Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Genet ; 103(3): 277-287, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36349847

RESUMEN

46,XY gonadal dysgenesis (GD) is a Disorder/Difference of Sex Development (DSD) that can present with phenotypes ranging from ambiguous genitalia to complete male-to-female sex reversal. Around 50% of 46,XY DSD cases receive a molecular diagnosis. In mice, Fibroblast growth factor 9 (FGF9) is an important component of the male sex-determining pathway. Two FGF9 variants reported to date disrupt testis development in mice, but not in humans. Here, we describe a female patient with 46,XY GD harbouring the rare FGF9 variant (missense mutation), NM_002010.2:c.583G > A;p.(Asp195Asn) (D195N). By biochemical and cell-based approaches, the D195N variant disrupts FGF9 protein homodimerisation and FGF9-heparin-binding, and reduces both Sertoli cell proliferation and Wnt4 repression. XY Fgf9D195N/D195N foetal mice show a transient disruption of testicular cord development, while XY Fgf9D195N/- foetal mice show partial male-to-female gonadal sex reversal. In the general population, the D195N variant occurs at an allele frequency of 2.4 × 10-5 , suggesting an oligogenic basis for the patient's DSD. Exome analysis of the patient reveals several known and novel variants in genes expressed in human foetal Sertoli cells at the time of sex determination. Taken together, our results indicate that disruption of FGF9 homodimerization impairs testis determination in mice and, potentially, also in humans in combination with other variants.


Asunto(s)
Factor 9 de Crecimiento de Fibroblastos , Disgenesia Gonadal 46 XY , Humanos , Masculino , Femenino , Ratones , Animales , Dimerización , Factor 9 de Crecimiento de Fibroblastos/genética , Testículo , Gónadas , Disgenesia Gonadal 46 XY/genética
2.
Cell Mol Life Sci ; 79(10): 522, 2022 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-36114905

RESUMEN

The transcription factor SOX9 is essential for the development of multiple organs including bone, testis, heart, lung, pancreas, intestine and nervous system. Mutations in the human SOX9 gene led to campomelic dysplasia, a haploinsufficiency disorder with several skeletal malformations frequently accompanied by 46, XY sex reversal. The mechanisms underlying the diverse SOX9 functions during organ development including its post-translational modifications, the availability of binding partners, and tissue-specific accessibility to target gene chromatin. Here we summarize the expression, activities, and downstream target genes of SOX9 in molecular genetic pathways essential for organ development, maintenance, and function. We also provide an insight into understanding the mechanisms that regulate the versatile roles of SOX9 in different organs.


Asunto(s)
Organogénesis , Factor de Transcripción SOX9/metabolismo , Displasia Campomélica , Cromatina , Trastornos del Desarrollo Sexual/genética , Humanos , Masculino , Mutación , Factor de Transcripción SOX9/genética
3.
Hum Mol Genet ; 29(13): 2148-2161, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32452519

RESUMEN

In mice, male sex determination depends on FGF9 signalling via FGFR2c in the bipotential gonads to maintain the expression of the key testis gene SOX9. In humans, however, while FGFR2 mutations have been linked to 46,XY disorders of sex development (DSD), the role of FGF9 is unresolved. The only reported pathogenic mutations in human FGF9, FGF9S99N and FGF9R62G, are dominant and result in craniosynostosis (fusion of cranial sutures) or multiple synostoses (fusion of limb joints). Whether these synostosis-causing FGF9 mutations impact upon gonadal development and DSD etiology has not been explored. We therefore examined embryonic gonads in the well-characterized Fgf9 missense mouse mutants, Fgf9S99N and Fgf9N143T, which phenocopy the skeletal defects of FGF9S99N and FGF9R62G variants, respectively. XY Fgf9S99N/S99N and XY Fgf9N143T/N143T fetal mouse gonads showed severely disorganized testis cords and partial XY sex reversal at 12.5 days post coitum (dpc), suggesting loss of FGF9 function. By 15.5 dpc, testis development in both mutants had partly recovered. Mitotic analysis in vivo and in vitro suggested that the testicular phenotypes in these mutants arise in part through reduced proliferation of the gonadal supporting cells. These data raise the possibility that human FGF9 mutations causative for dominant skeletal conditions can also lead to loss of FGF9 function in the developing testis, at least in mice. Our data suggest that, in humans, testis development is largely tolerant of deleterious FGF9 mutations which lead to skeletal defects, thus offering an explanation as to why XY DSDs are rare in patients with pathogenic FGF9 variants.


Asunto(s)
Factor 9 de Crecimiento de Fibroblastos/genética , Trastornos Ovotesticulares del Desarrollo Sexual/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Sinostosis/genética , Animales , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Gónadas/crecimiento & desarrollo , Gónadas/patología , Humanos , Masculino , Ratones , Mutación Missense/genética , Trastornos Ovotesticulares del Desarrollo Sexual/patología , Factor de Transcripción SOX9/genética , Procesos de Determinación del Sexo/genética , Desarrollo Sexual/genética
4.
Proc Natl Acad Sci U S A ; 116(33): 16577-16582, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31371505

RESUMEN

Parkinson's disease (PD) is a debilitating neurodegenerative disorder caused by the loss of midbrain dopamine (DA) neurons. While the cause of DA cell loss in PD is unknown, male sex is a strong risk factor. Aside from the protective actions of sex hormones in females, emerging evidence suggests that sex-chromosome genes contribute to the male bias in PD. We previously showed that the Y-chromosome gene, SRY, directly regulates adult brain function in males independent of gonadal hormone influence. SRY protein colocalizes with DA neurons in the male substantia nigra, where it regulates DA biosynthesis and voluntary movement. Here we demonstrate that nigral SRY expression is highly and persistently up-regulated in animal and human cell culture models of PD. Remarkably, lowering nigral SRY expression with antisense oligonucleotides in male rats diminished motor deficits and nigral DA cell loss in 6-hydroxydopamine (6-OHDA)-induced and rotenone-induced rat models of PD. The protective effect of the SRY antisense oligonucleotides was associated with male-specific attenuation of DNA damage, mitochondrial degradation, and neuroinflammation in the toxin-induced rat models of PD. Moreover, reducing nigral SRY expression diminished or removed the male bias in nigrostriatal degeneration, mitochondrial degradation, DNA damage, and neuroinflammation in the 6-OHDA rat model of PD, suggesting that SRY directly contributes to the sex differences in PD. These findings demonstrate that SRY directs a previously unrecognized male-specific mechanism of DA cell death and suggests that suppressing nigral Sry synthesis represents a sex-specific strategy to slow or prevent DA cell loss in PD.


Asunto(s)
Genes Ligados a Y , Neuroprotección/genética , Enfermedad de Parkinson/genética , Animales , Daño del ADN , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/patología , Masculino , Mitofagia/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Oligonucleótidos Antisentido/farmacología , Oxidopamina , Enfermedad de Parkinson/fisiopatología , Ratas , Proteína de la Región Y Determinante del Sexo/genética , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
5.
Hum Genet ; 140(12): 1625-1634, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34524523

RESUMEN

ATR-X, an acronym for alpha thalassemia and mental retardation X-linked, syndrome is a congenital condition predominantly affecting males, characterized by mild to severe intellectual disability, facial, skeletal, urogenital, and hematopoietic anomalies. Less common are heart defects, eye anomalies, renal abnormalities, and gastrointestinal dysfunction. ATR-X syndrome is caused by germline variants in the ATRX gene. Until recently, the diagnosis of the ATR-X syndrome had been guided by the classical clinical manifestations and confirmed by molecular techniques. However, our new systematic analysis shows that the only clinical sign shared by all affected individuals is intellectual disability, with the other manifestations varying even within the same family. More than 190 different germline ATRX mutations in some 200 patients have been analyzed. With improved and more frequent analysis by molecular technologies, more subtle deletions and insertions have been detected recently. Moreover, emerging technologies reveal non-classic phenotypes of ATR-X syndrome as well as the description of a new clinical feature, the development of osteosarcoma which suggests an increased cancer risk in ATR-X syndrome. This review will focus on the different types of inherited ATRX mutations and their relation to clinical features in the ATR-X syndrome. We will provide an update of the frequency of clinical manifestations, the affected organs, and the genotype-phenotype correlations. Finally, we propose a shift in the diagnosis of ATR-X patients, from a clinical diagnosis to a molecular-based approach. This may assist clinicians in patient management, risk assessment and genetic counseling.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X/genética , Discapacidad Intelectual Ligada al Cromosoma X/fisiopatología , Discapacidad Intelectual Ligada al Cromosoma X/terapia , Talasemia alfa/genética , Talasemia alfa/fisiopatología , Talasemia alfa/terapia , Animales , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual Ligada al Cromosoma X/diagnóstico , Técnicas de Diagnóstico Molecular , Mutación , Talasemia alfa/diagnóstico
6.
Clin Genet ; 99(2): 325-329, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33174625

RESUMEN

Human multiple synostoses syndrome 3 is an autosomal dominant disorder caused by pathogenic variants in FGF9. Only two variants have been described in FGF9 in humans so far, and one in mice. Here we report a novel missense variant c.566C > G, p.(Pro189Arg) in FGF9. Functional studies showed this variant impairs FGF9 homodimerization, but not FGFR3c binding. We also review the findings of cases reported previously and report on additional features not described previously.


Asunto(s)
Factor 9 de Crecimiento de Fibroblastos/genética , Mutación Missense , Sinostosis/genética , Anomalías Múltiples/genética , Adolescente , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Heterocigoto , Humanos , Masculino , Fenotipo , Unión Proteica , Radiografía , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Sinostosis/diagnóstico por imagen , Sinostosis/patología
7.
Nucleic Acids Res ; 45(12): 7191-7211, 2017 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-28472341

RESUMEN

In mammalian embryonic gonads, SOX9 is required for the determination of Sertoli cells that orchestrate testis morphogenesis. To identify genetic networks directly regulated by SOX9, we combined analysis of SOX9-bound chromatin regions from murine and bovine foetal testes with sequencing of RNA samples from mouse testes lacking Sox9. We found that SOX9 controls a conserved genetic programme that involves most of the sex-determining genes. In foetal testes, SOX9 modulates both transcription and directly or indirectly sex-specific differential splicing of its target genes through binding to genomic regions with sequence motifs that are conserved among mammals and that we called 'Sertoli Cell Signature' (SCS). The SCS is characterized by a precise organization of binding motifs for the Sertoli cell reprogramming factors SOX9, GATA4 and DMRT1. As SOX9 biological role in mammalian gonads is to determine Sertoli cells, we correlated this genomic signature with the presence of SOX9 on chromatin in foetal testes, therefore equating this signature to a genomic bar code of the fate of foetal Sertoli cells. Starting from the hypothesis that nuclear factors that bind to genomic regions with SCS could functionally interact with SOX9, we identified TRIM28 as a new SOX9 partner in foetal testes.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Morfogénesis/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Factor de Transcripción SOX9/genética , Células de Sertoli/metabolismo , Transcriptoma , Animales , Bovinos , Cromatina/química , Cromatina/metabolismo , Embrión de Mamíferos , Femenino , Feto , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Redes Reguladoras de Genes , Masculino , Ratones , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas Represoras/metabolismo , Factor de Transcripción SOX9/metabolismo , Análisis de Secuencia de ARN , Células de Sertoli/citología , Procesos de Determinación del Sexo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína 28 que Contiene Motivos Tripartito
8.
Hum Mutat ; 39(12): 1861-1874, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30067310

RESUMEN

Nuclear receptor subfamily 5 group A member 1/Steroidogenic factor 1 (NR5A1; SF-1; Ad4BP) mutations cause 46,XY disorders of sex development (DSD), with phenotypes ranging from developmentally mild (e.g., hypospadias) to severe (e.g., complete gonadal dysgenesis). The molecular mechanism underlying this spectrum is unclear. During sex determination, SF-1 regulates SOX9 (SRY [sex determining region Y]-box 9) expression. We hypothesized that SF-1 mutations in 46,XY DSD patients affect SOX9 expression via the Testis-specific Enhancer of Sox9 core element, TESCO. Our objective was to assess the ability of 20 SF-1 mutants found in 46,XY DSD patients to activate TESCO. Patient DNA was sequenced for SF-1 mutations and mutant SF-1 proteins were examined for transcriptional activity, protein expression, sub-cellular localization and in silico structural defects. Fifteen of the 20 mutants showed reduced SF-1 activation on TESCO, 11 with atypical sub-cellular localization. Fourteen SF-1 mutants were predicted in silico to alter DNA, ligand or cofactor interactions. Our study may implicate aberrant SF-1-mediated transcriptional regulation of SOX9 in 46,XY DSDs.


Asunto(s)
Trastorno del Desarrollo Sexual 46,XY/genética , Elementos de Facilitación Genéticos , Mutación , Factor de Transcripción SOX9/genética , Factor Esteroidogénico 1/genética , Adolescente , Adulto , Niño , Preescolar , Simulación por Computador , Regulación de la Expresión Génica , Células HEK293 , Humanos , Lactante , Recién Nacido , Ligandos , Masculino , Unión Proteica , Análisis de Secuencia de ADN/métodos , Factor Esteroidogénico 1/química , Factor Esteroidogénico 1/metabolismo
9.
BMC Evol Biol ; 18(1): 3, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29338681

RESUMEN

BACKGROUND: Maleness in mammals is genetically determined by the Y chromosome. On the Y chromosome SRY is known as the mammalian male-determining gene. Both placental mammals (Eutheria) and marsupial mammals (Metatheria) have SRY genes. However, only eutherian SRY genes have been empirically examined by functional analyses, and the involvement of marsupial SRY in male gonad development remains speculative. RESULTS: In order to demonstrate that the marsupial SRY gene is similar to the eutherian SRY gene in function, we first examined the sequence differences between marsupial and eutherian SRY genes. Then, using a parsimony method, we identify 7 marsupial-specific ancestral substitutions, 13 eutherian-specific ancestral substitutions, and 4 substitutions that occurred at the stem lineage of therian SRY genes. A literature search and molecular dynamics computational simulations support that the lineage-specific ancestral substitutions might be involved with the functional differentiation between marsupial and eutherian SRY genes. To address the function of the marsupial SRY gene in male determination, we performed luciferase assays on the testis enhancer of Sox9 core (TESCO) using the marsupial SRY. The functional assay shows that marsupial SRY gene can weakly up-regulate the luciferase expression via TESCO. CONCLUSIONS: Despite the sequence differences between the marsupial and eutherian SRY genes, our functional assay indicates that the marsupial SRY gene regulates SOX9 as a transcription factor in a similar way to the eutherian SRY gene. Our results suggest that SRY genes obtained the function of male determination in the common ancestor of Theria (placental mammals and marsupials). This suggests that the marsupial SRY gene has a function in male determination, but additional experiments are needed to be conclusive.


Asunto(s)
Evolución Molecular , Genes sry , Marsupiales/genética , Procesos de Determinación del Sexo/genética , Secuencia de Aminoácidos , Animales , ADN/metabolismo , Genes Reporteros , Luciferasas/metabolismo , Masculino , Filogenia , Unión Proteica , Termodinámica
10.
Hum Mol Genet ; 24(23): 6699-710, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26362256

RESUMEN

Patients with 46,XY gonadal dysgenesis (GD) exhibit genital anomalies, which range from hypospadias to complete male-to-female sex reversal. However, a molecular diagnosis is made in only 30% of cases. Heterozygous mutations in the human FGFR2 gene cause various craniosynostosis syndromes including Crouzon and Pfeiffer, but testicular defects were not reported. Here, we describe a patient whose features we would suggest represent a new FGFR2-related syndrome, craniosynostosis with XY male-to-female sex reversal or CSR. The craniosynostosis patient was chromosomally XY, but presented as a phenotypic female due to complete GD. DNA sequencing identified the FGFR2c heterozygous missense mutation, c.1025G>C (p.Cys342Ser). Substitution of Cys342 by Ser or other amino acids (Arg/Phe/Try/Tyr) has been previously reported in Crouzon and Pfeiffer syndrome. We show that the 'knock-in' Crouzon mouse model Fgfr2c(C342Y/C342Y) carrying a Cys342Tyr substitution displays XY gonadal sex reversal with variable expressivity. We also show that despite FGFR2c-Cys342Tyr being widely considered a gain-of-function mutation, Cys342Tyr substitution in the gonad leads to loss of function, as demonstrated by sex reversal in Fgfr2c(C342Y/-) mice carrying the knock-in allele on a null background. The rarity of our patient suggests the influence of modifier genes which exacerbated the testicular phenotype. Indeed, patient whole exome analysis revealed several potential modifiers expressed in Sertoli cells at the time of testis determination in mice. In summary, this study identifies the first FGFR2 mutation in a 46,XY GD patient. We conclude that, in certain rare genetic contexts, maintaining normal levels of FGFR2 signaling is important for human testis determination.


Asunto(s)
Craneosinostosis/genética , Disgenesia Gonadal 46 XY/genética , Mutación Missense , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Adolescente , Animales , Craneosinostosis/metabolismo , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones , Ratones Mutantes , Síndrome
11.
Biol Reprod ; 92(6): 145, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25855264

RESUMEN

Male sex determination hinges on the development of testes in the embryo, beginning with the differentiation of Sertoli cells under the influence of the Y-linked gene SRY. Sertoli cells then orchestrate fetal testis formation including the specification of fetal Leydig cells (FLCs) that produce steroid hormones to direct virilization of the XY embryo. As the majority of XY disorders of sex development (DSDs) remain unexplained at the molecular genetic level, we reasoned that genes involved in FLC development might represent an unappreciated source of candidate XY DSD genes. To identify these genes, and to gain a more detailed understanding of the regulatory networks underpinning the specification and differentiation of the FLC population, we developed methods for isolating fetal Sertoli, Leydig, and interstitial cell-enriched subpopulations using an Sf1-eGFP transgenic mouse line. RNA sequencing followed by rigorous bioinformatic filtering identified 84 genes upregulated in FLCs, 704 genes upregulated in nonsteroidogenic interstitial cells, and 1217 genes upregulated in the Sertoli cells at 12.5 days postcoitum. The analysis revealed a trend for expression of components of neuroactive ligand interactions in FLCs and Sertoli cells and identified factors potentially involved in signaling between the Sertoli cells, FLCs, and interstitial cells. We identified 61 genes that were not known previously to be involved in specification or differentiation of FLCs. This dataset provides a platform for exploring the biology of FLCs and understanding the role of these cells in testicular development. In addition, it provides a basis for targeted studies designed to identify causes of idiopathic XY DSD.


Asunto(s)
Diferenciación Celular/genética , Células Intersticiales del Testículo/metabolismo , Células de Sertoli/metabolismo , Procesos de Determinación del Sexo/genética , Testículo/metabolismo , Transcriptoma , Animales , Feto/metabolismo , Células Intersticiales del Testículo/citología , Masculino , Ratones , Ratones Transgénicos , Células de Sertoli/citología , Testículo/citología
12.
Bioessays ; 34(6): 454-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22408002

RESUMEN

The SRY gene, which is located on the Y chromosome and directs male development, may promote aggression and other traditionally male behavioural traits, resulting in the fight-or-flight reaction to stress.


Asunto(s)
Catecolaminas/genética , Genes sry , Proteína de la Región Y Determinante del Sexo/genética , Animales , Catecolaminas/metabolismo , Regulación hacia Abajo , Humanos , Masculino , Modelos Animales , Proteína de la Región Y Determinante del Sexo/metabolismo , Cromosoma Y/genética , Cromosoma Y/metabolismo
13.
Biol Sex Differ ; 15(1): 24, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38520033

RESUMEN

BACKGROUND: Disorders/differences of sex development (DSD) are congenital conditions in which the development of chromosomal, gonadal, or anatomical sex is atypical. With overlapping phenotypes and multiple genes involved, poor diagnostic yields are achieved for many of these conditions. The current DSD diagnostic regimen can be augmented by investigating transcriptome/proteome in vivo, but it is hampered by the unavailability of affected gonadal tissue at the relevant developmental stage. We try to mitigate this limitation by reprogramming readily available skin tissue-derived dermal fibroblasts into Sertoli cells (SC), which could then be deployed for different diagnostic strategies. SCs form the target cell type of choice because they act like an organizing center of embryonic gonadal development and many DSD arise when these developmental processes go awry. METHODS: We employed a computational predictive algorithm for cell conversions called Mogrify to predict the transcription factors (TFs) required for direct reprogramming of human dermal fibroblasts into SCs. We established trans-differentiation culture conditions where stable transgenic expression of these TFs was achieved in 46, XY adult dermal fibroblasts using lentiviral vectors. The resulting Sertoli like cells (SLCs) were validated for SC phenotype using several approaches. RESULTS: SLCs exhibited Sertoli-like morphological and cellular properties as revealed by morphometry and xCelligence cell behavior assays. They also showed Sertoli-specific expression of molecular markers such as SOX9, PTGDS, BMP4, or DMRT1 as revealed by IF imaging, RNAseq and qPCR. The SLC transcriptome shared about two thirds of its differentially expressed genes with a human adult SC transcriptome and expressed markers typical of embryonic SCs. Notably, SLCs lacked expression of most markers of other gonadal cell types such as Leydig, germ, peritubular myoid or granulosa cells. CONCLUSIONS: The trans-differentiation method was applied to a variety of commercially available 46, XY fibroblasts derived from patients with DSD and to a 46, XX cell line. The DSD SLCs displayed altered levels of trans-differentiation in comparison to normal 46, XY-derived SLCs, thus showcasing the robustness of this new trans-differentiation model. Future applications could include using the SLCs to improve definitive diagnosis of DSD in patients with variants of unknown significance.


Individuals with disorders/differences of sex development (DSD) frequently do not get a specific genetic diagnostic. A limitation in the field is that the relevant cell types that would be needed to study the molecular events occurring at the time of onset of many DSD are found in the embryonic gonad. This, of course, is not accessible for research or diagnostic purposes. We set out to develop a method for directly transforming more accessible cells, from adult skin, into the cells known to organize the male gonad in the embryo, Sertoli cells. A combination of unique transcription factors was stably integrated into skin fibroblasts, and culture under appropriate conditions allowed differentiation into Sertoli-like cells (SLC), but not other gonadal cell types. The SLCs recapitulated known patterns of gene expression, shape, and behavior of Sertoli cells. The method was also tested on commercially available fibroblasts from a variety of DSD genetic backgrounds. The resulting cells exhibited condition-specific behavior (gene expression, adhesion to substrate, division rate…). This method provides a new tool to study molecular events occurring at the time of onset of DSD in the embryonic gonad, and the impact of patient-specific mutations on those. It could allow identification of new developmental pathways (and, thus, new candidate genes for DSD), as well as a provide models to validate the impact of variants of unknown significance, or to test approaches to correct the genetic anomaly in patient cells.


Asunto(s)
Gónadas , Células de Sertoli , Masculino , Adulto , Femenino , Humanos , Células de Sertoli/metabolismo , Diferenciación Celular , Transcriptoma
14.
Front Cell Dev Biol ; 12: 1337714, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38425503

RESUMEN

SOX9 is a key transcription factor for testis determination and development. Mutations in and around the SOX9 gene contribute to Differences/Disorders of Sex Development (DSD). However, a substantial proportion of DSD patients lack a definitive genetic diagnosis. SOX9 target genes are potentially DSD-causative genes, yet only a limited subset of these genes has been investigated during testis development. We hypothesize that SOX9 target genes play an integral role in testis development and could potentially be causative genes in DSD. In this study, we describe a novel testicular target gene of SOX9, Trpc3. Trpc3 exhibits high expression levels in the SOX9-expressing male Sertoli cells compared to female granulosa cells in mouse fetal gonads between embryonic day 11.5 (E11.5) and E13.5. In XY Sox9 knockout gonads, Trpc3 expression is markedly downregulated. Moreover, culture of E11.5 XY mouse gonads with TRPC3 inhibitor Pyr3 resulted in decreased germ cell numbers caused by reduced germ cell proliferation. Trpc3 is also expressed in endothelial cells and Pyr3-treated E11.5 XY mouse gonads showed a loss of the coelomic blood vessel due to increased apoptosis of endothelial cells. In the human testicular cell line NT2/D1, TRPC3 promotes cell proliferation and controls cell morphology, as observed by xCELLigence and HoloMonitor real-time analysis. In summary, our study suggests that SOX9 positively regulates Trpc3 in mouse testes and TRPC3 may mediate SOX9 function during Sertoli, germ and endothelial cell development.

15.
iScience ; 27(5): 109629, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38616920

RESUMEN

ATR-X (alpha thalassemia, mental retardation, X-linked) syndrome features genital and testicular abnormalities including atypical genitalia and small testes with few seminiferous tubules. Our mouse model recapitulated the testicular defects when Atrx was deleted in Sertoli cells (ScAtrxKO) which displayed G2/M arrest and apoptosis. Here, we investigated the mechanisms underlying these defects. In control mice, Sertoli cells contain a single novel "GATA4 PML nuclear body (NB)" that contained the transcription factor GATA4, ATRX, DAXX, HP1α, and PH3 and co-localized with the Y chromosome short arm (Yp). ScAtrxKO mice contain single giant GATA4 PML-NBs with frequent DNA double-strand breaks (DSBs) in G2/M-arrested apoptotic Sertoli cells. HP1α and PH3 were absent from giant GATA4 PML-NBs indicating a failure in heterochromatin formation and chromosome condensation. Our data suggest that ATRX protects a Yp region from DNA damage, thereby preventing Sertoli cell death. We discuss Y chromosome damage/decondensation as a mechanism for testicular failure.

16.
Hum Mol Genet ; 20(11): 2213-24, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21427128

RESUMEN

X-linked ATR-X (alpha thalassemia, mental retardation, X-linked) syndrome in males is characterized by mental retardation, facial dysmorphism, alpha thalassemia and urogenital abnormalities, including small testes. It is unclear how mutations in the chromatin-remodeling protein ATRX cause these highly specific clinical features, since ATRX is widely expressed during organ development. To investigate the mechanisms underlying the testicular defects observed in ATR-X syndrome, we generated ScAtrxKO (Sertoli cell Atrx knockout) mice with Atrx specifically inactivated in the supporting cell lineage (Sertoli cells) of the mouse testis. ScAtrxKO mice developed small testes and discontinuous tubules, due to prolonged G2/M phase and apoptosis of proliferating Sertoli cells during fetal life. Apoptosis might be a consequence of the cell cycle defect. We also found that the onset of spermatogenesis was delayed in postnatal mice, with a range of spermatogenesis defects evident in adult ScAtrxKO mice. ATRX and the androgen receptor (AR) physically interact in the testis and in the Sertoli cell line TM4 and co-operatively activate the promoter of Rhox5, an important direct AR target. We also demonstrate that ATRX directly binds to the Rhox5 promoter in TM4 cells. Finally, gene expression of Rhox5 and of another AR-dependent gene, Spinlw1, was reduced in ScAtrxKO testes. These data suggest that ATRX can directly enhance the expression of androgen-dependent genes through physical interaction with AR. Recruitment of ATRX by DNA sequence-specific transcription factors could be a general mechanism by which ATRX achieves tissue-specific transcriptional regulation which could explain the highly specific clinical features of ATR-X syndrome when ATRX is mutated.


Asunto(s)
Proliferación Celular , ADN Helicasas/genética , Proteínas Nucleares/genética , Receptores Androgénicos/metabolismo , Células de Sertoli/metabolismo , Animales , Apoptosis , Ciclo Celular , Células Cultivadas , Ensamble y Desensamble de Cromatina , ADN Helicasas/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunoprecipitación , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Testículo/anomalías , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Nuclear Ligada al Cromosoma X , Talasemia alfa/genética
17.
Biol Reprod ; 89(2): 34, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23843232

RESUMEN

MicroRNAs are important regulators of developmental gene expression, but their contribution to fetal gonad development is not well understood. We have identified the evolutionarily conserved gonadal microRNAs miR-202-5p and miR-202-3p as having a potential role in regulating mouse embryonic gonad differentiation. These microRNAs are expressed in a sexually dimorphic pattern as the primordial XY gonad differentiates into a testis, with strong expression in Sertoli cells. In vivo, ectopic expression of pri-miR-202 in XX gonads did not result in molecular changes to the ovarian determination pathway. Expression of the primary transcript of miR-202-5p/3p remained low in XY gonads in a conditional Sox9-null mouse model, suggesting that pri-miR-202 transcription is downstream of SOX9, a transcription factor that is both necessary and sufficient for male sex determination. We identified the pri-miR-202 promoter that is sufficient to drive expression in XY but not XX fetal gonads ex vivo. Mutation of SOX9 and SF1 binding sites reduced ex vivo transactivation of the pri-miR-202 promoter, demonstrating that pri-miR-202 may be a direct transcriptional target of SOX9/SF1 during testis differentiation. Our findings indicate that expression of the conserved gonad microRNA, miR-202-5p/3p, is downstream of the testis-determining factor SOX9, suggesting an early role in testis development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , Organogénesis/genética , Factor de Transcripción SOX9/metabolismo , Testículo/embriología , Animales , Diferenciación Celular/genética , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Regiones Promotoras Genéticas , Factor de Transcripción SOX9/genética , Células de Sertoli/citología , Células de Sertoli/metabolismo , Diferenciación Sexual/genética , Testículo/citología , Testículo/metabolismo , Transcripción Genética
18.
Chromosome Res ; 20(1): 191-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22215485

RESUMEN

In most mammals, the Y chromosomal Sry gene initiates testis formation within the bipotential gonad, resulting in male development. SRY is a transcription factor and together with SF1 it directly up-regulates the expression of the pivotal sex-determining gene Sox9 via a 1.3-kb cis-regulatory element (TESCO) which contains an evolutionarily conserved region (ECR) of 180 bp. Remarkably, several rodent species appear to determine sex in the absence of Sry and a Y chromosome, including the mole voles Ellobius lutescens and Ellobius tancrei, whereas Ellobius fuscocapillus of the same genus retained Sry. The sex-determining mechanisms in the Sry-negative species remain elusive. We have cloned and sequenced 1.1 kb of E. lutescens TESCO which shares 75% sequence identity with mouse TESCO indicating that testicular Sox9 expression in E. lutescens might still be regulated via TESCO. We have also cloned and sequenced the ECRs of E. tancrei and E. fuscocapillus. While the three Ellobius ECRs are highly similar (94-97% sequence identity), they all display a 14-bp deletion (Δ14) removing a highly conserved SOX/TCF site. Introducing Δ14 into mouse TESCO increased both basal activity and SF1-mediated activation of TESCO in HEK293T cells. We propose a model whereby Δ14 may have triggered up-regulation of Sox9 in XX gonads leading to destabilization of the XY/XX sex-determining mechanism in Ellobius. E. lutescens/E. tancrei and E. fuscocapillus could have independently stabilized their sex determination mechanisms by Sry-independent and Sry-dependent approaches, respectively.


Asunto(s)
Arvicolinae/genética , Regulación de la Expresión Génica , Factor de Transcripción SOX9/metabolismo , Procesos de Determinación del Sexo , Cromosoma Y/metabolismo , Animales , Arvicolinae/metabolismo , Arvicolinae/fisiología , Secuencia de Bases , Sitios de Unión , Clonación Molecular , Secuencia Conservada , Evolución Molecular , Femenino , Variación Genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Factor de Transcripción SOX9/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Eliminación de Secuencia , Testículo/citología , Testículo/metabolismo , Testículo/fisiología , Cromosoma Y/genética
19.
Chromosome Res ; 20(1): 127-38, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22215486

RESUMEN

The basal lineage of monotremes features an extraordinarily complex sex chromosome system which has provided novel insights into the evolution of mammalian sex chromosomes. Recently, sequence information from autosomes, X chromosomes, and XY-shared pseudoautosomal regions has become available. However, no gene has so far been described on any of the Y chromosome-specific regions. We analyzed sequences derived from Y-specific BAC clones to identify genes with potentially male-specific function. Here, we report the identification and characterization of the mediator complex protein gametologs on platypus Y5 (Crspy). We also identified the X-chromosomal copy which unexpectedly maps to X1 (Crspx). Sequence comparison shows extensive divergence between the X and Y copy, but we found no significant positive selection on either gametolog. Expression analysis shows widespread expression of Crspx. Crspy is expressed exclusively in males with particularly strong expression in testis and kidney. Reporter gene assays to investigate whether Crspx/y can act on the recently discovered mouse Sox9 testis-specific enhancer element did reveal a modest effect together with mouse Sox9 + Sf1, but showed overall no significant upregulation of the reporter gene. This is the first report of a differentiated functional male-specific gene on platypus Y chromosomes, providing new insights into sex chromosome evolution and a candidate gene for male-specific function in monotremes.


Asunto(s)
Complejo Mediador/genética , Ornitorrinco/genética , Testículo/fisiología , Cromosoma X/genética , Cromosoma Y/genética , Animales , Cromosomas Artificiales Bacterianos/genética , Cromosomas de los Mamíferos/genética , Evolución Molecular , Regulación de la Expresión Génica , Genes Reporteros , Genes sry , Células HEK293 , Humanos , Riñón/citología , Riñón/metabolismo , Masculino , Ratones , Filogenia , Mapeo Físico de Cromosoma , Factor de Transcripción SOX9/genética , Procesos de Determinación del Sexo , Testículo/citología , Transfección
20.
Nat Rev Urol ; 20(7): 434-451, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37020056

RESUMEN

Sex development relies on the sex-specific action of gene networks to differentiate the bipotential gonads of the growing fetus into testis or ovaries, followed by the differentiation of internal and external genitalia depending on the presence or absence of hormones. Differences in sex development (DSD) arise from congenital alterations during any of these processes, and are classified depending on sex chromosomal constitution as sex chromosome DSD, 46,XY DSD or 46,XX DSD. Understanding the genetics and embryology of typical and atypical sex development is essential for diagnosing, treating and managing DSD. Advances have been made in understanding the genetic causes of DSD over the past 10 years, especially for 46,XY DSD. Additional information is required to better understand ovarian and female development and to identify further genetic causes of 46,XX DSD, besides congenital adrenal hyperplasia. Ongoing research is focused on the discovery of further genes related to typical and atypical sex development and, therefore, on improving diagnosis of DSD.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX , Trastorno del Desarrollo Sexual 46,XY , Trastornos del Desarrollo Sexual , Masculino , Humanos , Femenino , Trastornos del Desarrollo Sexual/diagnóstico , Trastornos del Desarrollo Sexual/genética , Testículo , Desarrollo Sexual , Trastorno del Desarrollo Sexual 46,XY/complicaciones , Trastorno del Desarrollo Sexual 46,XY/genética , Trastorno del Desarrollo Sexual 46,XY/terapia , Trastornos del Desarrollo Sexual 46, XX/complicaciones , Trastornos del Desarrollo Sexual 46, XX/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA