Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 134(5)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38426493

RESUMO

Early gestational loss occurs in approximately 20% of all clinically recognized human pregnancies and is an important cause of morbidity. Either embryonic or maternal defects can cause loss, but a functioning and receptive uterine endometrium is crucial for embryo implantation. We report that the switch/sucrose nonfermentable (SWI/SNF) remodeling complex containing polybromo-1 (PBRM1) and Brahma-related gene 1 (BRG1) is essential for implantation of the embryonic blastocyst on the wall of the uterus in mice. Although preimplantation development is unaffected, conditional ablation of Pbrm1 in uterine stromal cells disrupts progesterone pathways and uterine receptivity. Heart and neural crest derivatives expressed 2 (Hand2) encodes a basic helix-loop-helix (bHLH) transcription factor required for embryo implantation. We identify an enhancer of the Hand2 gene in stromal cells that requires PBRM1 for epigenetic histone modifications/coactivator recruitment and looping with the promoter. In Pbrm1cKO mice, perturbation of chromatin assembly at the promoter and enhancer sites compromises Hand2 transcription, adversely affects fibroblast growth factor signaling pathways, prevents normal stromal-epithelial crosstalk, and disrupts embryo implantation. The mutant female mice are infertile and provide insight into potential causes of early pregnancy loss in humans.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina , Animais , Feminino , Humanos , Camundongos , Gravidez , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cromatina/metabolismo , Implantação do Embrião/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Útero
2.
Trends Genet ; 40(3): 238-249, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38262796

RESUMO

Maternal mRNAs accumulate during egg growth and must be judiciously degraded or translated to ensure successful development of mammalian embryos. In this review we integrate recent investigations into pathways controlling rapid degradation of maternal mRNAs during the maternal-to-zygotic transition. Degradation is not indiscriminate, and some mRNAs are selectively protected and rapidly translated after fertilization for reprogramming the zygotic genome during early embryogenesis. Oocyte specific cofactors and pathways have been illustrated to control different futures of maternal mRNAs. We discuss mechanisms that control the fate of maternal mRNAs during late oogenesis and after fertilization. Issues to be resolved in current maternal mRNA research are described, and future research directions are proposed.


Assuntos
Desenvolvimento Embrionário , RNA Mensageiro Estocado , Animais , RNA Mensageiro Estocado/genética , RNA Mensageiro Estocado/metabolismo , Desenvolvimento Embrionário/genética , Oócitos , Oogênese/genética , Zigoto , Regulação da Expressão Gênica no Desenvolvimento/genética , Mamíferos/genética
3.
Cell Rep ; 42(10): 113247, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37831603

RESUMO

Perturbing the transcriptome of mammalian oocytes results in meiotic failure. We previously reported that RNA-exosome-associated RNase, EXOSC10, degrades unwanted protein-coding RNA and processes ribosomal RNA to ensure proper oocyte maturation. Here, we establish oocyte-specific knockout mice of another RNA-exosome-associated RNase, DIS3. Mutant females (Dis3cKO) exhibit significantly reduced fertility because oocytes arrest after the growth phase. Single-oocyte RNA sequencing (RNA-seq) and CUT&Tag analyses show that DIS3 degrades intergenic RNA and mediates transcription silencing that is essential for chromatin condensation and resumption of meiosis. Dis3cKO oocytes exhibit elevated H3K27me3 in a pre-defined manner due to insufficient demethylation. During oocyte growth, EXOSC10 functions with DIS3 to degrade intergenic RNA. Double-knockout oocytes have earlier growth defects and more accumulated transcripts. We conclude that RNA exosomes synergistically degrade unwanted RNA and mediate transcription termination to ensure transcriptome integrity during oocyte development.


Assuntos
Exossomos , RNA Polimerase II , Camundongos , Animais , Feminino , RNA Polimerase II/metabolismo , Exossomos/metabolismo , Oócitos/metabolismo , Meiose , RNA/metabolismo , Endorribonucleases/metabolismo , Fertilidade/genética , Mamíferos/metabolismo
4.
Dev Cell ; 58(18): 1716-1732.e8, 2023 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-37714160

RESUMO

Early mammalian development occurs during embryo transit of the female reproductive tract. Transport is orchestrated by secreted oviduct fluid, unidirectional beating of epithelial cilia, and smooth muscle contractions. Using gene-edited mice, we document that conditional disruption of a component of the SWI/SNF chromatin remodeling complex in smooth muscle cells prevents transport through the oviduct without perturbing embryogenesis. Analysis with RNA sequencing (RNA-seq), transposase-accessible chromatin with sequencing (ATAC-seq), chromatin immunocleavage sequencing (ChIC-seq), and pharmacologic rescue experiments implicated prostaglandin signaling pathways. In comparison with controls, gene-edited mice had compromised chromatin accessibility at enhancer/promoters of Ptgs2, Pla2g16, Pla2r1, and Ptger3 (EP3) as well as decreased enhancer-promoter interactive looping critical for Ptgs2 (aka Cox-2) expression in a SWI/SNF complex-dependent manner. Treatment of wild-type mice with prostaglandin inhibitors phenocopied the genetically induced defect.


Assuntos
Montagem e Desmontagem da Cromatina , Prostaglandinas , Feminino , Animais , Camundongos , Ciclo-Oxigenase 2/genética , Músculo Liso , Cromatina , Mamíferos
5.
Genes Dev ; 37(9-10): 418-431, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37257918

RESUMO

Translation of maternal mRNAs is detected before transcription of zygotic genes and is essential for mammalian embryo development. How certain maternal mRNAs are selected for translation instead of degradation and how this burst of translation affects zygotic genome activation remain unknown. Using gene-edited mice, we document that the oocyte-specific eukaryotic translation initiation factor 4E family member 1b (eIF4E1b) is the regulator of maternal mRNA expression that ensures subsequent reprogramming of the zygotic genome. In oocytes, eIF4E1b binds to transcripts encoding translation machinery proteins, chromatin remodelers, and reprogramming factors to promote their translation in zygotes and protect them from degradation. The protein products are thought to establish an open chromatin landscape in one-cell zygotes to enable transcription of genes required for cleavage stage development. Our results define a program for rapid resetting of the zygotic epigenome that is regulated by maternal mRNA expression and provide new insights into the mammalian maternal-to-zygotic transition.


Assuntos
RNA Mensageiro Estocado , Zigoto , Animais , Camundongos , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Oócitos , Biossíntese de Proteínas , RNA Mensageiro Estocado/genética , RNA Mensageiro Estocado/metabolismo , Zigoto/metabolismo
6.
Nucleic Acids Res ; 51(7): 3078-3093, 2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-36727488

RESUMO

During oocyte development in mice, transcripts accumulate in the growth phase and are subsequently degraded during maturation. At the transition point between growth and maturation, oocytes have an intact nucleus or germinal vesicle (GV), and terminal uridylation labels RNA for degradation in meiosis I. By profiling the transcriptome using single-oocyte long-read PacBio RNA sequencing, we document that a small cohort of mRNAs are polyadenylated after terminal uridylation in GV oocytes [designated uridylated-poly(A) RNA]. Because DIS3L2 ribonuclease is known to degrade uridylated transcripts, we established oocyte-specific Dis3l2 knockout mice (Dis3l2cKO). Upon DIS3L2 depletion, uridylated-poly(A) RNAs remain intact which increases their abundance, and they predominate in the transcriptome of Dis3l2cKO oocytes. The abundance of uridylated-poly(A) RNA in Dis3l2cKO oocytes arises not only from insufficient degradation, but also from the stabilizing effect of subsequent polyadenylation. Uridylated-poly(A) RNAs have shorter poly(A) tails and their translation activity decreases in Dis3l2cKO oocytes. Almost all Dis3l2cKO oocytes arrest at the GV stage, and female mice are infertile. Our study demonstrates multiple fates for RNA after terminal uridylation and highlights the role of DIS3L2 ribonuclease in safeguarding the transcriptome and ensuring female fertility.


Assuntos
Exorribonucleases , Fertilidade , Animais , Feminino , Camundongos , Oócitos/metabolismo , RNA/metabolismo , RNA Mensageiro/metabolismo , Exorribonucleases/metabolismo
7.
Cell Rep ; 42(2): 112047, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36724075

RESUMO

Mammalian development is precisely controlled by cell differentiation. Identifying new regulators and investigating their interactions provide insight into genetic networks defining pre-implantation development. We established a knockout mouse model of Dis3, an exosome associated ribonuclease. Homozygous Dis3 null embryos arrest at the morula stage of development. Using single-embryo RNA sequencing (RNA-seq), we observed persistence of Pou6f1 mRNA in homozygous null Dis3 embryos and that the cognate protein represses transcription of Nanog and Cdx2. The resultant defects in cell differentiation disrupt the morula-to-blastocyst transition and are embryonic lethal. Microinjection of Dis3 mRNA into zygotes rescues the phenotype. Point mutations of Dis3 ribonuclease in individual blastomeres prevents their incorporation into embryos. To overcome the paucity of embryos, we derived homozygous Dis3 null mouse embryonic stem cells to identify additional gene targets of POU6F1. Our findings delineate a regulatory pathway of DIS3-POU6F1 in pre-implantation mammalian embryogenesis.


Assuntos
Diferenciação Celular , Desenvolvimento Embrionário , Ribonucleases , Animais , Camundongos , Blastocisto/metabolismo , Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Mamíferos/metabolismo , Ribonucleases/metabolismo , RNA Mensageiro/metabolismo
8.
PLoS Genet ; 17(4): e1009485, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33831001

RESUMO

piRNAs are small non-coding RNAs required to maintain genome integrity and preserve RNA homeostasis during male gametogenesis. In murine adult testes, the highest levels of piRNAs are present in the pachytene stage of meiosis, but their mode of action and function remain incompletely understood. We previously reported that BTBD18 binds to 50 pachytene piRNA-producing loci. Here we show that spermatozoa in gene-edited mice lacking a BTBD18 targeted pachytene piRNA cluster on Chr18 have severe sperm head dysmorphology, poor motility, impaired acrosome exocytosis, zona pellucida penetration and are sterile. The mutant phenotype arises from aberrant formation of proacrosomal vesicles, distortion of the trans-Golgi network, and up-regulation of GOLGA2 transcripts and protein associated with acrosome dysgenesis. Collectively, our findings reveal central role of pachytene piRNAs in controlling spermiogenesis and male fertility.


Assuntos
Infertilidade Masculina/genética , RNA Interferente Pequeno/genética , Espermatogênese/genética , Espermatozoides/patologia , Acrossomo/patologia , Animais , Cromossomos/genética , Humanos , Infertilidade Masculina/patologia , Masculino , Meiose/genética , Camundongos , Estágio Paquíteno/genética , Espermátides/crescimento & desenvolvimento , Espermátides/patologia , Testículo/crescimento & desenvolvimento , Testículo/patologia
9.
Curr Top Dev Biol ; 140: 317-340, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32591079

RESUMO

Mammalian embryogenesis depends on maternal factors accumulated in eggs prior to fertilization and on placental transfers later in gestation. In this review, we focus on initial events when the organism has insufficient newly synthesized embryonic factors to sustain development. These maternal factors regulate preimplantation embryogenesis both uniquely in pronuclear formation, genome reprogramming and cell fate determination and more universally in regulating cell division, transcription and RNA metabolism. Depletion, disruption or inappropriate persistence of maternal factors can result in developmental defects in early embryos. To better understand the origins of these maternal effects, we include oocyte maturation processes that are responsible for their production. We focus on recent publications and reference comprehensive reviews that include earlier scientific literature of early mouse development.


Assuntos
Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Genoma/genética , Oócitos/metabolismo , Zigoto/metabolismo , Animais , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Feminino , Herança Materna/genética , Camundongos , Oócitos/citologia , Zigoto/citologia
10.
Nucleic Acids Res ; 48(10): 5349-5365, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32313933

RESUMO

Growing mammalian oocytes accumulate substantial amounts of RNA, most of which is degraded during subsequent meiotic maturation. The growth-to-maturation transition begins with germinal vesicle or nuclear envelope breakdown (GVBD) and is critical for oocyte quality and early development. The molecular machinery responsible for the oocyte transcriptome transition remains unclear. Here, we report that an exosome-associated RNase, EXOSC10, sculpts the transcriptome to facilitate the growth-to-maturation transition of mouse oocytes. We establish an oocyte-specific conditional knockout of Exosc10 in mice using CRISPR/Cas9 which results in female subfertility due to delayed GVBD. By performing multiple single oocyte RNA-seq, we document dysregulation of several types of RNA, and the mRNAs that encode proteins important for endomembrane trafficking and meiotic cell cycle. As expected, EXOSC10-depleted oocytes have impaired endomembrane components including endosomes, lysosomes, endoplasmic reticulum and Golgi. In addition, CDK1 fails to activate, possibly due to persistent WEE1 activity, which blocks lamina phosphorylation and disassembly. Moreover, we identified rRNA processing defects that cause higher percentage of developmentally incompetent oocytes after EXOSC10 depletion. Collectively, we propose that EXOSC10 promotes normal growth-to-maturation transition in mouse oocytes by sculpting the transcriptome to degrade RNAs encoding growth-phase factors and, thus, support the maturation phase of oogenesis.


Assuntos
Exorribonucleases/fisiologia , Complexo Multienzimático de Ribonucleases do Exossomo/fisiologia , Oócitos/crescimento & desenvolvimento , Oócitos/metabolismo , Oogênese , Transcriptoma , Animais , Proteína Quinase CDC2/metabolismo , Exorribonucleases/genética , Complexo Multienzimático de Ribonucleases do Exossomo/genética , Feminino , Infertilidade Feminina/genética , Membranas Intracelulares/metabolismo , Camundongos , Lâmina Nuclear/metabolismo , Poli A , RNA/metabolismo , Precursores de RNA/metabolismo , Processamento Pós-Transcricional do RNA , RNA Ribossômico/metabolismo , RNA-Seq
11.
Nucleic Acids Res ; 48(7): 3525-3541, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32086523

RESUMO

Germ-cell transcription factors control gene networks that regulate oocyte differentiation and primordial follicle formation during early, postnatal mouse oogenesis. Taking advantage of gene-edited mice lacking transcription factors expressed in female germ cells, we analyzed global gene expression profiles in perinatal ovaries from wildtype, FiglaNull, Lhx8Null and Sohlh1Null mice. Figla deficiency dysregulates expression of meiosis-related genes (e.g. Sycp3, Rad51, Ybx2) and a variety of genes (e.g. Nobox, Lhx8, Taf4b, Sohlh1, Sohlh2, Gdf9) associated with oocyte growth and differentiation. The absence of FIGLA significantly impedes meiotic progression, causes DNA damage and results in oocyte apoptosis. Moreover, we find that FIGLA and other transcriptional regulator proteins (e.g. NOBOX, LHX8, SOHLH1, SOHLH2) are co-expressed in the same subset of germ cells in perinatal ovaries and Figla ablation dramatically disrupts KIT, NOBOX, LHX8, SOHLH1 and SOHLH2 abundance. In addition, not only do FIGLA, LHX8 and SOHLH1 cross-regulate each other, they also cooperate by direct interaction with each during early oocyte development and share downstream gene targets. Thus, our findings substantiate a major role for FIGLA, LHX8 and SOHLH1 as multifunctional regulators of networks necessary for oocyte maintenance and differentiation during early folliculogenesis.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Redes Reguladoras de Genes , Proteínas com Homeodomínio LIM/metabolismo , Oócitos/metabolismo , Oogênese/genética , Fatores de Transcrição/metabolismo , Animais , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proliferação de Células/genética , Dano ao DNA , Feminino , Regulação da Expressão Gênica , Células HEK293 , Humanos , Proteínas com Homeodomínio LIM/genética , Meiose/genética , Camundongos , Oócitos/citologia , Ovário/metabolismo , Fatores de Transcrição/genética
12.
Nat Commun ; 10(1): 5196, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31729367

RESUMO

Spermatogonial stem cells (SSCs) have the dual capacity to self-renew and differentiate into progenitor spermatogonia that develop into mature spermatozoa. Here, we document that preferentially expressed antigen of melanoma family member 12 (PRAMEF12) plays a key role in maintenance of the spermatogenic lineage. In male mice, genetic ablation of Pramef12 arrests spermatogenesis and results in sterility which can be rescued by transgenic expression of Pramef12. Pramef12 deficiency globally decreases expression of spermatogenic-related genes, and single-cell transcriptional analysis of post-natal male germline cells identifies four spermatogonial states. In the absence of Pramef12 expression, there are fewer spermatogonial stem cells which exhibit lower expression of SSC maintenance-related genes and are defective in their ability to differentiate. The disruption of the first wave of spermatogenesis in juvenile mice results in agametic seminiferous tubules. These observations mimic a Sertoli cell-only syndrome in humans and may have translational implications for reproductive medicine.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células de Sertoli/metabolismo , Espermatogênese , Espermatogônias/metabolismo , Animais , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Análise de Sequência de RNA , Células de Sertoli/citologia , Espermatogônias/citologia
13.
Nat Commun ; 10(1): 2926, 2019 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-31249315

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

14.
Nat Commun ; 10(1): 2271, 2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-31118423

RESUMO

Following fertilization, cortical granules exocytose ovastacin, a metalloendopeptidase that cleaves ZP2 in the zona pellucida surrounding mouse eggs to prevent additional sperm binding. Using high- and super-resolution imaging with ovastacinmCherry as a fluorescent marker, we characterize cortical granule dynamics at single granule resolution in transgenic mouse eggs. Newly-developed imaging protocols provide an unprecedented view of vesicular dynamics near the plasma membrane in mouse eggs. We discover that cortical granule anchoring in the cortex is dependent on maternal MATER and document that myosin IIA is required for biphasic trafficking to the plasma membrane. We observe local clearance of cortical actin during exocytosis and determine that pharmacologic or genetic disruption of trafficking to the plasma membrane impairs secretion of cortical granules and results in polyspermy. Thus, the regulation of cortical granule dynamics at the cortex-plasma membrane interface is critical for exocytosis and the post-fertilization block to sperm binding that ensures monospermic fertilization.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Exocitose/fisiologia , Interações Espermatozoide-Óvulo/fisiologia , Zona Pelúcida/metabolismo , Animais , Antígenos/metabolismo , Membrana Celular/metabolismo , Proteínas do Ovo/metabolismo , Feminino , Microscopia Intravital , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Masculino , Metaloproteases/química , Metaloproteases/genética , Metaloproteases/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Glicoproteínas da Zona Pelúcida/metabolismo
15.
Dev Cell ; 46(5): 627-640.e5, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30122633

RESUMO

The zona pellucida surrounding ovulated eggs regulates monospermic fertilization necessary for successful development. Using mouse transgenesis, we document that the N terminus of ZP2 is sufficient for sperm binding to the zona matrix and for in vivo fertility. Sperm binding is independent of ZP2 glycans and does not occur after complete cleavage of ZP2 by ovastacin, a zinc metalloendopeptidase stored in egg cortical granules. Immediately following fertilization, a rapid block to sperm penetration of the zona pellucida is established that precedes ZP2 cleavage but requires ovastacin enzymatic activity. This block to penetration is associated with release of zinc from cortical granules coincident with exocytosis. High levels of zinc affect forward motility of sperm to prevent their passage through the zona matrix. This transient, post-fertilization block to sperm penetration provides a temporal window to complete the cleavage of ZP2, which prevents sperm binding to ensure monospermy.


Assuntos
Polissacarídeos/farmacologia , Interações Espermatozoide-Óvulo , Espermatozoides/metabolismo , Zinco/metabolismo , Glicoproteínas da Zona Pelúcida/metabolismo , Zona Pelúcida/metabolismo , Animais , Comunicação Celular , Exocitose , Feminino , Fertilização , Masculino , Camundongos , Camundongos Transgênicos , Espermatozoides/citologia , Espermatozoides/efeitos dos fármacos , Zona Pelúcida/efeitos dos fármacos , Glicoproteínas da Zona Pelúcida/genética
16.
Nat Commun ; 8(1): 1643, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29158485

RESUMO

Degradation of maternal proteins by the ubiquitin-proteasome system (UPS) accompanies the maternal-to-zygotic transition. DPPA3/Stella/PGC7, encoded by a maternal effect gene, is present in the nucleus and cytoplasm of zygotes and has been associated with protecting the female pronucleus from TET3-mediated demethylation. We now report that cytoplasmic DPPA3 is partially cleaved by the ubiquitin-proteasome system and an N-terminus fragment remains in the cytoplasm where it associates with early and re-cycling endosomes. If DPPA3 is absent or if cleavage is prevented, multiple vesicles coalesce/aggregate and markers of lysosomes are decreased. Fertilized eggs develop poorly into blastocysts, which results in significantly decreased fecundity of Dppa3 R60A transgenic mice. This phenocopies aspects of Lamp1/2 knockdowns and Dppa3 KO embryos can be partially rescued in vitro by DPPA31-60 and to a lesser extent by LAMP1/2. Thus, the N-terminus of DPPA3 has a significant role in cytoplasmic vesicular trafficking in addition to its previously reported nuclear function.


Assuntos
Citoplasma/metabolismo , Camundongos/embriologia , Proteínas Repressoras/metabolismo , Motivos de Aminoácidos , Animais , Blastocisto/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteínas Cromossômicas não Histona , Citoplasma/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Desenvolvimento Embrionário , Feminino , Regulação da Expressão Gênica no Desenvolvimento , /metabolismo , Masculino , Camundongos/genética , Camundongos/metabolismo , Camundongos Knockout , Gravidez , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Transporte Proteico , Proteólise , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Zigoto/citologia , Zigoto/metabolismo
17.
Dev Cell ; 40(5): 453-466.e5, 2017 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-28292424

RESUMO

PIWI-interacting RNAs (piRNAs) are small non-coding RNAs essential for animal germ cell development. Despite intense investigation of post-transcriptional processing, chromatin regulators for piRNA biogenesis in mammals remain largely unexplored. Here we document that BTBD18 is a pachytene nuclear protein in mouse testes that occupies a subset of pachytene piRNA-producing loci. Ablation of Btbd18 in mice disrupts piRNA biogenesis, prevents spermiogenesis, and results in male sterility. Transcriptome profiling, chromatin accessibility, and RNA polymerase II occupancy demonstrate that BTBD18 facilitates expression of pachytene piRNA precursors by promoting transcription elongation. Thus, our study identifies BTBD18 as a specific controller for transcription activation through RNA polymerase II elongation at a subset of genomic piRNA loci.


Assuntos
Loci Gênicos , Proteínas Nucleares/metabolismo , RNA Interferente Pequeno/metabolismo , Elongação da Transcrição Genética , Animais , Apoptose/genética , Deleção de Genes , Células Germinativas/citologia , Células Germinativas/metabolismo , Masculino , Meiose/genética , Camundongos , Precursores de RNA/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espermatogênese/genética , Testículo/citologia
18.
PLoS Genet ; 13(1): e1006580, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28114310

RESUMO

Monospermic fertilization is mediated by the extracellular zona pellucida composed of ZP1, ZP2 and ZP3. Sperm bind to the N-terminus of ZP2 which is cleaved after fertilization by ovastacin (encoded by Astl) exocytosed from egg cortical granules to prevent sperm binding. AstlNull mice lack the post-fertilization block to sperm binding and the ability to rescue this phenotype with AstlmCherry transgenic mice confirms the role of ovastacin in providing a definitive block to polyspermy. During oogenesis, endogenous ovastacin traffics through the endomembrane system prior to storage in peripherally located cortical granules. Deletion mutants of ovastacinmCherry expressed in growing oocytes define a unique 7 amino acid motif near its N-terminus that is necessary and sufficient for cortical granule localization. Deletion of the 7 amino acids by CRISPR/Cas9 at the endogenous locus (AstlΔ) prevents cortical granule localization of ovastacin. The misdirected enzyme is present within the endomembrane system and ZP2 is prematurely cleaved. Sperm bind poorly to the zona pellucida of AstlΔ/Δ mice with partially cleaved ZP2 and female mice are sub-fertile.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Fertilização , Metaloproteases/metabolismo , Oócitos/metabolismo , Sinais Direcionadores de Proteínas , Glicoproteínas da Zona Pelúcida/metabolismo , Animais , Feminino , Metaloproteases/química , Metaloproteases/genética , Camundongos , Transporte Proteico , Proteólise
19.
Development ; 144(3): 519-528, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27993980

RESUMO

During development from oocyte to embryo, genetic programs in mouse germ cells are reshaped by chromatin remodeling to orchestrate the onset of development. Epigenetic modifications of specific amino acid residues of core histones and their isoforms can dramatically alter activation and suppression of gene expression. H3.3 is a histone H3 variant that plays essential roles in mouse oocytes and early embryos, but the functional role of individual amino acid residues has been unclear because of technical hurdles. Here, we describe two strategies that successfully investigated the functions of three individual H3.3 residues in oogenesis, cleavage-stage embryogenesis and early development. We first generated genetic mosaic ovaries and blastocysts with stochastic expression of wild-type or mutant H3.3 alleles and showed dominant negative effects of H3.3R26 and H3.3K27 in modulating oogenesis and partitioning cells to the inner cell mass of the early embryo. Time-lapse imaging assays also revealed the essential roles of H3.3K56 in efficient H2B incorporation and paternal pronuclei formation. Application of these strategies can be extended to investigate roles of additional H3.3 residues and has implications for use in other developmental systems.


Assuntos
Blastocisto/metabolismo , Histonas/metabolismo , Oócitos/metabolismo , Animais , Blastocisto/citologia , Massa Celular Interna do Blastocisto/citologia , Massa Celular Interna do Blastocisto/metabolismo , Blastômeros/citologia , Blastômeros/metabolismo , Linhagem da Célula , Montagem e Desmontagem da Cromatina , Epigênese Genética , Feminino , Histonas/química , Histonas/genética , Masculino , Camundongos , Camundongos Transgênicos , Mosaicismo , Oogênese , Imagem com Lapso de Tempo , Zigoto/citologia , Zigoto/metabolismo
20.
J Mol Cell Biol ; 8(4): 369-70, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27339058
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...