Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
3.
Nat Commun ; 14(1): 3403, 2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-37296101

RESUMO

Squamous cell carcinoma antigen recognized by T cells 3 (SART3) is an RNA-binding protein with numerous biological functions including recycling small nuclear RNAs to the spliceosome. Here, we identify recessive variants in SART3 in nine individuals presenting with intellectual disability, global developmental delay and a subset of brain anomalies, together with gonadal dysgenesis in 46,XY individuals. Knockdown of the Drosophila orthologue of SART3 reveals a conserved role in testicular and neuronal development. Human induced pluripotent stem cells carrying patient variants in SART3 show disruption to multiple signalling pathways, upregulation of spliceosome components and demonstrate aberrant gonadal and neuronal differentiation in vitro. Collectively, these findings suggest that bi-allelic SART3 variants underlie a spliceosomopathy which we tentatively propose be termed INDYGON syndrome (Intellectual disability, Neurodevelopmental defects and Developmental delay with 46,XY GONadal dysgenesis). Our findings will enable additional diagnoses and improved outcomes for individuals born with this condition.


Assuntos
Disgenesia Gonadal , Células-Tronco Pluripotentes Induzidas , Deficiência Intelectual , Masculino , Humanos , Testículo/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Antígenos de Neoplasias
4.
Endocrinology ; 164(5)2023 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-36786658

RESUMO

During sex determination in the mouse, fibroblast growth factor 9 signals through the fibroblast growth factor receptor 2c isoform (FGFR2c) to trigger Sertoli cell and testis development from 11.5 days post coitum (dpc). In the XX gonad, the FOXL2 and WNT4/RSPO1 pathways drive granulosa cell and ovarian development. The function of FGFR2 in the developing ovary, and whether FGFR2 is required in the testis after sex determination, is not clear. In fetal mouse gonads from 12.5 dpc, FGFR2 shows sexually dimorphic expression. In XX gonads, FGFR2c is coexpressed with FOXL2 in pregranulosa cells, whereas XY gonads show FGFR2b expression in germ cells. Deletion of Fgfr2c in XX mice led to a marked decrease/absence of germ cells by 13.5 dpc in the ovary. This indicates that FGFR2c in the somatic pregranulosa cells is required for the maintenance of germ cells. Surprisingly, on the Fgfr2c-/- background, the germ cell phenotype could be rescued by ablation of Foxl2, suggesting a novel mechanism whereby FGFR2 and FOXL2 act antagonistically during germ cell development. Consistent with low/absent FGFR2 expression in the Sertoli cells of 12.5 and 13.5 dpc XY gonads, XY AMH:Cre; Fgfr2flox/flox mice showed normal testis morphology and structures during fetal development and in adulthood. Thus, FGFR2 is not essential for maintaining Sertoli cell fate after sex determination. Combined, these data show that FGFR2 is not necessary for Sertoli cell function after sex determination but does play an important role in the ovary.


Assuntos
Ovário , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Masculino , Feminino , Camundongos , Animais , Ovário/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Gônadas/metabolismo , Testículo/metabolismo , Células Germinativas/metabolismo , Processos de Determinação Sexual
5.
Hum Mol Genet ; 32(6): 1032-1047, 2023 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-36282544

RESUMO

Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is a congenital condition characterized by aplasia or hypoplasia of the uterus and vagina in women with a 46,XX karyotype. This condition can occur as type I when isolated or as type II when associated with extragenital anomalies including kidney and skeletal abnormalities. The genetic basis of MRKH syndrome remains unexplained and several candidate genes have been proposed to play a role in its etiology, including HNF1B, LHX1 and WNT4. Here, we conducted a microarray analysis of 13 women affected by MRKH syndrome, resulting in the identification of chromosomal changes, including the deletion at 17q12, which contains both HNF1B and LHX1. We focused on HNF1B for further investigation due to its known association with, but unknown etiological role in, MRKH syndrome. We ablated Hnf1b specifically in the epithelium of the Müllerian ducts in mice and found that this caused hypoplastic development of the uterus, as well as kidney anomalies, closely mirroring the MRKH type II phenotype. Using single-cell RNA sequencing of uterine tissue in the Hnf1b-ablated embryos, we analyzed the molecules and pathways downstream of Hnf1b, revealing a dysregulation of processes associated with cell proliferation, migration and differentiation. Thus, we establish that loss of Hnf1b function leads to an MRKH phenotype and generate the first mouse model of MRKH syndrome type II. Our results support the investigation of HNF1B in clinical genetic settings of MRKH syndrome and shed new light on the molecular mechanisms underlying this poorly understood condition in women's reproductive health.


Assuntos
Transtornos 46, XX do Desenvolvimento Sexual , Ductos Paramesonéfricos , Animais , Feminino , Camundongos , Transtornos 46, XX do Desenvolvimento Sexual/genética , Diferenciação Celular , Genômica , Fator 1-beta Nuclear de Hepatócito/genética , Humanos
6.
Sex Dev ; 16(4): 270-282, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35306493

RESUMO

INTRODUCTION: Sex determination in eutherian mammals is controlled by the Y-linked gene Sry, which drives the formation of testes in male embryos. Despite extensive study, the genetic steps linking Sry action and male sex determination remain largely unknown. Here, we focused on Mmd2, a gene that encodes a member of the progestin and adipoQ receptor (PAQR) family. Mmd2 is expressed during the sex-determining period in XY but not XX gonads, suggesting a specific role in testis development. METHODS: We used CRISPR to generate mouse strains deficient in Mmd2 and its 2 closely related PAQR family members, Mmd and Paqr8, which are also expressed during testis development. Following characterization of Mmd2 expression in the developing testis, we studied sex determination in embryos from single knockout as well as Mmd2;Mmd and Mmd2;Paqr8 double knockout lines using quantitative RT-PCR and immunofluorescence. RESULTS: Analysis of knockout mice deficient in Sox9 and Nr5a1 revealed that Mmd2 operates downstream of these known sex-determining genes. However, fetal testis development progressed normally in Mmd2-null embryos. To determine if other genes might have compensated for the loss of Mmd2, we analyzed Paqr8 and Mmd-null embryos and confirmed that in both knockout lines, sex determination occurred normally. Finally, we generated Mmd2;Mmd and Mmd2;Paqr8 double-null embryos and again observed normal testis development. DISCUSSION: These results may reflect functional redundancy among PAQR factors, or their dispensability in gonadal development. Our findings highlight the difficulties involved in identifying genes with a functional role in sex determination and gonadal development through expression screening and loss-of-function analyses of individual candidate genes and may help to explain the paucity of genes in which variations have been found to cause human disorders/differences of sex development.


Assuntos
Gônadas , Processos de Determinação Sexual , Humanos , Camundongos , Masculino , Animais , Processos de Determinação Sexual/genética , Proteína da Região Y Determinante do Sexo/genética , Proteína da Região Y Determinante do Sexo/metabolismo , Gônadas/metabolismo , Testículo/metabolismo , Diferenciação Sexual/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mamíferos/genética , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
7.
Hum Mol Genet ; 31(13): 2223-2235, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35134173

RESUMO

The genetic regulation of ovarian development remains largely unclear. Indeed, in most cases of impaired ovarian development-such as 46,XX disorders of sex development (DSD) without SRY, and premature ovarian insufficiency (POI)-the genetic causes have not been identified, and the vast majority of disease-associated sequence variants could lie within non-coding regulatory sequences. In this study, we aimed to identify enhancers of five ovarian genes known to play key roles in early ovarian development, basing our analysis on the expression of enhancer derived transcripts (eRNAs), which are considered to characterize active enhancers. Temporal expression profile changes in mouse WT1-positive ovarian cells were obtained from cap analysis of gene expression at E13.5, E16.5 and P0. We compared the chronological expression profiles of ovarian-specific eRNA with expression profiles for each of the ovarian-specific genes, yielding two candidate sequences for enhancers of Wnt4 and Rspo1. Both sequences are conserved between mouse and human, and we confirmed their enhancer activities using transient expression assays in murine granulosa cells. Furthermore, by sequencing the region in patients with impaired ovarian development in 24 patients, such as POI, gonadal dysgenesis and 46,XX DSD, we identified rare single nucleotide variants in both sequences. Our results demonstrate that combined analysis of the temporal expression profiles of eRNA and mRNA of target genes presents a powerful tool for locating cis-element enhancers, and a means of identifying disease-associated sequence variants that lie within non-coding regulatory sequences, thus advancing an important unmet need in forward human genetics.


Assuntos
Menopausa Precoce , Insuficiência Ovariana Primária , Animais , Elementos Facilitadores Genéticos/genética , Feminino , Variação Genética , Humanos , Menopausa Precoce/genética , Camundongos , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/metabolismo , RNA/genética , Fatores de Tempo
8.
Hum Mutat ; 43(3): 362-379, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34918413

RESUMO

SRY is the Y-chromosomal gene that determines male sex development in humans and most other mammals. After three decades of study, we still lack a detailed understanding of which domains of the SRY protein are required to engage the pathway of gene activity leading to testis development. Some insight has been gained from the study of genetic variations underlying differences/disorders of sex determination (DSD), but the lack of a system of experimentally generating SRY mutations and studying their consequences in vivo has limited progress in the field. To address this issue, we generated a mouse model carrying a human SRY transgene able to drive testis determination in XX mice. Using CRISPR-Cas9 gene editing, we generated novel genetic modifications in each of SRY's three domains (N-terminal, HMG box, and C-terminal) and performed a detailed analysis of their molecular and cellular effects on embryonic testis development. Our results provide new functional insights unique to human SRY and present a versatile and powerful system in which to functionally analyze variations of SRY including known and novel pathogenic variants found in DSD.


Assuntos
Edição de Genes , Camundongos Transgênicos , Proteína da Região Y Determinante do Sexo , Testículo , Animais , Humanos , Masculino , Camundongos , Domínios Proteicos , Proteína da Região Y Determinante do Sexo/genética , Testículo/metabolismo
9.
Dev Dyn ; 251(2): 336-349, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34174014

RESUMO

BACKGROUND: Lymphatic vascular development is regulated by well-characterized signaling and transcriptional pathways. These pathways regulate lymphatic endothelial cell (LEC) migration, motility, polarity, and morphogenesis. Canonical and non-canonical WNT signaling pathways are known to control LEC polarity and development of lymphatic vessels and valves. PKD1, encoding Polycystin-1, is the most commonly mutated gene in polycystic kidney disease but has also been shown to be essential in lymphatic vascular morphogenesis. The mechanism by which Pkd1 acts during lymphangiogenesis remains unclear. RESULTS: Here we find that loss of non-canonical WNT signaling components Wnt5a and Ryk phenocopy lymphatic defects seen in Pkd1 knockout mice. To investigate genetic interaction, we generated Pkd1;Wnt5a double knockout mice. Loss of Wnt5a suppressed phenotypes seen in the lymphatic vasculature of Pkd1-/- mice and Pkd1 deletion suppressed phenotypes observed in Wnt5a-/- mice. Thus, we report mutually suppressive roles for Pkd1 and Wnt5a, with developing lymphatic networks restored to a more wild type state in double mutant mice. This genetic interaction between Pkd1 and the non-canonical WNT signaling pathway ultimately controls LEC polarity and the morphogenesis of developing vessel networks. CONCLUSION: Our work suggests that Pkd1 acts at least in part by regulating non-canonical WNT signaling during the formation of lymphatic vascular networks.


Assuntos
Vasos Linfáticos , Doenças Renais Policísticas , Animais , Vasos Linfáticos/metabolismo , Camundongos , Camundongos Knockout , Morfogênese/genética , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Proteína Quinase C , Receptores Proteína Tirosina Quinases/metabolismo , Via de Sinalização Wnt/genética , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
10.
Nucleic Acids Res ; 49(19): 10931-10955, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34570228

RESUMO

Few genetically dominant mutations involved in human disease have been fully explained at the molecular level. In cases where the mutant gene encodes a transcription factor, the dominant-negative mode of action of the mutant protein is particularly poorly understood. Here, we studied the genome-wide mechanism underlying a dominant-negative form of the SOX18 transcription factor (SOX18RaOp) responsible for both the classical mouse mutant Ragged Opossum and the human genetic disorder Hypotrichosis-lymphedema-telangiectasia-renal defect syndrome. Combining three single-molecule imaging assays in living cells together with genomics and proteomics analysis, we found that SOX18RaOp disrupts the system through an accumulation of molecular interferences which impair several functional properties of the wild-type SOX18 protein, including its target gene selection process. The dominant-negative effect is further amplified by poisoning the interactome of its wild-type counterpart, which perturbs regulatory nodes such as SOX7 and MEF2C. Our findings explain in unprecedented detail the multi-layered process that underpins the molecular aetiology of dominant-negative transcription factor function.


Assuntos
Glomerulonefrite/genética , Hipotricose/genética , Linfedema/genética , Fatores de Transcrição SOXF/genética , Telangiectasia/genética , Transcrição Gênica , Animais , Células COS , Chlorocebus aethiops , Modelos Animais de Doenças , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Genes Reporter , Glomerulonefrite/metabolismo , Glomerulonefrite/patologia , Células HeLa , Células Endoteliais da Veia Umbilical Humana , Humanos , Hipotricose/metabolismo , Hipotricose/patologia , Luciferases/genética , Luciferases/metabolismo , Linfedema/metabolismo , Linfedema/patologia , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Camundongos , Mutação , Fatores de Transcrição SOXF/metabolismo , Imagem Individual de Molécula , Telangiectasia/metabolismo , Telangiectasia/patologia
11.
Development ; 148(5)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33574039

RESUMO

In mice, the entry of germ cells into meiosis crucially depends on the expression of stimulated by retinoic acid gene 8 (Stra8). Stra8 is expressed specifically in pre-meiotic germ cells of females and males, at fetal and postnatal stages, respectively, but the mechanistic details of its spatiotemporal regulation are yet to be defined. In particular, there has been considerable debate regarding whether retinoic acid is required, in vivo, to initiate Stra8 expression in the mouse fetal ovary. We show that the distinctive anterior-to-posterior pattern of Stra8 initiation, characteristic of germ cells in the fetal ovary, is faithfully recapitulated when 2.9 kb of the Stra8 promoter is used to drive eGFP expression. Using in vitro transfection assays of cutdown and mutant constructs, we identified two functional retinoic acid responsive elements (RAREs) within this 2.9 kb regulatory element. We also show that the transcription factor DMRT1 enhances Stra8 expression, but only in the presence of RA and the most proximal RARE. Finally, we used CRISPR/Cas9-mediated targeted mutation studies to demonstrate that both RAREs are required for optimal Stra8 expression levels in vivo.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Germinativas/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sítios de Ligação , Sistemas CRISPR-Cas/genética , Feminino , Desenvolvimento Fetal/genética , Feto/citologia , Feto/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células Germinativas/citologia , Meiose , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutagênese , Ovário/citologia , Ovário/metabolismo , Regiões Promotoras Genéticas , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/farmacologia , Tretinoína/farmacologia
12.
Science ; 370(6512): 121-124, 2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-33004521

RESUMO

The mammalian sex-determining gene Sry induces male development. Since its discovery 30 years ago, Sry has been believed to be a single-exon gene. Here, we identified a cryptic second exon of mouse Sry and a corresponding two-exon type Sry (Sry-T) transcript. XY mice lacking Sry-T were sex-reversed, and ectopic expression of Sry-T in XX mice induced male development. Sry-T messenger RNA is expressed similarly to that of canonical single-exon type Sry (Sry-S), but SRY-T protein is expressed predominantly because of the absence of a degron in the C terminus of SRY-S. Sry exon2 appears to have evolved recently in mice through acquisition of a retrotransposon-derived coding sequence to replace the degron. Our findings suggest that in nature, SRY-T, not SRY-S, is the bona fide testis-determining factor.


Assuntos
Éxons , Genes Essenciais , Processos de Determinação Sexual , Proteína da Região Y Determinante do Sexo/genética , Animais , Sistemas CRISPR-Cas , Edição de Genes , Loci Gênicos , Masculino , Camundongos , Camundongos Transgênicos , RNA não Traduzido/genética , Deleção de Sequência , Transcrição Gênica
13.
Hum Mol Genet ; 29(13): 2148-2161, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32452519

RESUMO

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.


Assuntos
Fator 9 de Crescimento de Fibroblastos/genética , Transtornos Ovotesticulares do Desenvolvimento Sexual/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Sinostose/genética , Animais , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Gônadas/crescimento & desenvolvimento , Gônadas/patologia , Humanos , Masculino , Camundongos , Mutação de Sentido Incorreto/genética , Transtornos Ovotesticulares do Desenvolvimento Sexual/patologia , Fatores de Transcrição SOX9/genética , Processos de Determinação Sexual/genética , Desenvolvimento Sexual/genética
14.
G3 (Bethesda) ; 9(11): 3645-3651, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31492696

RESUMO

The emergence of the CRISPR-Cas9 system has triggered a technical revolution in mammalian genome editing. Compared to traditional gene-targeting strategies, CRISPR-Cas9 technology offers a more efficient and cost-effective approach for generating genetically modified animal models. However, off-target cleavage in CRISPR-mediated genome editing is a major concern in the analysis of phenotypes as well as the selection of therapeutic targets. Here, we analyzed whole-genome sequencing (WGS) data from two knock-out (KO) mouse strains generated by using the CRISPR-Cas9 system targeting the Mmd and Paqr8 loci. A total of nine individuals were sequenced including two parents, four F1 offspring and three uninjected control mice. Using GATK and bcftools software, we identified two off-target events in the founder mice. The two CRISPR-Cas9-induced off-target events were predictable using Cas-OFFinder and were not passed on to the offspring that we investigated. In addition, our results indicated that the number of CRISPR-Cas9-induced mutations was not statistically distinguishable from the background de novo mutations (DNMs).


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Animais , Feminino , Marcação de Genes , Genoma , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Receptores de Progesterona/genética , Sequenciamento Completo do Genoma
15.
Sci Rep ; 9(1): 11953, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31420575

RESUMO

The endocardium is the endothelial component of the vertebrate heart and plays a key role in heart development. Where, when, and how the endocardium segregates during embryogenesis have remained largely unknown, however. We now show that Nkx2-5+ cardiac progenitor cells (CPCs) that express the Sry-type HMG box gene Sox17 from embryonic day (E) 7.5 to E8.5 specifically differentiate into the endocardium in mouse embryos. Although Sox17 is not essential or sufficient for endocardium fate, it can bias the fate of CPCs toward the endocardium. On the other hand, Sox17 expression in the endocardium is required for heart development. Deletion of Sox17 specifically in the mesoderm markedly impaired endocardium development with regard to cell proliferation and behavior. The proliferation of cardiomyocytes, ventricular trabeculation, and myocardium thickening were also impaired in a non-cell-autonomous manner in the Sox17 mutant, likely as a consequence of down-regulation of NOTCH signaling. An unknown signal, regulated by Sox17 and required for nurturing of the myocardium, is responsible for the reduction in NOTCH-related genes in the mutant embryos. Our results thus provide insight into differentiation of the endocardium and its role in heart development.


Assuntos
Diferenciação Celular , Embrião de Mamíferos/embriologia , Endocárdio/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas HMGB/biossíntese , Fatores de Transcrição SOXF/biossíntese , Transdução de Sinais , Células-Tronco/metabolismo , Animais , Embrião de Mamíferos/citologia , Endocárdio/citologia , Proteínas HMGB/genética , Mesoderma/citologia , Mesoderma/embriologia , Camundongos , Camundongos Transgênicos , Receptores Notch/genética , Receptores Notch/metabolismo , Fatores de Transcrição SOXF/genética , Células-Tronco/citologia
17.
J Cell Sci ; 132(8)2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30877223

RESUMO

The nuclear receptor NR5A1 is equally expressed and required for development of the gonadal primordia of both sexes, but, after sex determination, it is upregulated in XY testes and downregulated in XX ovaries. We have recently demonstrated, in mice, that this downregulation is mediated by forkhead box L2 (FOXL2) and hypothesized that adequate suppression of Nr5a1 is essential for normal ovarian development. Further, analysis of human patients with disorders/differences of sex development suggests that overexpression of NR5A1 can result in XX (ovo)testicular development. Here, we tested the role of Nr5a1 by overexpression in fetal gonads using a Wt1-BAC (bacterial artificial chromosome) transgene system. Enforced Nr5a1 expression compromised ovarian development in 46,XX mice, resulting in late-onset infertility, but did not induce (ovo)testis differentiation. The phenotype was similar to that of XX mice lacking Notch signaling. The expression level of Notch2 was significantly reduced in Nr5a1 transgenic mice, and the ovarian phenotype was almost completely rescued by in utero treatment with a NOTCH2 agonist. We conclude that suppression of Nr5a1 during the fetal period optimizes ovarian development by fine-tuning Notch signaling.


Assuntos
Ovário/fisiologia , Receptor Notch2/fisiologia , Desenvolvimento Sexual , Fator Esteroidogênico 1/fisiologia , Testículo/fisiologia , Animais , Diferenciação Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , Receptor Notch2/genética , Transdução de Sinais , Fator Esteroidogênico 1/genética
18.
Genome Biol ; 20(1): 23, 2019 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-30704500

RESUMO

BACKGROUND: Altering the biochemical makeup of chromatin by the incorporation of histone variants during development represents a key mechanism in regulating gene expression. The histone variant H2A.B, H2A.B.3 in mice, appeared late in evolution and is most highly expressed in the testis. In the mouse, it is encoded by three different genes. H2A.B expression is spatially and temporally regulated during spermatogenesis being most highly expressed in the haploid round spermatid stage. Active genes gain H2A.B where it directly interacts with polymerase II and RNA processing factors within splicing speckles. However, the importance of H2A.B for gene expression and fertility are unknown. RESULTS: Here, we report the first mouse knockout of this histone variant and its effects on fertility, nuclear organization, and gene expression. In view of the controversy related to the generation of off-target mutations by gene editing approaches, we test the specificity of TALENs by disrupting the H2A.B multi-copy gene family using only one pair of TALENs. We show that TALENs do display a high level of specificity since no off-target mutations are detected by bioinformatics analyses of exome sequences obtained from three consecutive generations of knockout mice and by Sanger DNA sequencing. Male H2A.B.3 knockout mice are subfertile and display an increase in the proportion of abnormal sperm and clogged seminiferous tubules. Significantly, a loss of proper RNA Pol II targeting to distinct transcription-splicing territories and changes to pre-mRNA splicing are observed. CONCLUSION: We have produced the first H2A.B knockout mouse using the TALEN approach.


Assuntos
Fertilidade/genética , Edição de Genes/métodos , Histonas/genética , Infertilidade Masculina/etiologia , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição , Animais , Sequência de Bases , Proteínas Cromossômicas não Histona/metabolismo , Feminino , Expressão Gênica , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Masculino , Camundongos Knockout , Mutação , RNA Polimerase II/metabolismo , Espermatozoides/metabolismo , Espermatozoides/patologia
19.
Sex Dev ; 13(1): 26-34, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30504698

RESUMO

Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is a disorder of sex development which affects 1 in 4,500 females and is characterized by agenesis of müllerian structures, including the uterus, cervix, and upper vagina. It can occur in isolation (type 1) or in conjunction with various anomalies (type 2), with a subset of these comprising müllerian, renal, and cervicothoracic abnormalities (MURCS) association. The genetic causes of MRKH have been investigated previously yielding limited results, with massive parallel sequencing becoming increasingly utilized. We sought to identify genetic contributions to MRKH using a combination of microarray and whole exome sequencing (WES) on a cohort of 8 unrelated women with MRKH and MURCS. WES data were analysed using a candidate gene approach to identify potential contributing variants. Microarray analysis identified a 0.6-Mb deletion in the previously implicated 16p11.2 region in a patient with MRKH type 2. WES revealed 16 rare nonsynonymous variants in MRKH candidate genes across the cohort. These included variants in several genes, such as LRP10 and DOCK4, associated with disorders with müllerian anomalies. Further functional studies of these variants will help to delineate their biological significance and expand the genotypic spectrum of MRKH.


Assuntos
Transtornos 46, XX do Desenvolvimento Sexual/genética , Anormalidades Congênitas/genética , Estudos de Associação Genética , Genômica/métodos , Ductos Paramesonéfricos/anormalidades , Adulto , Variações do Número de Cópias de DNA/genética , Feminino , Humanos , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Sequenciamento do Exoma , Adulto Jovem
20.
Nat Commun ; 9(1): 5319, 2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30552336

RESUMO

Disorders of sex development (DSDs) are conditions affecting development of the gonads or genitalia. Variants in two key genes, SRY and its target SOX9, are an established cause of 46,XY DSD, but the genetic basis of many DSDs remains unknown. SRY-mediated SOX9 upregulation in the early gonad is crucial for testis development, yet the regulatory elements underlying this have not been identified in humans. Here, we identified four DSD patients with overlapping duplications or deletions upstream of SOX9. Bioinformatic analysis identified three putative enhancers for SOX9 that responded to different combinations of testis-specific regulators. All three enhancers showed synergistic activity and together drive SOX9 in the testis. This is the first study to identify SOX9 enhancers that, when duplicated or deleted, result in 46,XX or 46,XY sex reversal, respectively. These enhancers provide a hitherto missing link by which SRY activates SOX9 in humans, and establish SOX9 enhancer mutations as a significant cause of DSD.


Assuntos
Transtornos do Desenvolvimento Sexual/genética , Duplicação Gênica/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Deleção de Sequência/genética , Proteína da Região Y Determinante do Sexo/genética , Transtornos 46, XX do Desenvolvimento Sexual/genética , Animais , Linhagem Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Transtorno 46,XY do Desenvolvimento Sexual/genética , Feminino , Técnicas de Inativação de Genes , Genitália/metabolismo , Gônadas/metabolismo , Humanos , Masculino , Camundongos , Mutação , Fatores de Processamento de RNA/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Fatores de Transcrição SOX/genética , Diferenciação Sexual , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...