. In contrast, deletion of GHR in LepR cells reduced fat mass and increased gastrocnemius muscle hypertrophy, energy expenditure and voluntary locomotor activity in trained mice. Although glucose tolerance was not significantly affected by targeted deletions, glycemia before and immediately after maximum running tests was altered by GHR ablation. In conclusion, GHR signaling in hypothalamic neurons regulates the adaptation capacity to aerobic exercise in a cell-specific manner. These findings suggest that GH may represent a hormonal cue that informs specific hypothalamic neurons to produce exercise-induced acute and chronic metabolic adaptations.


Subject(s)
Exercise/physiology , Physical Conditioning, Animal , Receptors, Leptin/genetics , Receptors, Somatotropin/genetics , Steroidogenic Factor 1/genetics , Adaptation, Physiological/genetics , Animals , Energy Metabolism/genetics , Gene Expression Regulation , Growth Hormone/metabolism , Humans , Hypothalamus/metabolism , Leptin/genetics , Locomotion/genetics , Male , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neurons/metabolism
7.
Genes (Basel) ; 11(12)2020 11 27.
Article in English | MEDLINE | ID: mdl-33261069

ABSTRACT

BACKGROUND: Pituitary adenomas (PA) are the second most common tumor in the central nervous system and have low counts of mutated genes. Splicing occurs in 95% of the coding RNA. There is scarce information about the spliceosome and mRNA-isoforms in PA, and therefore we carried out proteomic and transcriptomic analysis to identify spliceosome components and mRNA isoforms in PA. METHODS: Proteomic profile analysis was carried out by nano-HPLC and mass spectrometry with a quadrupole time-of-flight mass spectrometer. The mRNA isoforms and transcriptomic profiles were carried out by microarray technology. With proteins and mRNA information we carried out Gene Ontology and exon level analysis to identify splicing-related events. RESULTS: Approximately 2000 proteins were identified in pituitary tumors. Spliceosome proteins such as SRSF1, U2AF1 and RBM42 among others were found in PA. These results were validated at mRNA level, which showed up-regulation of spliceosome genes in PA. Spliceosome-related genes segregate and categorize PA tumor subtypes. The PA showed alterations in CDK18 and THY1 mRNA isoforms which could be tumor specific. CONCLUSIONS: Spliceosome components are significant constituents of the PA molecular machinery and could be used as molecular markers and therapeutic targets. Splicing-related genes and mRNA-isoforms profiles characterize tumor subtypes.


Subject(s)
Adenoma/metabolism , Pituitary Neoplasms/metabolism , Proteome , Spliceosomes , Steroidogenic Factor 1/genetics , Transcription Factor Pit-1/genetics , Transcriptome , Adenoma/genetics , Adenoma/pathology , Alternative Splicing , Biomarkers, Tumor , Cell Lineage , Chromatography, High Pressure Liquid , Exons/genetics , Gene Ontology , Hormones/analysis , Humans , Nanotechnology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Principal Component Analysis , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Tandem Mass Spectrometry , Transcription Factors/analysis
8.
Hum Mutat ; 41(1): 58-68, 2020 01.
Article in English | MEDLINE | ID: mdl-31513305

ABSTRACT

Nuclear receptor subfamily 5 group A member 1 (NR5A1), also named steroidogenic factor 1, is an essential transcription factor that regulates a number of target genes crucial for normal reproductive physiology and endocrine function. It is encoded by NR5A1 gene and is expressed in high doses mainly in steroidogenic tissues, where it controls several steps of adrenal and gonadal development. NR5A1 mutations are associated with a wide phenotypic spectrum of disorders/differences of sex development (DSD), a group of conditions in which development of chromosomal, gonadal, or anatomic sex is atypical. Here, we reviewed 188 NR5A1 mutations from 238 cases reported in literature so far. Additionally, we report the variations p.Ser4*, p.(Cys55Ser), p.(Met78Leu), and p.Met98Glyfs*45, which have not been annotated for NR5A1 before and were identified in some of the 205 46,XY patients of our own cohort. This is the first NR5A1 mutation review which includes both 46,XX and 46,XY karyotype, with the purpose of discussing the complexity of genotype-phenotype correlations among DSD and infertile male patients and also females with primary ovarian failure.


Subject(s)
Disorders of Sex Development/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Infertility/genetics , Mutation , Steroidogenic Factor 1/genetics , Adolescent , Alleles , Child , Child, Preschool , Databases, Genetic , Disorder of Sex Development, 46,XY/diagnosis , Disorder of Sex Development, 46,XY/genetics , Disorders of Sex Development/diagnosis , Female , Genetic Association Studies/methods , Genotype , Humans , Infant , Infant, Newborn , Infertility/diagnosis , Karyotype , Male , Phenotype
9.
FASEB J ; 33(11): 11909-11924, 2019 11.
Article in English | MEDLINE | ID: mdl-31366244

ABSTRACT

Growth hormone (GH) is secreted during hypoglycemia, and GH-responsive neurons are found in brain areas containing glucose-sensing neurons that regulate the counter-regulatory response (CRR). However, whether GH modulates the CRR to hypoglycemia via specific neuronal populations is currently unknown. Mice carrying ablation of GH receptor (GHR) either in leptin receptor (LepR)- or steroidogenic factor-1 (SF1)-expressing cells were studied. We also investigated the importance of signal transducer and activator of transcription 5 (STAT5) signaling in SF1 cells for the CRR. GHR ablation in LepR cells led to impaired capacity to recover from insulin-induced hypoglycemia and to a blunted CRR caused by 2-deoxy-d-glucose (2DG) administration. GHR inactivation in SF1 cells, which include ventromedial hypothalamic neurons, also attenuated the CRR. The reduced CRR was prevented by parasympathetic blockers. Additionally, infusion of 2DG produced an abnormal hyperactivity of parasympathetic preganglionic neurons, whereas the 2DG-induced activation of anterior bed nucleus of the stria terminalis neurons was reduced in mice without GHR in SF1 cells. Mice carrying ablation of Stat5a/b genes in SF1 cells showed no defects in the CRR. In summary, GHR expression in SF1 cells is required for a normal CRR, and these effects are largely independent of STAT5 pathway.-Furigo, I. C., de Souza, G. O., Teixeira, P. D. S., Guadagnini, D., Frazão, R., List, E. O., Kopchick, J. J., Prada, P. O., Donato, J., Jr. Growth hormone enhances the recovery of hypoglycemia via ventromedial hypothalamic neurons.


Subject(s)
Growth Hormone/pharmacology , Hypoglycemia/drug therapy , Hypothalamus/drug effects , Neurons/drug effects , Recovery of Function/drug effects , Animals , Deoxyglucose/pharmacology , Hypoglycemia/physiopathology , Hypothalamus/cytology , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/physiology , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism
10.
Biol Reprod ; 99(6): 1303-1312, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29985989

ABSTRACT

Testicular anti-Müllerian hormone (AMH) production is inhibited by androgens around pubertal onset, as observed under normal physiological conditions and in patients with precocious puberty. In agreement, AMH downregulation is absent in patients with androgen insensitivity. The molecular mechanisms underlying the negative regulation of AMH by androgens remain unknown. Our aim was to elucidate the mechanisms through which androgens downregulate AMH expression in the testis. A direct negative effect of androgens on the transcriptional activity of the AMH promoter was found using luciferase reporter assays in the mouse prepubertal Sertoli cell line SMAT1. A strong inhibition of AMH promoter activity was seen in the presence of both testosterone and DHT and of the androgen receptor. By site-directed mutagenesis and chromatin immunoprecipitation assays, we showed that androgen-mediated inhibition involved the binding sites for steroidogenic factor 1 (SF1) present in the proximal promoter of the AMH gene. In this study, we describe for the first time the mechanism behind AMH inhibition by androgens, as seen in physiological and pathological conditions in males. Inhibition of AMH promoter activity by androgens could be due to protein-protein interactions between the ligand-bound androgen receptor and SF1 or by blockage of SF1 binding to its sites on the AMH promoter.


Subject(s)
Androgens/pharmacology , Anti-Mullerian Hormone/metabolism , Sertoli Cells/physiology , Steroidogenic Factor 1/metabolism , Animals , Anti-Mullerian Hormone/genetics , Cell Line , Chromatin Immunoprecipitation , Down-Regulation , Humans , Immunohistochemistry , Male , Mice , Promoter Regions, Genetic , Receptors, Androgen/metabolism , Steroidogenic Factor 1/genetics , Transcriptome
11.
Mol Hum Reprod ; 24(4): 203-210, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29438521

ABSTRACT

STUDY QUESTION: Is the expression of steroidogenic enzyme 17α-Hydroxylase/17,20-Lyase (CYP17A1) down-regulated in Leydig cells (LCs) of men with spermatogenic failure and compensated impairment of LC function, i.e. a low testosterone to LH (T/LH) ratio? SUMMARY ANSWER: Although the transcriptional expression of CYP17A1 is increased, its protein expression is decreased, in isolated LCs of men with spermatogenic failure and reduced serum T/LH. WHAT IS KNOWN ALREADY: Primary spermatogenic defects have been associated with functional and morphological abnormalities of LCs, characterized by decreased serum testosterone (T) levels, decreased T/LH, increased 17ß-estradiol (E2) and E2/T ratio, and larger clusters of LCs (LC hyperplasia). CYP17A1 is a key enzyme in the testosterone pathway and has been implicated in the steroidogenic lesion produced by E2 stimulation. STUDY DESIGN, SIZE, DURATION: We studied 18 azoospermic patients with Sertoli cell-only syndrome (SCOS) and signs of LC dysfunction (cases) and 10 obstructive azoospermic/oligozoospermic men with normal spermatogenesis (controls). The SCOS patients were sub-grouped into 9 cases with T/LH <2 and 9 cases with T/LH ≥2. All of the men underwent testicular biopsy for sperm retrieval at the Reproductive Unit of a University Hospital. PARTICIPANTS/MATERIALS, SETTING, METHODS: The transcriptional expression of CYP17A1 and SF-1 (steroidogenic factor 1) was quantified by SYBR®Green-based qPCR in LCs isolated by laser capture microdissection (LCM), and relative expression to the control pool was assessed. CYP17A1 protein expression was semi-quantified by indirect immunofluorescence (IFI) using Image-Pro Plus v7.0 (Media Cybernetics) in testicular tissue. FSH and LH serum concentrations, and serum and intratesticular T (ITT) and E2 (ITE2) were measured by IRMA and RIA, respectively. MAIN RESULTS AND THE ROLE OF CHANCE: Relative CYP17A1 mRNA expression was increased in cases with T/LH <2 compared to cases with T/LH ≥2, by a mean of 3.3-fold (P = 0.002). No corresponding increase in protein expression was found; in fact, CYP17A1 immunostaining intensity assessed by the Integrated Optical Density (IOD) parameter was lower in the cases with T/LH <2 compared to controls (P = 0.008). Relative SF-1 mRNA expression was similar in both case subgroups. CYP17A1 mRNA expression correlated with ITE2 and intratesticular E2/T (r = 0.536; P = 0.026 and r = 0.542; P = 0.016, respectively), while an inverse association was observed for ITE2 and protein level expression (r = -0.421; P = 0.05). LARGE SCALE DATA: Not applicable. LIMITATIONS REASONS FOR CAUTION: We should interpret the results of the semi-quantification of immunofluorescent staining by Image-Pro Plus software with caution, because it is a semi-quantitative method that may have certain difficulties regarding the disposition of protein in the cells. However, it is not influenced by variations in the number of cells that express the protein, as could be the case of western blot analysis in testicular tissue. WIDER IMPLICATIONS OF THE FINDINGS: Dysfunctional LCs of men with SCOS show post-transcriptional deregulation of CYP17A1, with increased mRNA and decreased protein expression, which may be modulated by increased ITE2 levels. In addition, transcriptional expression of CYP17A1 was not associated with changes in SF-1 mRNA expression. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the National Fund for Scientific and Technological Development (FONDECYT) of Chile to A.C. [grant number 1120176]. The authors declare no conflict of interest.


Subject(s)
Leydig Cells/metabolism , Sertoli Cell-Only Syndrome/metabolism , Steroid 17-alpha-Hydroxylase/metabolism , Humans , Luteinizing Hormone/metabolism , Male , Steroidogenic Factor 1/metabolism , Testosterone/metabolism
12.
J Pediatr Endocrinol Metab ; 31(2): 191-194, 2018 Jan 26.
Article in English | MEDLINE | ID: mdl-29306929

ABSTRACT

BACKGROUND: Inhibin B is a hormone produced by the Sertoli cells that can provide important information for the investigation of disorders of sex development (DSD) with 46,XY karyotype. The aim of this study is to compare two enzyme-linked immunosorbent assay (ELISA) assays for dosage of serum inhibin B in patients with 46,XY DSD with normal testosterone secretion. METHODS: Twenty-nine patients with 46,XY DSD and normal testosterone secretion (partial androgen insensitivity syndrome [PAIS] [n=8]; 5α-reductase deficiency [n=7] and idiopathic 46,XY DSD [n=14]) were included. Molecular analysis of the AR and SRD5A2 genes were performed in all patients and the NR5A1 gene analysis in the idiopathic group. Measurements of inhibin B were performed by two second-generation ELISA assays (Beckman-Coulter and AnshLabs). Assays were compared using the interclass correlation coefficient (ICC) and the Bland-Altman method. RESULTS: ICC was 0.915 [95% confidence interval (CI): 0.828-0.959], however, a discrepancy was observed between trials, which is more evident among higher values when analyzed by the Bland-Altman method. CONCLUSIONS: It is recommended to perform the inhibin B measurement always using the same ELISA kit when several evaluations are required for a specific patient.


Subject(s)
Disorder of Sex Development, 46,XY/blood , Enzyme-Linked Immunosorbent Assay , Inhibin-beta Subunits/blood , Testis/metabolism , Testosterone/metabolism , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/blood , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/deficiency , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Adolescent , Adult , Androgen-Insensitivity Syndrome/blood , Androgen-Insensitivity Syndrome/diagnosis , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/physiopathology , Child , Child, Preschool , Diagnosis, Differential , Disorder of Sex Development, 46,XY/diagnosis , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/physiopathology , Hospitals, University , Humans , Hypospadias/blood , Hypospadias/diagnosis , Hypospadias/genetics , Hypospadias/physiopathology , Karyotype , Male , Membrane Proteins/genetics , Outpatient Clinics, Hospital , Receptors, Androgen/genetics , Reproducibility of Results , Severity of Illness Index , Steroid Metabolism, Inborn Errors/blood , Steroid Metabolism, Inborn Errors/diagnosis , Steroid Metabolism, Inborn Errors/genetics , Steroid Metabolism, Inborn Errors/physiopathology , Steroidogenic Factor 1/genetics , Testis/physiopathology , Young Adult
13.
Hum Mutat ; 39(1): 114-123, 2018 01.
Article in English | MEDLINE | ID: mdl-29027717

ABSTRACT

Steroidogenic factor-1 (SF1), encoded by the NR5A1 gene, is a key regulator of steroidogenesis and reproductive development. NR5A1 mutations described in 46,XY patients with disorders of sex development (DSD) can be associated with a range of conditions of phenotypes; however, the genotype-phenotype correlation remains elusive in many cases. In the present study, we describe the impact of five NR5A1 variants (three novel: p.Arg39Cys, p.Ser32Asn, and p.Lys396Argfs*34; and two previously described: p.Cys65Tyr and p.Cys247*) on protein function, identified in seven patients with 46,XY DSD. In vitro functional analyses demonstrate that NR5A1 mutations impair protein functions and result in the DSD phenotype observed in our patients. Missense mutations in the DNA binding domain and the frameshift mutation p.Lys396Argfs*34 lead to both, markedly affected transactivation assays, and loss of DNA binding, whereas the mutation p.Cys247* retained partial transactivation capacity and the ability to bind a consensus SF1 responsive element. SF1 acts in a dose-dependent manner and regulates a cascade of genes involved in the sex determination and steroidogenesis, but in most cases reported so far, still lead to a sufficient adrenal steroidogenesis and function, just like in our cases, in which heterozygous mutations are associated to 46,XY DSD with intact adrenal steroid biosynthesis.


Subject(s)
Disorder of Sex Development, 46,XY/diagnosis , Disorder of Sex Development, 46,XY/genetics , Mutation , Phenotype , Steroidogenic Factor 1/genetics , Adolescent , Alleles , Amino Acid Substitution , Child , Child, Preschool , Female , Genetic Association Studies , Genotype , Humans , Infant , Infant, Newborn , Male , Models, Molecular , Protein Conformation , Sequence Analysis, DNA , Steroidogenic Factor 1/chemistry , Structure-Activity Relationship , Young Adult
14.
Neuroscience ; 365: 114-124, 2017 Dec 04.
Article in English | MEDLINE | ID: mdl-28987511

ABSTRACT

Previous studies have shown that leptin resistance is a key feature that leads to gestational metabolic adaptions. We hypothesized that leptin sensitivity in the ventromedial nucleus of the hypothalamus (VMH) plays a critical role regulating gestational metabolic changes. In the present study, we generated a mouse model carrying ablation of the suppressor of cytokine signaling 3 (SOCS3) in steroidogenic factor-1 (SF1) cells, which include the VMH, in order to investigate whether increased leptin sensitivity in this neuronal population prevents at least part of the metabolic changes typically observed during gestation and lactation. As predicted by the inhibitory effects of SOCS3 in leptin signaling, pregnant SF1 SOCS3 KO mice exhibited increased leptin sensitivity in the VMH, since an acute leptin injection induced a 95% increase in the STAT3 phosphorylation in this nucleus, compared to control animals (p = 0.02). Despite that, SF1 SOCS3 KO mice showed similar weight gain, food intake, hypothalamic neuropeptide expression and serum leptin levels during pregnancy compared to control littermates. Unexpectedly, SF1 SOCS3 KO mice exhibited glucose intolerance during pregnancy. SF1 SOCS3 KO mice also presented a lower body weight (-3%; p < 0.05) during mid and late lactation, although food intake, litter size and offspring growth were not affected. Our findings suggest that increased leptin sensitivity in the VMH causes modest metabolic effects and is not sufficient to prevent major metabolic adaptations of pregnancy and lactation.


Subject(s)
Lactation/metabolism , Neurons/metabolism , Pregnancy/metabolism , Steroidogenic Factor 1/metabolism , Suppressor of Cytokine Signaling 3 Protein/deficiency , Adiposity/drug effects , Adiposity/genetics , Animals , Body Weight/drug effects , Energy Metabolism/drug effects , Energy Metabolism/genetics , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glucose Tolerance Test , Insulin/metabolism , Lactation/drug effects , Leptin/pharmacology , Mice , Mice, Transgenic , Neurons/drug effects , RNA, Messenger/metabolism , Steroidogenic Factor 1/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Ventromedial Hypothalamic Nucleus/cytology
15.
J Endocrinol ; 235(3): 207-222, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28899903

ABSTRACT

Many hormones/cytokines are secreted in response to exercise and cytokine signaling may play a pivotal role in the training adaptations. To investigate the importance of cytokine signaling during vertical ladder climbing, a resistance exercise model, we produced mice lacking SOCS3 protein exclusively in steroidogenic factor-1 (SF1) cells (SF1 Socs3 KO mice). SF1 expression is found in steroidogenic cells of the adrenal cortex and gonads, as well as in neurons of the ventromedial nucleus of the hypothalamus. Histological markers of the fetal adrenal zone (or X-zone in rodents) were still present in adult males and postpartum SF1 Socs3 KO females, suggesting a previously unrecognized effect of SOCS3 on the terminal differentiation of the adrenal gland. This change led to a distinct distribution of lipid droplets along the adrenal cortex. Under basal conditions, adult SF1 Socs3 KO mice exhibited similar adrenal weight, and plasma ACTH and corticosterone concentrations. Nonetheless, SF1 Socs3 KO mice exhibited a blunted ACTH-induced corticosterone secretion. The overall metabolic responses induced by resistance training remained unaffected in SF1 Socs3 KO mice, including changes in body adiposity, glucose tolerance and energy expenditure. However, training performance and glucose control during intense resistance exercise were impaired in SF1 Socs3 KO mice. Furthermore, a reduced counter-regulatory response to 2-deoxy-d-glucose was observed in mutant mice. These findings revealed a novel participation of SOCS3 regulating several endocrine and metabolic aspects. Therefore, cytokine signaling in SF1 cells exerts an important role to sustain training performance possibly by promoting the necessary metabolic adjustments during exercise.


Subject(s)
Cell Differentiation/physiology , Steroidogenic Factor 1/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Adiposity/genetics , Adiposity/physiology , Adrenal Glands/metabolism , Animals , Cell Differentiation/genetics , Corticosterone/metabolism , Deoxyglucose/metabolism , Female , Male , Mice , Mice, Knockout , Pituitary Gland/metabolism , Steroidogenic Factor 1/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Testis/metabolism , Testosterone/metabolism
16.
Clinics (Sao Paulo) ; 72(6): 391-394, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28658440

ABSTRACT

OBJECTIVES:: Transcription Factor 21 represses steroidogenic factor 1, a nuclear receptor required for gonadal development, sex determination and the regulation of adrenogonadal steroidogenesis. The aim of this study was to investigate whether silencing or overexpression of the gene Transcription Factor 21 could modulate the gene and protein expression of steroidogenic factor 1 in adrenocortical tumors. METHODS:: We analyzed the gene expression of steroidogenic factor 1 using qPCR after silencing endogenous Transcription Factor 21 in pediatric adrenal adenoma-T7 cells through small interfering RNA. In addition, using overexpression of Transcription Factor 21 in human adrenocortical carcinoma cells, we analyzed the protein expression of steroidogenic factor 1 using Western blotting. RESULTS:: Transcription Factor 21 knockdown increased the mRNA expression of steroidogenic factor 1 by 5.97-fold in pediatric adrenal adenoma-T7 cells. Additionally, Transcription Factor 21 overexpression inhibited the protein expression of steroidogenic factor 1 by 0.41-fold and 0.64-fold in two different adult adrenocortical carcinoma cell cultures, H295R and T36, respectively. CONCLUSIONS:: Transcription Factor 21 is downregulated in adrenocortical carcinoma cells. Taken together, these findings support the hypothesis that Transcription Factor 21 is a regulator of steroidogenic factor 1 and is a tumor suppressor gene in pediatric and adult adrenocortical tumors.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/genetics , Steroidogenic Factor 1/metabolism , Adrenal Cortex Neoplasms/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Line, Tumor , Down-Regulation , Humans , Immunoblotting , Real-Time Polymerase Chain Reaction , Steroidogenic Factor 1/genetics
17.
Clinics ; Clinics;72(6): 391-394, June 2017. graf
Article in English | LILACS | ID: biblio-840089

ABSTRACT

OBJECTIVES: Transcription Factor 21 represses steroidogenic factor 1, a nuclear receptor required for gonadal development, sex determination and the regulation of adrenogonadal steroidogenesis. The aim of this study was to investigate whether silencing or overexpression of the gene Transcription Factor 21 could modulate the gene and protein expression of steroidogenic factor 1 in adrenocortical tumors. METHODS: We analyzed the gene expression of steroidogenic factor 1 using qPCR after silencing endogenous Transcription Factor 21 in pediatric adrenal adenoma-T7 cells through small interfering RNA. In addition, using overexpression of Transcription Factor 21 in human adrenocortical carcinoma cells, we analyzed the protein expression of steroidogenic factor 1 using Western blotting. RESULTS: Transcription Factor 21 knockdown increased the mRNA expression of steroidogenic factor 1 by 5.97-fold in pediatric adrenal adenoma-T7 cells. Additionally, Transcription Factor 21 overexpression inhibited the protein expression of steroidogenic factor 1 by 0.41-fold and 0.64-fold in two different adult adrenocortical carcinoma cell cultures, H295R and T36, respectively. CONCLUSIONS: Transcription Factor 21 is downregulated in adrenocortical carcinoma cells. Taken together, these findings support the hypothesis that Transcription Factor 21 is a regulator of steroidogenic factor 1 and is a tumor suppressor gene in pediatric and adult adrenocortical tumors.


Subject(s)
Humans , Adrenal Cortex Neoplasms/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/genetics , Steroidogenic Factor 1/metabolism , Adrenal Cortex Neoplasms/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Line, Tumor , Down-Regulation , Immunoblotting , Real-Time Polymerase Chain Reaction , Steroidogenic Factor 1/genetics
18.
Birth Defects Res C Embryo Today ; 108(4): 309-320, 2016 12.
Article in English | MEDLINE | ID: mdl-28033660

ABSTRACT

Steroidogenic factor 1 (NR5A1, SF-1, Ad4BP) is a transcriptional regulator of genes involved in adrenal and gonadal development and function. Mutations in NR5A1 have been among the most frequently identified genetic causes of gonadal development disorders and are associated with a wide phenotypic spectrum. In 46,XY individuals, NR5A1-related phenotypes may range from disorders of sex development (DSD) to oligo/azoospermia, and in 46,XX individuals, from 46,XX ovotesticular and testicular DSD to primary ovarian insufficiency (POI). The most common 46,XY phenotype is atypical or female external genitalia with clitoromegaly, palpable gonads, and absence of Müllerian derivatives. Notably, an undervirilized external genitalia is frequently seen at birth, while spontaneous virilization may occur later, at puberty. In 46,XX individuals, NR5A1 mutations are a rare genetic cause of POI, manifesting as primary or secondary amenorrhea, infertility, hypoestrogenism, and elevated gonadotropin levels. Mothers and sisters of 46,XY DSD patients carrying heterozygous NR5A1 mutations may develop POI, and therefore require appropriate counseling. Moreover, the recurrent heterozygous p.Arg92Trp NR5A1 mutation is associated with variable degrees of testis development in 46,XX patients. A clear genotype-phenotype correlation is not seen in patients bearing NR5A1 mutations, suggesting that genetic modifiers, such as pathogenic variants in other testis/ovarian-determining genes, may contribute to the phenotypic expression. Here, we review the published literature on NR5A1-related disease, and discuss our findings at a single tertiary center in Brazil, including ten novel NR5A1 mutations identified in 46,XY DSD patients. The ever-expanding phenotypic range associated with NR5A1 variants in XY and XX individuals confirms its pivotal role in reproductive biology, and should alert clinicians to the possibility of NR5A1 defects in a variety of phenotypes presenting with gonadal dysfunction. Birth Defects Research (Part C) 108:309-320, 2016. © 2016 The Authors Birth Defects Research Part C: Embryo Today: Reviews Published by Wiley Periodicals, Inc.


Subject(s)
Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/physiology , Adolescent , Adrenal Insufficiency , Adult , Brazil , Child , Child, Preschool , Disorders of Sex Development/genetics , Disorders of Sex Development/metabolism , Female , Gonadal Disorders/genetics , Gonadal Disorders/metabolism , Humans , Infant , Male , Mutation , Phenotype , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/metabolism , Steroidogenic Factor 1/metabolism
19.
Sex Dev ; 10(4): 191-199, 2016.
Article in English | MEDLINE | ID: mdl-27463801

ABSTRACT

Mutations in the NR5A1 gene, which encodes the steroidogenic factor 1 (SF1), are responsible for different phenotypes of disorders of sex development (DSD), such as bilateral anorchia and hypospadias. Furthermore, they can be associated with primary amenorrhea, premature ovarian failure, male infertility, adrenal tumors, and endometriosis. Direct sequencing of the 7 NR5A1 exons including ∼1,000 bp of the 5'-upstream and 3'-downstream regions and all intron-exon boundaries was performed in patients with DSD. Three different in silico tools were used to assess the consequences of a splice site mutation. As a result, 3 novel NR5A1 mutations were identified in 3 patients with 46,XY partial gonadal dysgenesis: p.Lys38* and p.Leu80Trpfs*8 lead to premature translation termination codons within the SF1 DNA-binding domain, and the intronic nucleotide substitution c.1138+1G>T at the intron 6 donor splice site is considered to modify correct splicing. We assume that the anomalous mRNA produced as a result of p.Lys38* and p.Leu80Trpfs*8 will be degraded by nonsense-mediated mRNA decay even before translation, leading to SF1 haploinsufficiency. The c.1138+1G>T mutation is expected to produce a truncated protein. Heterozygous SF1 loss-of-function mutations in these cases resulted in mild DSD manifestations, such as dysgenetic testes, spontaneous puberty, and preserved adrenal function.


Subject(s)
Disorder of Sex Development, 46,XY/genetics , Steroidogenic Factor 1/genetics , Codon, Nonsense/genetics , Exons/genetics , Gonadal Dysgenesis, 46,XY/genetics , Gonadal Dysgenesis, 46,XY/physiopathology , Gonads/metabolism , Gonads/physiology , Humans , Hypospadias/genetics , Hypospadias/physiopathology , Infant , Infertility, Male/genetics , Infertility, Male/physiopathology , Introns/genetics , Male , Mutation , Testis/abnormalities , Testis/physiopathology
20.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;48(12): 1087-1094, Dec. 2015. graf
Article in English | LILACS | ID: lil-762914

ABSTRACT

During gonad and adrenal development, the POD-1/capsulin/TCF21transcription factor negatively regulates SF-1/NR5A1expression, with higher SF-1 levels being associated with increased adrenal cell proliferation and tumorigenesis. In adrenocortical tumor cells, POD-1 binds to the SF-1 E-box promoter region, decreasing SF-1 expression. However, the modulation of SF-1 expression by POD-1 has not previously been described in normal adrenal cells. Here, we analyzed the basal expression of Pod-1 and Sf-1 in primary cultures of glomerulosa (G) and fasciculata/reticularis (F/R) cells isolated from male Sprague-Dawley rats, and investigated whether POD-1 overexpression modulates the expression of endogenous Sf-1 and its target genes in these cells. POD-1 overexpression, following the transfection of pCMVMycPod-1, significantly decreased the endogenous levels of Sf-1 mRNA and protein in F/R cells, but not in G cells, and also decreased the expression of the SF-1 target StAR in F/R cells. In G cells overexpressing POD-1, no modulation of the expression of SF-1 targets, StAR and CYP11B2, was observed. Our data showing that G and F/R cells respond differently to ectopic POD-1 expression emphasize the functional differences between the outer and inner zones of the adrenal cortex, and support the hypothesis that SF-1 is regulated by POD-1/Tcf21 in normal adrenocortical cells lacking the alterations in cellular physiology found in tumor cells.


Subject(s)
Animals , Male , Adrenal Cortex/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Phosphoproteins/metabolism , Steroidogenic Factor 1/metabolism , Adrenal Cortex/cytology , Basic Helix-Loop-Helix Transcription Factors/genetics , Electrophoresis, Polyacrylamide Gel , Gene Expression , Immunoblotting , Primary Cell Culture , Phosphoproteins/analysis , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , RNA, Messenger/analysis , Steroidogenic Factor 1/analysis , Zona Fasciculata/cytology , Zona Fasciculata/metabolism , Zona Glomerulosa/cytology , Zona Glomerulosa/metabolism , Zona Reticularis/cytology , Zona Reticularis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL