Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Development ; 147(6)2020 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-32108023

RESUMO

Members of the Iroquois B (IrxB) homeodomain cluster genes, specifically Irx3 and Irx5, are crucial for heart, limb and bone development. Recently, we reported their importance for oocyte and follicle survival within the developing ovary. Irx3 and Irx5 expression begins after sex determination in the ovary but remains absent in the fetal testis. Mutually antagonistic molecular signals ensure ovary versus testis differentiation with canonical Wnt/ß-catenin signals paramount for promoting the ovary pathway. Notably, few direct downstream targets have been identified. We report that Wnt/ß-catenin signaling directly stimulates Irx3 and Irx5 transcription in the developing ovary. Using in silico analysis of ATAC- and ChIP-Seq databases in conjunction with mouse gonad explant transfection assays, we identified TCF/LEF-binding sequences within two distal enhancers of the IrxB locus that promote ß-catenin-responsive ovary expression. Meanwhile, Irx3 and Irx5 transcription is suppressed within the developing testis by the presence of H3K27me3 on these same sites. Thus, we resolved sexually dimorphic regulation of Irx3 and Irx5 via epigenetic and ß-catenin transcriptional control where their ovarian presence promotes oocyte and follicle survival vital for future ovarian health.


Assuntos
Epigênese Genética/fisiologia , Gônadas/embriologia , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Diferenciação Celular/genética , Células Cultivadas , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/metabolismo , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Ovário/embriologia , Ovário/metabolismo , Caracteres Sexuais , Diferenciação Sexual/genética , Testículo/embriologia , Testículo/metabolismo , Fatores de Transcrição/metabolismo
2.
PLoS Genet ; 15(5): e1007895, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31116734

RESUMO

XX and XY fetal gonads are initially bipotential, poised between the ovary and testis fate. Multiple lines of evidence suggest that commitment to testis fate requires the repression of genes associated with ovary fate. It was previously shown that loss of CBX2, the subunit of the Polycomb Repressive Complex 1 (PRC1) that binds H3K27me3 and mediates silencing, leads to ovary development in XY mice and humans. While it had been proposed that CBX2 is an activator of the testis-determining gene Sry, we investigated the alternative possibility that CBX2 has a direct role as a repressor of the antagonistic ovary-promoting pathway. To investigate this possibility, we developed a quantitative genome-wide profile of the repressive histone mark H3K27me3 and its active counterpart H3K4me3 in isolated XY and XX gonadal supporting cells before and after sex determination. We show that testis and ovary sex-determining (SD) genes are bivalent before sex determination, providing insight into how the bipotential state of the gonad is established at the epigenetic level. After sex determination, many SD genes of the alternate pathway remain bivalent, possibly contributing to the ability of these cells to transdifferentiate even in adults. The finding that many genes in the Wnt signaling pathway were targeted for H3K27me3-mediated repression in Sertoli cells led us to test whether deletion of Wnt4 could rescue testis development in Cbx2 mutants. We show that Sry expression and testis development were rescued in XY Cbx2-/-;Wnt4-/- mice. Furthermore, we show that CBX2 directly binds the downstream Wnt signaler Lef1, an ovary-promoting gene that remains bivalent in Sertoli cells. Our results suggest that stabilization of the testis fate requires CBX2-mediated repression of bivalent ovary-determining genes, which would otherwise block testis development.


Assuntos
Epigênese Genética , Ovário/metabolismo , Complexo Repressor Polycomb 1/genética , Processos de Determinação Sexual , Testículo/metabolismo , Via de Sinalização Wnt/genética , Animais , Embrião de Mamíferos , Feminino , Fator 9 de Crescimento de Fibroblastos/genética , Fator 9 de Crescimento de Fibroblastos/metabolismo , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Histonas/genética , Histonas/metabolismo , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Masculino , Camundongos , Ovário/citologia , Ovário/crescimento & desenvolvimento , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Complexo Repressor Polycomb 1/deficiência , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Diferenciação Sexual , Testículo/citologia , Testículo/crescimento & desenvolvimento , Proteína Wnt4/genética , Proteína Wnt4/metabolismo
3.
Dev Biol ; 446(2): 168-179, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30594505

RESUMO

Cis-regulatory elements are critical for the precise spatiotemporal regulation of genes during development. However, identifying functional regulatory sites that drive cell differentiation in vivo has been complicated by the high numbers of cells required for whole-genome epigenetic assays. Here, we identified putative regulatory elements during sex determination by performing ATAC-seq and ChIP-seq for H3K27ac in purified XX and XY gonadal supporting cells before and after sex determination in mice. We show that XX and XY supporting cells initiate sex determination with similar chromatin landscapes and acquire sex-specific regulatory elements as they commit to the male or female fate. To validate our approach, we identified a functional gonad-specific enhancer downstream of Bmp2, an ovary-promoting gene. This work increases our understanding of the complex regulatory network underlying mammalian sex determination and provides a powerful resource for identifying non-coding regulatory elements that could harbor mutations that lead to Disorders of Sexual Development.


Assuntos
Cromatina/genética , Gônadas/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Processos de Determinação Sexual/genética , Acetilação , Animais , Cromatina/metabolismo , Feminino , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/citologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Histonas/genética , Histonas/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos
4.
Development ; 144(4): 720-730, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28087634

RESUMO

A current goal of molecular biology is to identify transcriptional networks that regulate cell differentiation. However, identifying functional gene regulatory elements has been challenging in the context of developing tissues where material is limited and cell types are mixed. To identify regulatory sites during sex determination, we subjected Sertoli cells from mouse fetal testes to DNaseI-seq and ChIP-seq for H3K27ac. DNaseI-seq identified putative regulatory sites around genes enriched in Sertoli and pregranulosa cells; however, active enhancers marked by H3K27ac were enriched proximal to only Sertoli-enriched genes. Sequence analysis identified putative binding sites of known and novel transcription factors likely controlling Sertoli cell differentiation. As a validation of this approach, we identified a novel Sertoli cell enhancer upstream of Wt1, and used it to drive expression of a transgenic reporter in Sertoli cells. This work furthers our understanding of the complex genetic network that underlies sex determination and identifies regions that potentially harbor non-coding mutations underlying disorders of sexual development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Elementos Reguladores de Transcrição , Células de Sertoli/metabolismo , Animais , Sítios de Ligação , Diferenciação Celular , Desoxirribonuclease I/metabolismo , Elementos Facilitadores Genéticos , Genes Reporter , Genoma , Histonas/metabolismo , Homozigoto , Masculino , Camundongos , Mutação , Regiões Promotoras Genéticas , Processos de Determinação Sexual , Testículo/embriologia , Transgenes
5.
PLoS Genet ; 8(3): e1002575, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22438826

RESUMO

The divergence of distinct cell populations from multipotent progenitors is poorly understood, particularly in vivo. The gonad is an ideal place to study this process, because it originates as a bipotential primordium where multiple distinct lineages acquire sex-specific fates as the organ differentiates as a testis or an ovary. To gain a more detailed understanding of the process of gonadal differentiation at the level of the individual cell populations, we conducted microarrays on sorted cells from XX and XY mouse gonads at three time points spanning the period when the gonadal cells transition from sexually undifferentiated progenitors to their respective sex-specific fates. We analyzed supporting cells, interstitial/stromal cells, germ cells, and endothelial cells. This work identified genes specifically depleted and enriched in each lineage as it underwent sex-specific differentiation. We determined that the sexually undifferentiated germ cell and supporting cell progenitors showed lineage priming. We found that germ cell progenitors were primed with a bias toward the male fate. In contrast, supporting cells were primed with a female bias, indicative of the robust repression program involved in the commitment to XY supporting cell fate. This study provides a molecular explanation reconciling the female default and balanced models of sex determination and represents a rich resource for the field. More importantly, it yields new insights into the mechanisms by which different cell types in a single organ adopt their respective fates.


Assuntos
Desenvolvimento Embrionário/genética , Células Endoteliais , Células Germinativas , Gônadas , Células Estromais , Animais , Diferenciação Celular , Linhagem da Célula , Células Endoteliais/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Gônadas/crescimento & desenvolvimento , Gônadas/metabolismo , Masculino , Camundongos , Análise em Microsséries , Processos de Determinação Sexual , Células Estromais/metabolismo
6.
Dev Biol ; 383(2): 295-306, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24036309

RESUMO

Mammalian sex determination is controlled by antagonistic pathways that are initially co-expressed in the bipotential gonad and subsequently become male- or female-specific. In XY gonads, testis development is initiated by upregulation of Sox9 by SRY in pre-Sertoli cells. Disruption of either gene leads to complete male-to-female sex reversal. Ovarian development is dependent on canonical Wnt signaling through Wnt4, Rspo1 and ß-catenin. However, only a partial female-to-male sex reversal results from disruption of these ovary-promoting genes. In Wnt4 and Rspo1 mutants, there is evidence of pregranulosa cell-to-Sertoli cell transdifferentiation near birth, following a severe decline in germ cells. It is currently unclear why primary sex reversal does not occur at the sex-determining stage, but instead occurs near birth in these mutants. Here we show that Wnt4-null and Rspo1-null pregranulosa cells transition through a differentiated granulosa cell state prior to transdifferentiating towards a Sertoli cell fate. This transition is preceded by a wave of germ cell death that is closely associated with the disruption of pregranulosa cell quiescence. Our results suggest that maintenance of mitotic arrest in pregranulosa cells may preclude upregulation of Sox9 in cases where female sex-determining genes are disrupted. This may explain the lack of complete sex reversal in such mutants at the sex-determining stage.


Assuntos
Pontos de Checagem do Ciclo Celular , Diferenciação Celular , Transdiferenciação Celular , Células da Granulosa/citologia , Mitose , Mutação/genética , Proteína Wnt4/metabolismo , Animais , Animais Recém-Nascidos , Hormônio Antimülleriano/metabolismo , Morte Celular , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Feminino , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/metabolismo , Células Germinativas/citologia , Células Germinativas/metabolismo , Células da Granulosa/metabolismo , Masculino , Meiose , Camundongos , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Trombospondinas/metabolismo
7.
Biol Reprod ; 86(2): 37, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21976597

RESUMO

The embryonic origins of ovarian granulosa cells have been a subject of debate for decades. By tamoxifen-induced lineage tracing of Foxl2-expressing cells, we show that descendants of the bipotential supporting cell precursors in the early gonad contribute granulosa cells to a specific population of follicles in the medulla of the ovary that begin to grow immediately after birth. These precursor cells arise from the proliferative ovarian surface epithelium and enter mitotic arrest prior to upregulating Foxl2. Granulosa cells that populate the cortical primordial follicles activated in adult life derive from the surface epithelium perinatally, and enter mitotic arrest at that stage. Ingression from the surface epithelium dropped to undetectable levels by Postnatal Day 7, when most surviving oocytes were individually encapsulated by granulosa cells. These findings add complexity to the standard model of sex determination in which the Sertoli and granulosa cells of the adult testis and ovary directly stem from the supporting cell precursors of the bipotential gonad.


Assuntos
Linhagem da Célula , Células da Granulosa/citologia , Folículo Ovariano/citologia , Ovário/embriologia , Animais , Diferenciação Celular , Desenvolvimento Embrionário , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/metabolismo , Células da Granulosa/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Animais , Folículo Ovariano/metabolismo , Ovário/citologia
8.
Dev Biol ; 327(2): 516-23, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19210962

RESUMO

The apical ectodermal ridge (AER) in the vertebrate limb is required for limb outgrowth and patterning. To investigate the role BMP ligands expressed in the AER play in limb development we selectively inactivated both Bmp2 and Bmp4 in this tissue. The autopods of mice lacking both of these genes contained extra digits, digit bifurcations and interdigital webbing due to a decrease in programmed cell death and an increase in cell proliferation in the underlying mesoderm. Upon removal of Bmp2 and Bmp4 in the AER, no defects in proximal-distal patterning were observed. At the molecular level, removal of Bmp2 and Bmp4 in the AER caused an increase in Fgf expression, which correlated with an increase in both the width and length of the AER. Investigation of Engrailed-1 (En1) expression in the AER of limb buds in which Bmp2 and Bmp4 had been removed indicated that En1 expression was absent from this tissue. Our data suggests that AER expression of Bmp2 and Bmp4 is required for digit and dorsal-ventral patterning but surprisingly not for limb outgrowth.


Assuntos
Padronização Corporal/fisiologia , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Morte Celular/fisiologia , Ectoderma , Extremidades , Animais , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 4/genética , Proliferação de Células , Citocinas , Ectoderma/anatomia & histologia , Ectoderma/fisiologia , Extremidades/anatomia & histologia , Extremidades/embriologia , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Deformidades Congênitas dos Membros , Masculino , Camundongos , Transdução de Sinais/fisiologia
9.
Hum Mol Genet ; 17(19): 2949-55, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18617533

RESUMO

During mammalian sex determination, expression of the Y-linked gene Sry shifts the bipotential gonad toward a testicular fate by upregulating a feed-forward loop between FGF9 and SOX9 to establish SOX9 expression in somatic cells. We previously proposed that these signals are mutually antagonistic with counteracting signals in XX gonads and that a shift in the balance of these factors leads to either male or female development. Evidence in mice and humans suggests that the male pathway is opposed by the expression of two signals, WNT4 and R-SPONDIN-1 (RSPO1), that promote the ovarian fate and block testis development. Both of these ligands can activate the canonical Wnt signaling pathway. Duplication of the distal portion of chromosome 1p, which includes both WNT4 and RSPO1, overrides the male program and causes male-to-female sex reversal in XY patients. To determine whether activation of beta-catenin is sufficient to block the testis pathway, we have ectopically expressed a stabilized form of beta-catenin in the somatic cells of XY gonads. Our results show that activation of beta-catenin in otherwise normal XY mice effectively disrupts the male program and results in male-to-female sex-reversal. The identification of beta-catenin as a key pro-ovarian and anti-testis signaling molecule will further our understanding of the mechanisms controlling sex determination and the molecular mechanisms that lead to sex-reversal.


Assuntos
Transtornos do Desenvolvimento Sexual , Regulação da Expressão Gênica no Desenvolvimento , Ovário/crescimento & desenvolvimento , Testículo/crescimento & desenvolvimento , beta Catenina/metabolismo , Animais , Feminino , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovário/metabolismo , Fenótipo , Diferenciação Sexual , Transdução de Sinais , Testículo/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt4 , beta Catenina/genética
10.
Science ; 360(6396): 1469-1473, 2018 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-29903884

RESUMO

Cell fate decisions require appropriate regulation of key genes. Sox9, a direct target of SRY, is pivotal in mammalian sex determination. In vivo high-throughput chromatin accessibility techniques, transgenic assays, and genome editing revealed several novel gonadal regulatory elements in the 2-megabase gene desert upstream of Sox9 Although others are redundant, enhancer 13 (Enh13), a 557-base pair element located 565 kilobases 5' from the transcriptional start site, is essential to initiate mouse testis development; its deletion results in XY females with Sox9 transcript levels equivalent to those in XX gonads. Our data are consistent with the time-sensitive activity of SRY and indicate a strict order of enhancer usage. Enh13 is conserved and embedded within a 32.5-kilobase region whose deletion in humans is associated with XY sex reversal, suggesting that it is also critical in humans.


Assuntos
Elementos Facilitadores Genéticos/genética , Disgenesia Gonadal 46 XY/genética , Fatores de Transcrição SOX9/genética , Processos de Determinação Sexual/genética , Proteína da Região Y Determinante do Sexo/metabolismo , Testículo/embriologia , Animais , Sequência Conservada , Feminino , Humanos , Masculino , Camundongos , Deleção de Sequência , Proteína da Região Y Determinante do Sexo/genética , Sítio de Iniciação de Transcrição
11.
PLoS One ; 7(10): e47238, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23091613

RESUMO

The fetal gonad is composed of a mixture of somatic cell lineages and germ cells. The fate of the gonad, male or female, is determined by a population of somatic cells that differentiate into Sertoli or granulosa cells and direct testis or ovary development. It is well established that germ cells are not required for the establishment or maintenance of Sertoli cells or testis cords in the male gonad. However, in the agametic ovary, follicles do not form suggesting that germ cells may influence granulosa cell development. Prior investigations of ovaries in which pre-meiotic germ cells were ablated during fetal life reported no histological changes during stages prior to birth. However, whether granulosa cells underwent normal molecular differentiation was not investigated. In cases where germ cell loss occurred secondary to other mutations, transdifferentiation of granulosa cells towards a Sertoli cell fate was observed, raising questions about whether germ cells play an active role in establishing or maintaining the fate of granulosa cells. We developed a group of molecular markers associated with ovarian development, and show here that the loss of pre-meiotic germ cells does not disrupt the somatic ovarian differentiation program during fetal life, or cause transdifferentiation as defined by expression of Sertoli markers. Since we do not find defects in the ovarian somatic program, the subsequent failure to form follicles at perinatal stages is likely attributable to the absence of germ cells rather than to defects in the somatic cells.


Assuntos
Células Germinativas/metabolismo , Gônadas/embriologia , Processos de Determinação Sexual , Diferenciação Sexual , Animais , Biomarcadores , Linhagem da Célula , Transdiferenciação Celular , Feminino , Células Germinativas/citologia , Gônadas/citologia , Masculino , Camundongos , Ovário/citologia , Ovário/embriologia , Ovário/metabolismo , Células de Sertoli
12.
PLoS One ; 7(5): e37826, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22662233

RESUMO

Outgrowth and patterning of the vertebrate limb requires a functional apical ectodermal ridge (AER). The AER is a thickening of ectodermal tissue located at the distal end of the limb bud. Loss of this structure, either through genetic or physical manipulations results in truncation of the limb. A number of genes, including Bmps, are expressed in the AER. Previously, it was shown that removal of the BMP receptor Bmpr1a specifically from the AER resulted in complete loss of hindlimbs suggesting that Bmp signaling in the AER is required for limb outgrowth. In this report, we genetically removed the three known AER-expressed Bmp ligands, Bmp2, Bmp4 and Bmp7 from the AER of the limb bud using floxed conditional alleles and the Msx2-cre allele. Surprisingly, only defects in digit patterning and not limb outgrowth were observed. In triple mutants, the anterior and posterior AER was present but loss of the central region of the AER was observed. These data suggest that Bmp ligands expressed in the AER are not required for limb outgrowth but instead play an essential role in maintaining the AER and patterning vertebrate digits.


Assuntos
Padronização Corporal/genética , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 7/genética , Extremidades/embriologia , Botões de Extremidades/crescimento & desenvolvimento , Alelos , Animais , Feminino , Fator 10 de Crescimento de Fibroblastos/genética , Fator 10 de Crescimento de Fibroblastos/metabolismo , Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/metabolismo , Deleção de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Deformidades Congênitas dos Membros/genética , Fator de Transcrição MSX1/genética , Fator de Transcrição MSX1/metabolismo , Masculino , Camundongos , Mutação , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo
14.
Curr Top Dev Biol ; 83: 151-83, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19118666

RESUMO

Sex determination in mammals results in two discrete sexes, male and female. The sexes are genetically distinct at fertilization (XY = male and XX = female). However, there is little evidence for differences in their development until mid-gestation when the gonadal primordium forms. Recent research suggests that signals within this tissue maintain the gonad in a bipotential state, balanced between two alternative fates, testis or ovary. At mid-gestation, expression of the Y-linked gene Sry in gonadal cells triggers the divergence of gonad development toward the testis pathway. The critical role of Sry may be simply to promote the stable expression of SOX9 in the key cell population in the gonad. Establishment of SOX9 in this lineage is opposed by female promoting factors that push the gonad toward an ovarian fate. Both the male and female sides of these antagonistic pathways are augmented by feedback loops and reinforcing signals that canalize development, once the initial choice is made. Hormones and growth factors produced by the developing testis regulate the male differentiation of the sex ducts and the external genitalia during fetal life. In contrast, the ovary is not required for the fetal development of female genital ducts or female external genitalia, as these organs develop in the absence of a gonad. At puberty, hormones produced by the testis or the ovary control the sex-specific differentiation of the musculature, mammary tissue, and body hair. Male or female development of the brain was previously thought to depend on hormones produced by the testis or ovary. However, recent evidence reveals expression differences between XX and XY brains prior to the time that hormones are circulating, suggesting that some influences on brain development may be autonomous to the cells of the brain.


Assuntos
Gônadas/embriologia , Processos de Determinação Sexual , Desenvolvimento Sexual , Animais , Diferenciação Celular , Feminino , Gônadas/citologia , Gônadas/crescimento & desenvolvimento , Gônadas/metabolismo , Humanos , Masculino , Camundongos , Modelos Biológicos , Caracteres Sexuais , Diferenciação Sexual , Desenvolvimento Sexual/genética
15.
Biol Reprod ; 79(4): 696-703, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18633141

RESUMO

MicroRNAs (miRNAs) are small noncoding RNAs that posttranscriptionally regulate gene expression. Hundreds of miRNAs are expressed in mammals; however, their functions are just starting to be uncovered. MicroRNAs are processed from a long hairpin mRNA transcript, down to a approximately 23-nucleotide duplex. The enzyme Dicer1 is required for miRNA processing, and mouse knockouts of Dicer1 are embryonic lethal before 7.5 days postcoitus. To examine the function of miRNAs specifically in the germline, we used a mouse model that expresses Cre recombinase from the TNAP locus and a floxed Dicer1 conditional allele. Removal of Dicer1 from germ cells resulted in male infertility. Germ cells were present in adult testes, but few tubules contained elongating spermatids. Germ cells that did differentiate to elongating spermatids exhibited abnormal morphology and motility. Rarely, sperm lacking Dicer1 could fertilize wild-type eggs to generate viable offspring. These results show that Dicer1 and miRNAs are essential for proper differentiation of the male germline.


Assuntos
Diferenciação Celular/genética , RNA Helicases DEAD-box/fisiologia , Endorribonucleases/fisiologia , Células Germinativas/fisiologia , Animais , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Endorribonucleases/genética , Endorribonucleases/metabolismo , Feminino , Células Germinativas/metabolismo , Infertilidade Masculina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/metabolismo , Processamento Pós-Transcricional do RNA/genética , Ribonuclease III , Motilidade dos Espermatozoides/genética , Espermatogênese/genética , Espermatogênese/fisiologia , Espermatozoides/metabolismo , Espermatozoides/fisiologia
16.
Development ; 133(17): 3411-8, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16887828

RESUMO

DNA methylation is necessary for the silencing of endogenous retrotransposons and the maintenance of monoallelic gene expression at imprinted loci and on the X chromosome. Dynamic changes in DNA methylation occur during the initial stages of primordial germ cell development; however, all consequences of this epigenetic reprogramming are not understood. DNA demethylation in postmigratory primordial germ cells coincides with erasure of genomic imprints and reactivation of the inactive X chromosome, as well as ongoing germ cell differentiation events. To investigate a possible role for DNA methylation changes in germ cell differentiation, we have studied several marker genes that initiate expression at this time. Here, we show that the postmigratory germ cell-specific genes Mvh, Dazl and Scp3 are demethylated in germ cells, but not in somatic cells. Premature loss of genomic methylation in Dnmt1 mutant embryos leads to early expression of these genes as well as GCNA1, a widely used germ cell marker. In addition, GCNA1 is ectopically expressed by somatic cells in Dnmt1 mutants. These results provide in vivo evidence that postmigratory germ cell-specific genes are silenced by DNA methylation in both premigratory germ cells and somatic cells. This is the first example of ectopic gene activation in Dnmt1 mutant mice and suggests that dynamic changes in DNA methylation regulate tissue-specific gene expression of a set of primordial germ cell-specific genes.


Assuntos
Metilação de DNA , Inativação Gênica , Células Germinativas/metabolismo , Animais , Sequência de Bases , Diferenciação Celular , Linhagem da Célula , Movimento Celular/genética , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Primers do DNA , Feminino , Células Germinativas/citologia , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sistema Urogenital/embriologia , Cromossomo X
17.
Stem Cells ; 23(9): 1314-23, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16051982

RESUMO

The capacity for cellular differentiation is governed not only by the repertoire of available transcription factors but by the accessibility of cis-regulatory elements. Studying changes in epigenetic modifications during stem cell differentiation will help us understand how cells maintain or lose differentiation potential. We investigated changes in DNA methylation during the transition of pluripotent embryonic stem cells (ESCs) into differentiated cell types. Using a methylation-sensitive restriction fingerprinting method, we identified a novel adenine nucleotide (ADP/ATP) translocase gene, Ant4, that was selectively hypomethylated and expressed in undifferentiated mouse ESCs. In contrast to other pluripotent stem cell-specific genes such as Oct-4 and Nanog, the Ant4 gene was readily derepressed in differentiated cells after 5-aza-2'-deoxycytidine treatment. Moreover, expression of de novo DNA methyltransferases Dnmt3a and Dnmt3b was essential for repression and DNA methylation of the Ant4 gene during ESC differentiation. Although the deduced amino acid sequence of Ant4 is highly homologous to the previously identified Ant isoforms, the expression of Ant4 was uniquely restricted to developing gametes in adult mice, and its promoter hypomethylation was observed only in testis. Additionally, Ant4 was expressed in primordial germ cells. These data indicate that Ant4 is a pluripotent stem cell- and germ cell-specific isoform of adenine nucleotide translocase in mouse and that DNA methylation plays a primary role in its transcriptional silencing in somatic cells.


Assuntos
Metilação de DNA , Inativação Gênica , Translocases Mitocondriais de ADP e ATP/genética , Células-Tronco/enzimologia , Animais , Sequência de Bases , Diferenciação Celular/genética , DNA (Citosina-5-)-Metiltransferases/deficiência , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica , Masculino , Camundongos , Translocases Mitocondriais de ADP e ATP/biossíntese , Dados de Sequência Molecular , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Espermatozoides/enzimologia , Células-Tronco/citologia , Testículo/citologia , DNA Metiltransferase 3B
18.
Dev Biol ; 258(1): 201-8, 2003 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12781693

RESUMO

The initial cohort of mammalian gametes is established by the proliferation of primordial germ cells in the early embryo. Primordial germ cells first appear in extraembyronic tissues and subsequently migrate to the developing gonad. Soon after they arrive in the gonad, the germ cells cease dividing and undertake sexually dimorphic patterns of development. Male germ cells arrest mitotically, while female germ cells directly enter meiotic prophase I. These sex-specific differentiation events are imposed upon a group of sex-common differentiation events that are shared by XX and XY germ cells. We have studied the appearance of GCNA1, a postmigratory sex-common germ cell marker, in cultures of premigratory germ cells to investigate how this differentiation program is regulated. Cultures in which proliferation was either inhibited or stimulated displayed a similar extent of differentiation as controls, suggesting that some differentiation events are the result of a cell-intrinsic program and are independent of cell proliferation. We also found that GCNA1 expression was accelerated by agents which promote DNA demethylation or histone acetylation. These results suggest that genomic demethylation of proliferative phase primordial germ cells is a mechanism by which germ cell maturation is coordinated.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Metilação de DNA , Células Germinativas/fisiologia , Fosfatase Alcalina/análise , Animais , Antígenos Nucleares/metabolismo , Azacitidina/farmacologia , Biomarcadores , Divisão Celular , Células Cultivadas , Feminino , Células Germinativas/efeitos dos fármacos , Células Germinativas/metabolismo , Ácidos Hidroxâmicos/farmacologia , Masculino , Meiose , Camundongos , Camundongos Endogâmicos , Proteínas Nucleares/biossíntese , Oogônios/citologia , Oogônios/efeitos dos fármacos , Oogônios/metabolismo , Diferenciação Sexual , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Espermatogônias/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Tretinoína/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa