Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Development ; 146(23)2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31704792

RESUMO

The number and quality of oocytes within the ovarian reserve largely determines fertility and reproductive lifespan in mammals. An oocyte-specific transcription factor cascade controls oocyte development, and some of these transcription factors, such as newborn ovary homeobox gene (NOBOX), are candidate genes for primary ovarian insufficiency in women. Transcription factors are frequently modified by the post-translational modification SUMOylation, but it is not known whether SUMOylation is required for function of the oocyte-specific transcription factors or if SUMOylation is required in oocytes during their development within the ovarian follicle. To test this, the sole E2 SUMO-conjugating enzyme, Ube2i, was ablated in mouse oocytes beginning in primordial follicles. Loss of oocyte Ube2i resulted in female infertility with major defects in stability of the primordial follicle pool, ovarian folliculogenesis, ovulation and meiosis. Transcriptomic profiling of ovaries suggests that loss of oocyte Ube2i caused defects in both oocyte- and granulosa cell-expressed genes, including NOBOX and some of its known target genes. Together, these studies show that SUMOylation is required in the mammalian oocyte during folliculogenesis for both oocyte development and communication with ovarian somatic cells.


Assuntos
Comunicação Celular , Células da Granulosa , Infertilidade Feminina , Oócitos/metabolismo , Sumoilação , Enzimas de Conjugação de Ubiquitina/deficiência , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células da Granulosa/metabolismo , Células da Granulosa/patologia , Infertilidade Feminina/embriologia , Infertilidade Feminina/genética , Infertilidade Feminina/patologia , Camundongos , Camundongos Knockout , Oócitos/patologia , Enzimas de Conjugação de Ubiquitina/metabolismo
2.
Mol Biol Cell ; 29(22): 2784-2799, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30188763

RESUMO

Mice that lack the epidermal growth factor receptor (EGFR) fail to develop a hair coat, but the mechanism responsible for this deficit is not completely understood. Here, we show that EGFR plays a critical role to attenuate wingless-type MMTV integration site family member (Wnt)/ß-catenin signaling during postnatal hair follicle development. Genetic ablation of EGFR in mice resulted in increased mitotic activity in matrix cells, apoptosis in hair follicles, and impaired differentiation of epithelial lineages that form hair. EGFR is activated in wild-type hair follicle stem cells marked with SOX9 or NFATc1 and is essential to restrain proliferation and support stem cell numbers and their quiescence. We observed elevated levels of Wnt4, 6, 7b, 10a, 10b, and 16 transcripts and hyperactivation of the ß-catenin pathway in EGFR knockout follicles. Using primary keratinocytes, we linked ligand-induced EGFR activation to suppression of nascent mRNA synthesis of Wnt genes. Overexpression of the Wnt antagonist sFRP1 in mice lacking EGFR demonstrated that elevated Wnts are a major cause for the hair follicle defects. Colocalization of transforming growth factor α and Wnts regulated by EGFR in stem cells and progeny indicates that EGFR autocrine loops control Wnts. Our findings define a novel mechanism that integrates EGFR and Wnt/ß-catenin pathways to coordinate the delicate balance between proliferation and differentiation during development.


Assuntos
Receptores ErbB/metabolismo , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/metabolismo , Via de Sinalização Wnt , Animais , Apoptose/efeitos dos fármacos , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Introdução de Genes , Cabelo/crescimento & desenvolvimento , Folículo Piloso/citologia , Ligantes , Camundongos , Mitose/efeitos dos fármacos , Modelos Biológicos , Morfogênese/efeitos dos fármacos , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Pele/crescimento & desenvolvimento , Transcrição Gênica/efeitos dos fármacos , Fator de Crescimento Transformador alfa/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Proteínas ras/metabolismo
3.
J Cell Sci ; 130(10): 1835-1844, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28389581

RESUMO

Spermiogenesis includes transcriptional silencing, chromatin condensation and extensive morphological changes as spermatids transform into sperm. Chromatin condensation involves histone hyperacetylation, transitory DNA breaks, histone H2AX (also known as H2AFX) phosphorylation at Ser139 (γH2AX), and replacement of histones by protamines. Previously, we have reported that the spermatid protein kinase TSSK6 is essential for fertility in mice, but its specific role in spermiogenesis is unknown. Here, we show that TSSK6 expression is spatiotemporally coincident with γH2AX formation in the nuclei of developing mouse spermatids. RNA-sequencing analysis demonstrates that genetic ablation of Tssk6 does not impact gene expression or silencing in spermatids. However, loss of TSSK6 blocks γH2AX formation, even though the timing and level of the transient DNA breaks is unaltered. Further, Tssk6-knockout sperm contained increased levels of histones H3 and H4, and protamine 2 precursor and intermediate(s) indicative of a defective histone-to-protamine transition. These results demonstrate that TSSK6 is required for γH2AX formation during spermiogenesis, and also link γH2AX to the histone-to-protamine transition and male fertility.


Assuntos
Histonas/metabolismo , Protaminas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Espermatogênese , Animais , Núcleo Celular/metabolismo , Quebras de DNA de Cadeia Dupla , Deleção de Genes , Masculino , Camundongos Knockout , Espermátides/metabolismo , Fatores de Tempo , Transcriptoma/genética
4.
Biol Reprod ; 95(2): 44, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27335065

RESUMO

Pregnancy is a complex physiological process tightly controlled by the interplay among hormones, morphogens, transcription factors, and signaling pathways. Although recent studies using genetically engineered mouse models have revealed that ligands and receptors of transforming growth factor beta (TGFbeta) and bone morphogenetic protein (BMP) signaling pathways are essential for multiple reproductive events during pregnancy, the functional role of SMAD transcription factors, which serve as the canonical signaling platform for the TGFbeta/BMP pathways, in the oviduct and uterus is undefined. Here, we used a mouse model containing triple conditional deletion of the BMP receptor signaling Smads (Smad1 and Smad5) and Smad4, the central mediator of both TGFbeta and BMP signaling, to investigate the role of the SMADs in reproductive tract structure and function in cells from the Amhr2 lineage. Unlike the respective single- or double-knockouts, female Smad1(flox/flox) Smad5(flox/flox) Smad4(flox/flox) Amhr2(cre/+)conditional knockout (i.e., Smad1/5/4-Amhr2-cre KO) mice are sterile. We discovered that Smad1/5/4-Amhr2-cre KO females have malformed oviducts that subsequently develop oviductal diverticuli. These oviducts showed dysregulation of multiple genes essential for oviduct and smooth muscle development. In addition, uteri from Smad1/5/4-Amhr2-cre KO females exhibit multiple defects in stroma, epithelium, and smooth muscle layers and fail to assemble a closed uterine lumen upon embryo implantation, with defective uterine decidualization that led to pregnancy loss at early to mid-gestation. Taken together, our study uncovers a new role for the SMAD transcription factors in maintaining the structural and functional integrity of oviduct and uterus, required for establishment and maintenance of pregnancy.


Assuntos
Tubas Uterinas/metabolismo , Oviductos/metabolismo , Reprodução/fisiologia , Transdução de Sinais/fisiologia , Proteínas Smad/metabolismo , Útero/metabolismo , Animais , Implantação do Embrião/fisiologia , Tubas Uterinas/anormalidades , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Músculo Liso/anormalidades , Músculo Liso/metabolismo , Oviductos/anormalidades , Gravidez , Proteínas Smad/genética , Útero/anormalidades , Útero/fisiologia
5.
Mol Endocrinol ; 28(11): 1887-98, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25243859

RESUMO

Molecular changes that give rise to granulosa cell tumors of the ovary are not well understood. Previously, we showed that deletion in granulosa cells of the bone morphogenetic protein receptor-signaling transcription factors, Smad1 and Smad5, causes development of metastatic granulosa cell tumors that phenocopy the juvenile form of granulosa cell tumors (JGCTs) in humans. The TGFß-SMAD2/3 pathway is active in JGCTs, but its role is unknown. We tested the in vivo contribution of TGFß-SMAD signaling to JGCT development by genetically deleting the common Smad4 from Smad1/5 double knockout mice. Smad1/5/4 triple knockout mice were sterile and had significantly increased survival and delayed tumor development compared to those for the Smad1/5 double knockout mice. The few tumors that did develop were smaller, showed no evidence of metastasis, and had increased apoptosis. In the human JGCT cell line COV434, TGFß1 increased viability by inhibiting apoptosis through a TGFß type I receptor-dependent repression of caspase activity and inhibition of poly(ADP-ribose) polymerase cleavage. These data support a tumor-promoting function of TGFß in JGCTs through its ability to repress apoptosis.


Assuntos
Apoptose/genética , Carcinogênese/genética , Células da Granulosa/metabolismo , Células da Granulosa/patologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Animais , Carcinogênese/patologia , Sobrevivência Celular/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Feminino , Tumor de Células da Granulosa/genética , Tumor de Células da Granulosa/metabolismo , Tumor de Células da Granulosa/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/genética , Proteínas Smad/genética , Proteínas Smad/metabolismo
6.
PLoS Genet ; 9(11): e1003863, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24244176

RESUMO

Implantation of a blastocyst in the uterus is a multistep process tightly controlled by an intricate regulatory network of interconnected ovarian, uterine, and embryonic factors. Bone morphogenetic protein (BMP) ligands and receptors are expressed in the uterus of pregnant mice, and BMP2 has been shown to be a key regulator of implantation. In this study, we investigated the roles of the BMP type 1 receptor, activin-like kinase 2 (ALK2), during mouse pregnancy by producing mice carrying a conditional ablation of Alk2 in the uterus (Alk2 cKO mice). In the absence of ALK2, embryos demonstrate delayed invasion into the uterine epithelium and stroma, and upon implantation, stromal cells fail to undergo uterine decidualization, resulting in sterility. Mechanistically, microarray analysis revealed that CCAAT/enhancer-binding protein ß (Cebpb) expression is suppressed during decidualization in Alk2 cKO females. These findings and the similar phenotypes of Cebpb cKO and Alk2 cKO mice lead to the hypothesis that BMPs act upstream of CEBPB in the stroma to regulate decidualization. To test this hypothesis, we knocked down ALK2 in human uterine stromal cells (hESC) and discovered that ablation of ALK2 alters hESC decidualization and suppresses CEBPB mRNA and protein levels. Chromatin immunoprecipitation (ChIP) analysis of decidualizing hESC confirmed that BMP signaling proteins, SMAD1/5, directly regulate expression of CEBPB by binding a distinct regulatory sequence in the 3' UTR of this gene; CEBPB, in turn, regulates the expression of progesterone receptor (PGR). Our work clarifies the conserved mechanisms through which BMPs regulate peri-implantation in rodents and primates and, for the first time, uncovers a linear pathway of BMP signaling through ALK2 to regulate CEBPB and, subsequently, PGR during decidualization.


Assuntos
Receptores de Ativinas Tipo I/genética , Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular/genética , Implantação do Embrião/genética , Útero/metabolismo , Receptores de Ativinas Tipo I/metabolismo , Ativinas/metabolismo , Animais , Proteína Morfogenética Óssea 2/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proliferação de Células , Implantação do Embrião/fisiologia , Feminino , Humanos , Camundongos , Gravidez , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Transdução de Sinais/genética , Células Estromais/metabolismo , Útero/embriologia
7.
PLoS One ; 8(11): e78260, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24223784

RESUMO

This study investigated the development of rat oocytes in vitro and in vivo following intracytoplasmic injection of heads from spermatozoa heat-dried at 50°C for 8 h and stored at 4°C in different gas phases. Sperm membrane and chromosome are damaged by the process of heat-drying. Oocyte activation and cleavage of oocytes were worse in oocytes injected with spermatozoa heat-dried and stored for 1 week than unheated, fresh spermatozoa, but in heat-dried spermatozoa, there were no differences in these abilities of oocytes between the samples stored in nitrogen gas and in air. The oocytes injected with heat-dried spermatozoa stored for 1 week could develop to the morula and blastocyst stages without difference between the samples stored in nitrogen gas and in air after artificial stimulation. Cleavage of oocytes and development of cleaved embryos were higher when heat-dried spermatozoa were stored for 3 and 6 months in nitrogen gas than in air. However, the ability of injected oocytes to develop to the morula and blastocyst stages was not inhibited even when heat-dried spermatozoa stored in both atmosphere conditions for as long as 6 months were used. When 2-cell embryos derived from oocytes injected with heads from spermatozoa heat-dried and stored for 1 week and 1 month were transferred, each 1 of 4 recipients was conceived, and the conceived recipients delivered 1 live young each. These results demonstrate that rat oocytes can be fertilized with heat-dried spermatozoa and that the fertilized oocytes can develop to term.


Assuntos
Blastocisto/fisiologia , Implantação do Embrião , Mórula/fisiologia , Oócitos/fisiologia , Cabeça do Espermatozoide/fisiologia , Ar , Animais , Blastocisto/citologia , Dessecação , Feminino , Liofilização , Temperatura Alta , Masculino , Mórula/citologia , Nitrogênio , Oócitos/citologia , Ratos , Ratos Wistar , Preservação do Sêmen , Injeções de Esperma Intracitoplásmicas
8.
PLoS One ; 8(9): e76114, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086699

RESUMO

Factor in the germline alpha (FIGLA) is an oocyte-specific basic helix-loop-helix transcription factor essential for primordial follicle formation and expression of many genes required for folliculogenesis, fertilization and early embryonic survival. Here we report the characterization of bovine FIGLA gene and its regulation during early embryogenesis. Bovine FIGLA mRNA expression is restricted to gonads and is detected in fetal ovaries harvested as early as 90 days of gestation. FIGLA mRNA and protein are abundant in germinal vesicle and metaphase II stage oocytes, as well as in embryos from pronuclear to eight-cell stage but barely detectable at morula and blastocyst stages, suggesting that FIGLA might be a maternal effect gene. Recent studies in zebrafish and mice have highlighted the importance of non-coding small RNAs (microRNAs) as key regulatory molecules targeting maternal mRNAs for degradation during embryonic development. We hypothesized that FIGLA, as a maternal transcript, is regulated by microRNAs during early embryogenesis. Computational predictions identified a potential microRNA recognition element (MRE) for miR-212 in the 3' UTR of the bovine FIGLA mRNA. Bovine miR-212 is expressed in oocytes and tends to increase in four-cell and eight-cell stage embryos followed by a decline at morula and blastocyst stages. Transient transfection and reporter assays revealed that miR-212 represses the expression of FIGLA in a MRE dependent manner. In addition, ectopic expression of miR-212 mimic in bovine early embryos dramatically reduced the expression of FIGLA protein. Collectively, our results demonstrate that FIGLA is temporally regulated during bovine early embryogenesis and miR-212 is an important negative regulator of FIGLA during the maternal to zygotic transition in bovine embryos.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Bovinos/embriologia , Regulação da Expressão Gênica no Desenvolvimento/genética , MicroRNAs/metabolismo , Oócitos/metabolismo , RNA Mensageiro/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Northern Blotting , DNA Complementar/genética , Gônadas/metabolismo , Luciferases , MicroRNAs/genética , Plasmídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Reprod Biol Endocrinol ; 11: 86, 2013 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-24007267

RESUMO

BACKGROUND: Oocyte-specific genes play critical roles in oogenesis, folliculogenesis and early embryonic development. The objectives of this study were to characterize the expression of a novel oocyte-specific gene encoding an F-box protein during ovarian development in rainbow trout, and identify its potential interacting partners in rainbow trout oocytes. METHODS: Through analysis of expressed sequence tags (ESTs) from a rainbow trout oocyte cDNA library, a novel transcript represented by ESTs only from the oocyte library was identified. The complete cDNA sequence for the novel gene (named fbxoo) was obtained by assembling sequences from an EST clone and a 5'RACE product. The expression and localization of fbxoo mRNA and protein in ovaries of different developmental stages were analyzed by quantitative real time PCR, immunoblotting, in situ hybridization and immunohistochemistry. Identification of Fbxoo binding proteins was performed by yeast two-hybrid screening. RESULTS: fbxoo mRNA is specifically expressed in mature oocytes as revealed by tissue distribution analysis. The fbxoo cDNA sequence is 1,996 bp in length containing an open reading frame, which encodes a predicted protein of 514 amino acids. The novel protein sequence does not match any known protein sequences in the NCBI database. However, a search of the Pfam protein database revealed that the protein contains an F-box motif at the N-terminus, indicating that Fbxoo is a new member of the F-box protein family. The expression of fbxoo mRNA and protein is high in ovaries at early pre-vitellogenesis stage, and both fbxoo mRNA and protein are predominantly expressed in early pre-vitellogenic oocytes. Several proteins including tissue inhibitor of metalloproteinase 2 (Timp2) were identified as potential Fbxoo protein binding partners. CONCLUSIONS: Results suggest that the novel oocyte-specific F-box protein may play an important role in early oocyte development by regulating other critical proteins involved in oogenesis in rainbow trout.


Assuntos
Proteínas F-Box/metabolismo , Proteínas de Peixes/metabolismo , Oncorhynchus mykiss/genética , Oócitos/metabolismo , Animais , Clonagem Molecular , DNA Complementar/química , Etiquetas de Sequências Expressas , Proteínas F-Box/análise , Proteínas F-Box/química , Proteínas F-Box/genética , Proteínas de Peixes/análise , Proteínas de Peixes/química , Proteínas de Peixes/genética , Dados de Sequência Molecular , RNA Mensageiro/análise , Alinhamento de Sequência , Análise de Sequência de DNA
10.
Biol Reprod ; 86(1): 1-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21900682

RESUMO

Bidirectional signaling between oocytes and granulosa cells is required for normal folliculogenesis. Oocyte-secreted members of the transforming growth factor beta (TGFB) family, growth differentiation factor 9 (GDF9), and bone morphogenetic protein 15 (BMP15) are well-known mediators of granulosa cell function. Deletion in granulosa cells of Smad4, the common SMAD mediating all canonical TGFB-related protein signals, results in infertility. Reciprocal signaling by granulosa cell-expressed TGFB family ligands, such as activin, to the oocyte during follicle development has been proposed but not tested in vivo using conditional knockout mice. Therefore, we generated two oocyte-specific conditional knockout models for the common SMAD, Smad4, using cre recombinase expression from either the zona pellucida 3 (Zp3) or Gdf9 promoter. Cre expression from the Gdf9 promoter occurs at a slightly earlier time point in follicle development than from Zp3. Deletion of Smad4 using Zp3cre had no effect on fertility, while deletion of Smad4 with Gdf9icre resulted in a slight, but significant, reduction in litter size. These mouse models suggest a novel, although minor, role for Smad4 in the oocyte restricted to the primordial follicle stage.


Assuntos
Fertilidade/genética , Oócitos/metabolismo , Proteína Smad4/metabolismo , Animais , Proteínas do Ovo/genética , Proteínas do Ovo/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Células da Granulosa/metabolismo , Fator 9 de Diferenciação de Crescimento/genética , Fator 9 de Diferenciação de Crescimento/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Ovário/anatomia & histologia , Ovário/fisiologia , Ovulação/fisiologia , Regiões Promotoras Genéticas , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteína Smad4/genética , Glicoproteínas da Zona Pelúcida
11.
Biol Reprod ; 85(6): 1175-82, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21832168

RESUMO

The transforming growth factor beta (TGFB) protein family is renowned for its diverse roles in developmental biology including reproduction. Gremlin is a member of the differential screening-selected gene aberrative in neuroblastoma (DAN)/cerberus family of bone morphogenetic protein (BMP) antagonists. Recent studies on gremlin focus on its involvement in embryonic skeletal, lung, and kidney development. To define the role of gremlin (Grem1) in female reproduction, we analyzed postnatal folliculogenesis using global and conditional knockout (cKO) mice for gremlin. Grem1(-/-) mice die within 48 h after birth, and ovaries collected from neonatal Grem1(-/-) mice demonstrated reduced oocyte numbers and delayed primordial follicle development. Transplanting Grem1(-/-) neonatal ovaries showed that folliculogenesis proceeded to large antral follicle stage, but Grem1(-/-) ovaries contained corpora lutea-like structures not found in control-transplanted ovaries. However, Grem1 cKO mice had comparable fertility to control mice. These data suggest that gremlin plays a previously uncharacterized role in the regulation of oocyte numbers and the timing of primordial follicle development, but either it is not required for later folliculogenesis or its loss is possibly compensated by other BMP antagonists.


Assuntos
Fertilidade , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Folículo Ovariano/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Citocinas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovário/citologia , Ovário/metabolismo , Proteínas/metabolismo
12.
BMC Dev Biol ; 11: 25, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21548929

RESUMO

BACKGROUND: Oocyte-derived maternal RNAs drive early embryogenesis when the newly formed embryo is transcriptionally inactive. Recent studies in zebrafish have identified the role of microRNAs during the maternal-to-embryonic transition (MET). MicroRNAs are short RNAs that bind to the 3' UTR of target mRNAs to repress their translation and accelerate their decay. Newborn ovary homeobox gene (NOBOX) is a transcription factor that is preferentially expressed in oocytes and essential for folliculogenesis in mice. NOBOX knockout mice are infertile and lack of NOBOX disrupts expression of many germ-cell specific genes and microRNAs. We recently reported the cloning and expression of bovine NOBOX during early embryonic development and our gene knockdown studies indicate that NOBOX is a maternal effect gene essential for early embryonic development. As NOBOX is a maternal transcript critical for development and NOBOX is depleted during early embryogenesis, we hypothesized that NOBOX is targeted by microRNAs for silencing and/or degradation. RESULTS: Using an algorithm "MicroInspector", a potential microRNA recognition element (MRE) for miR-196a was identified in the 3' UTR of the bovine NOBOX mRNA. Expression analysis of miR-196a in bovine oocytes and during early embryonic development indicated that it is expressed both in oocytes and embryos and tends to increase at the four-cell and eight-cell stages. Ectopic expression of NOBOX and miR-196a in HeLa cells inhibited the expression of NOBOX protein compared to the control cells without miR-196a. Similarly, the activity of a luciferase construct containing the entire 3' UTR of bovine NOBOX was suppressed, and the regulation was abolished by mutations in the miR-196a binding site indicating that the predicted MRE is critical for the direct and specific binding of miR-196a to the NOBOX mRNA. Furthermore, ectopic expression of miR-196a mimic in bovine early embryos significantly reduced the NOBOX expression at the both mRNA and protein levels. CONCLUSION: Collectively, our results demonstrate that miR-196a is a bona fide negative regulator of NOBOX during bovine early embryogenesis.


Assuntos
Desenvolvimento Embrionário/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox , Proteínas de Homeodomínio/metabolismo , MicroRNAs/metabolismo , Ovário/embriologia , Fatores de Transcrição/metabolismo , Regiões 3' não Traduzidas , Algoritmos , Animais , Animais Recém-Nascidos , Sequência de Bases , Bovinos , Feminino , Proteínas de Homeodomínio/genética , Humanos , Camundongos , MicroRNAs/genética , Dados de Sequência Molecular , Oócitos/fisiologia , Ovário/citologia , Fatores de Transcrição/genética
13.
Reprod Biol Endocrinol ; 9: 40, 2011 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21447182

RESUMO

BACKGROUND: Nucleoplasmin 2 (NPM2) is an oocyte-specific nuclear protein essential for nuclear and nucleolar organization and early embryonic development. The aims of this study were to clone the bovine NPM2 gene, determine its temporal expression during oocyte development and early embryogenesis, and evaluate the potential role of miRNA-181a in regulation of its expression. METHODS: A 329 bp cDNA fragment was amplified from bovine fetal ovary using primers designed based on the conserved regions of the human and mouse NPM2 cDNA sequences. RACE experiments were performed to obtain the 5' and 3' ends of the bovine NPM2 cDNA. Real time PCR and Western blot analysis were used to examine the expression of bovine NPM2 in oocytes and early embryos. Co-expression of bovine NPM2 and miRNA-181a in Hela cells was performed to determine if expression of bovine NPM2 is regulated by miRNA-181a. RESULTS: The bovine NPM2 cDNA is 851 bp in length encoding a protein of 200 amino acids. The protein contains the conserved bipartite nuclear localization sequence and shows 53% and 62% identity with mouse and human NPM2, respectively. Expression of bovine NPM2 mRNA is restricted to ovaries. NPM2 mRNA is abundant in GV and MII stage oocytes, decreases in early cleavage stage embryos, and barely detectable in morula and blastocyst stage embryos. Similarly, expression of NPM2 protein is high in oocytes and early embryos but extremely low in blastocysts. The abundance of NPM2 mRNA is significantly lower in oocytes isolated from persistent versus growing dominant follicles (P < 0.05). A miR-181a binding site in the 3'UTR of the NPM2 transcript was identified. Transfection experiments showed that bovine NPM2 protein expression is reduced in Hela cells expressing miR-181a compared to control cells without miR-181a, indicating that translation of NPM2 is repressed by miR-181a. CONCLUSIONS: Our data suggest that expression of bovine NPM2 is temporally regulated during early embryogenesis and miR-181a may play a role in its regulation.


Assuntos
MicroRNAs/fisiologia , Nucleoplasminas/biossíntese , Oócitos/fisiologia , Sequência de Aminoácidos , Animais , Bovinos , Clonagem Molecular , Feminino , Dados de Sequência Molecular , Oogênese/fisiologia , Alinhamento de Sequência , Distribuição Tecidual
14.
Endocrinology ; 152(3): 1013-23, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21193554

RESUMO

Newborn ovary homeobox (NOBOX) is an oocyte-specific transcription factor essential for folliculogenesis and expression of many germ cell-specific genes in mice. Here we report the characterization of the bovine NOBOX gene and its role in early embryogenesis. The cloned cDNA for bovine NOBOX contains an open reading frame encoding a protein of 500 amino acids with a conserved homeodomain. mRNA for NOBOX is preferentially expressed in ovaries and undetectable by RT-PCR in somatic tissues examined. NOBOX protein is present in oocytes throughout folliculogenesis. NOBOX is expressed in a stage-specific manner during oocyte maturation and early embryonic development and of maternal origin. Knockdown of NOBOX in early embryos using small interfering RNA demonstrated that NOBOX is required for embryonic development to the blastocyst stage. Depletion of NOBOX in early embryos caused significant down-regulation of genes associated with transcriptional regulation, signal transduction, and cell cycle regulation during embryonic genome activation. In addition, NOBOX depletion in early embryos reduced expression of pluripotency genes (POU5F1/OCT4 and NANOG) and number of inner cell mass cells in embryos that reached the blastocyst stage. This study demonstrates that NOBOX is an essential maternal-derived transcription factor during bovine early embryogenesis, which functions in regulation of embryonic genome activation, pluripotency gene expression, and blastocyst cell allocation.


Assuntos
Bovinos/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Oócitos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Clonagem Molecular , Desenvolvimento Embrionário/fisiologia , Feminino , Genoma , Proteínas de Homeodomínio/genética , Ovário/metabolismo , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fatores de Tempo , Fatores de Transcrição/genética
15.
Anim Reprod Sci ; 120(1-4): 16-22, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20347535

RESUMO

Ovarian folliculogenesis and early embryogenesis are complex processes, which require tightly regulated expression and interaction of a multitude of genes. Small endogenous RNA molecules, termed microRNAs (miRNAs), are involved in the regulation of gene expression during folliculogenesis and early embryonic development. To identify miRNAs in bovine oocytes/ovaries, a bovine fetal ovary miRNA library was constructed. Sequence analysis of random clones from the library identified 679 miRNA sequences, which represent 58 distinct bovine miRNAs. Of these distinct miRNAs, 42 are known bovine miRNAs present in the miRBase database and the remaining 16 miRNAs include 15 new bovine miRNAs that are homologous to miRNAs identified in other species, and one novel miRNA, which does not match any miRNAs in the database. The precursor sequences for 14 of the new 15 miRNAs as well as the novel miRNA were identified from the bovine genome database and their hairpin structures were predicted. Expression analysis of the 58 miRNAs in fetal ovaries in comparison to somatic tissue pools identified 8 miRNAs predominantly expressed in fetal ovaries. Further analysis of the eight miRNAs in germinal vesicle (GV) stage oocytes identified two miRNAs (bta-mir424 and bta-mir-10b), that are highly abundant in GV oocytes. Both miRNAs show similar expression patterns during oocyte maturation and preimplantation development of bovine embryos, being abundant in GV and MII stage oocytes, as well as in early stage embryos (until 16-cell stage). The amount of the novel miRNA is relatively small in oocytes and early cleavage embryos but greater in blastocysts, suggesting a role of this miRNA in blastocyst cell differentiation.


Assuntos
Bovinos , Feto/metabolismo , MicroRNAs/genética , MicroRNAs/isolamento & purificação , Ovário/metabolismo , Animais , Bovinos/embriologia , Bovinos/genética , Clonagem Molecular , Embrião de Mamíferos , Feminino , Feto/química , Perfilação da Expressão Gênica , Biblioteca Gênica , Ovário/química , Ovário/embriologia , Gravidez , RNA/genética , RNA/isolamento & purificação , RNA Mitocondrial
16.
Biol Reprod ; 81(2): 333-42, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19420384

RESUMO

Nuclear proteins such as transcription and chromatin remodeling factors are required for initiation of transcription in early embryos before embryonic genome activation. The nuclear transport of these proteins is mediated by transport factors such as importins. Through analysis of expressed sequence tags from a bovine oocyte cDNA library, we identified a new member of the importin alpha family (named importin alpha8). The cloned cDNA for bovine importin alpha8 (KPNA7) is 1817 base pair in length, encoding a protein of 522 amino acids that contains a conserved importin beta-binding domain and seven armadillo motifs. The RT-PCR analysis revealed that KPNA7 mRNA is specifically expressed in ovaries and mature oocytes. Real-time PCR demonstrated that KPNA7 expression in germinal vesicle (GV) oocytes is 33 to 2396 times higher than that of other importin alpha genes and that KPNA7 mRNA is abundant in GV and metaphase II oocytes, as well as in early-stage embryos collected before embryonic genome activation, but is barely detectable in morula- and blastocyst-stage embryos. Similarly, expression of KPNA7 protein is very high in oocytes and early embryos but is low in blastocysts. A glutathione S-transferase pull-down assay revealed that KPNA7 has a strong binding affinity for the nuclear protein nucleoplasmin 2 relative to that of other importin alphas. RNA interference experiments demonstrated that knockdown of KPNA7 in early embryos results in a decreased proportion of embryos developing to the blastocyst stage. These results suggest that KPNA7 may have an important role in the transport of essential nuclear proteins required for early embryogenesis.


Assuntos
Bovinos/embriologia , Embrião de Mamíferos/metabolismo , Ovário/metabolismo , alfa Carioferinas/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Blastocisto/metabolismo , Bovinos/metabolismo , Desenvolvimento Embrionário , Etiquetas de Sequências Expressas , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Biblioteca Gênica , Células da Granulosa/metabolismo , Dados de Sequência Molecular , Nucleoplasminas/metabolismo , Oócitos/citologia , Oócitos/metabolismo , Especificidade de Órgãos , Ovário/embriologia , Filogenia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas , Interferência de RNA , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Células Tecais/metabolismo , alfa Carioferinas/química , alfa Carioferinas/genética , alfa Carioferinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...