Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Reprod Med Biol ; 17(2): 107-114, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29692667

RESUMO

BACKGROUND: The germ cell lineage transmits genetic and epigenetic information to the next generation. Primordial germ cells (PGCs), the early embryonic precursors of sperm or eggs, have been studied extensively. Recently, in vitro models of PGC induction have been established in the mouse. Many attempts are reported to enhance our understanding of PGC development in other mammals, including human. METHODS: Here, original and review articles that have been published on PubMed are reviewed in order to give an overview of the literature that is focused on PGC development, including the specification of in vivo and in vitro in mice, human, porcine, and bovine. RESULTS: Mammalian PGC development, in vivo and in vitro, have been studied primarily by using the mouse model as a template to study PGC specification in other mammals, including human, porcine, and bovine. CONCLUSION: The growing body of published works reveals similarities, as well as differences, in PGC establishment in and between mouse and human.

2.
Dev Biol ; 385(2): 155-9, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24269765

RESUMO

Primordial germ cells (PGCs) are the founder cells of the germline. Via gametogenesis and fertilisation this lineage generates a new embryo in the next generation. PGCs are also the cell of origin of multilineage teratocarcinomas. In vitro, mouse PGCs can give rise to embryonic germ (EG) cells - pluripotent stem cells that can contribute to primary chimaeras when introduced into pre-implantation embryos. Thus, PGCs can give rise to pluripotent cells in the course of the developmental cycle, during teratocarcinogenesis and by in vitro culture. However, there is no evidence that PGCs can differentiate directly into somatic cell types. Furthermore, it is generally assumed that PGCs do not contribute to chimaeras following injection into the early mouse embryo. However, these data have never been formally published. Here, we present the primary data from the original PGC-injection experiments performed 40 years ago, alongside results from more recent studies in three separate laboratories. These results have informed and influenced current models of the relationship between pluripotency and the germline cycle. Current technologies allow further experiments to confirm and expand upon these findings and allow definitive conclusions as to the developmental potency of PGCs.


Assuntos
Embrião de Mamíferos/citologia , Células Germinativas/citologia , Animais , Linhagem da Célula , Feminino , Camundongos , Gravidez
4.
6.
Differentiation ; 78(2-3): 116-23, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19683852

RESUMO

Embryonic stem (ES) cells, derived from pre-implantation embryo, embryonic germ (EG) cells, derived from embryonic precursors of gametes, primordial germ cells (PGCs), can differentiate into any cell type in the body. Moreover, ES cells have the capacity to differentiate into PGCs in vitro. In the present study we have shown the differentiation capacity of six EG cell lines to form PGCs in vitro, in comparison to ES cells. Cell lines were differentiated via embryoid body (EB) formation using the co-expression of mouse vasa homolog (Mvh) and Oct-4 to identify newly formed PGCs in vitro. We found an increase of PGC numbers in almost all analysed cell lines in 5-day-old EBs, thus suggesting that EG and ES cells have similar efficiency to generate PGCs. The addition of retinoic acid confirmed that the cultures had attained a PGC-like identity and continued to proliferate. Furthermore we have shown that the expression pattern of Prmt5 and H3K27me3 in newly formed PGCs is similar to that observed in embryonic day E11.5 PGCs in vivo. By co-culturing EBs with Chinese hamster ovary (CHO) cells some of the PGCs entered into meiosis, as judged by Scp3 expression. The derivation of germ cells from pluripotent stem cells in vitro could provide an invaluable model system to study both the genetic and epigenetic programming of germ cell development in vivo.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Animais , Células CHO , Proteínas de Ciclo Celular , Proliferação de Células , Células Cultivadas , Cricetinae , Cricetulus , Proteínas de Ligação a DNA , Antígenos CD15/metabolismo , Meiose , Camundongos , Proteínas Nucleares/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases
7.
PLoS One ; 3(10): e3531, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18953407

RESUMO

BACKGROUND: Specification of primordial germ cells (PGCs) results in the conversion of pluripotent epiblast cells into monopotent germ cell lineage. Blimp1/Prmt5 complex plays a critical role in the specification and maintenance of the early germ cell lineage. However, PGCs can be induced to dedifferentiate back to a pluripotent state as embryonic germ (EG) cells when exposed to exogenous signaling molecules, FGF-2, LIF and SCF. METHODOLOGY AND PRINCIPAL FINDINGS: Here we show that Trichostatin A (TSA), an inhibitor of histone deacetylases, is a highly potent agent that can replace FGF-2 to induce dedifferentiation of PGCs into EG cells. A key early event during dedifferentiation of PGCs in response to FGF-2 or TSA is the down-regulation of Blimp1, which reverses and apparently relieves the cell fate restriction imposed by it. Notably, the targets of Blimp1, which include c-Myc and Klf-4, which represent two of the key factors known to promote reprogramming of somatic cells to pluripotent state, are up-regulated. We also found early activation of the LIF/Stat-3 signaling pathway with the translocation of Stat-3 into the nucleus. By contrast, while Prmt5 is retained in EG cells, it translocates from the nucleus to the cytoplasm where it probably has an independent role in regulating pluripotency. CONCLUSIONS/SIGNIFICANCE: We propose that dedifferentiation of PGCs into EG cells may provide significant mechanistic insights on early events associated with reprogramming of committed cells to a pluripotent state.


Assuntos
Desdiferenciação Celular/fisiologia , Células Germinativas/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Desdiferenciação Celular/genética , Linhagem da Célula/genética , Linhagem da Célula/fisiologia , Células Cultivadas , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes myc , Células Germinativas/metabolismo , Fator Inibidor de Leucemia/metabolismo , Masculino , Camundongos , Modelos Biológicos , Células-Tronco Pluripotentes/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo , Gravidez , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Curr Protoc Stem Cell Biol ; Chapter 1: Unit1A.3, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18770625

RESUMO

In this unit we describe the derivation of pluripotent embryonic germ (EG) cells from mouse primordial germ cells (PGCs) isolated from both 8.5- and 11.5-days post-coitum (dpc) embryos. Once EG cells are derived we explain how to propagate and characterize the cell lines. We introduce readers to PGCs and explain differences between PGCs and their in vitro derivatives EG cells. Finally, we also compare mouse EG cells with ES cells. This unit will be of great interest to anyone interested in PGCs or studying the behavior of cultured PGCs or the derivation of new EG cell lines.


Assuntos
Técnicas de Cultura de Células/métodos , Reprogramação Celular , Embrião de Mamíferos/citologia , Células Germinativas/citologia , Células-Tronco/citologia , Fosfatase Alcalina/metabolismo , Animais , Linhagem Celular , Separação Celular , Criopreservação , Imunofluorescência , Antígenos CD15/metabolismo , Magnetismo , Camundongos , Microesferas , Mitose , Fator 3 de Transcrição de Octâmero/metabolismo , Reação em Cadeia da Polimerase , Análise para Determinação do Sexo
10.
Curr Top Dev Biol ; 83: 185-212, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19118667

RESUMO

In mammals, germ cells are induced from a population of cells at the base of the allantois. This regulative mechanism of germ line induction depends on Bmp signals and a combination of epigenetic changes that silence somatic differentiation genes and activate pluripotency genes. RNA binding proteins are a conserved feature of germ cell development in mammals, and play critical roles in the establishment and maintenance of pluripotency. After their specification, germ cells move through the gut to the gonads under the influence of migratory and attractive cues. In the gonad, germ cells initiate sex-specific differentiation. Germ cells that arrive in the ovary enter meiosis, whereas germ cells that arrive in the testis undergo mitotic arrest. Entry into meiosis is controlled by retinoic acid signals that are blocked in the testis. The signals regulating mitotic arrest in the testis are still not completely understood, but likely involve RNA-binding proteins. Epigenetic reprograming occurs during specification, migratory stages, and sex-specific stages, when maternal and paternal imprints are established. The facility of transitions between germ cells and stem cells suggests a close relationship among their genomic programs.


Assuntos
Diferenciação Celular , Células Germinativas/citologia , Células Germinativas/metabolismo , Animais , Ciclo Celular , Linhagem da Célula , Movimento Celular , Epigênese Genética , Feminino , Gônadas/citologia , Gônadas/embriologia , Gônadas/metabolismo , Masculino , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Caracteres Sexuais , Desenvolvimento Sexual
11.
Dev Biol ; 313(2): 674-81, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18062950

RESUMO

Pluripotent stem cells, termed embryonic germ (EG) cells, have been generated from both human and mouse primordial germ cells (PGCs). Like embryonic stem (ES) cells, EG cells have the potential to differentiate into all germ layer derivatives and may also be important for any future clinical applications. The development of PGCs in vivo is accompanied by major epigenetic changes including DNA demethylation and imprint erasure. We have investigated the DNA methylation pattern of several imprinted genes and repetitive elements in mouse EG cell lines before and after differentiation. Analysed cell lines were derived soon after PGC specification, "early", in comparison with EG cells derived after PGC colonisation of the genital ridge, "late" and embryonic stem (ES) cell lines, derived from the inner cell mass (ICM). Early EG cell lines showed strikingly heterogeneous DNA methylation patterns, in contrast to the uniformity of methylation pattern seen in somatic cells (control), late EG cell and ES cell lines. We also observed that all analysed XX cell lines exhibited less methylation than XY. We suggest that this heterogeneity may reflect the changes in DNA methylation taking place in the germ cell lineage soon after specification.


Assuntos
Metilação de DNA , Heterogeneidade Genética , Impressão Genômica , Células Germinativas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Cruzamentos Genéticos , Embrião de Mamíferos , Feminino , Cariotipagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Tempo , Transgenes
12.
Proc Natl Acad Sci U S A ; 103(30): 11184-8, 2006 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-16847261

RESUMO

Germ cells in XY male mice establish site-specific methylation on imprinted genes during spermatogenesis, whereas germ cells in XX females establish their imprints in growing oocytes. We showed previously that in vitro, sex-specific methylation patterns of pluripotent stem cell lines derived from germ cells were influenced more by the sex chromosome constitution of the cells themselves than by the gender of the embryo from which they had been derived. To see whether the same situation would prevail in vivo, we have now determined the methylation status of H19 expressed from the maternal allele, and the expression and methylation status of a paternally expressed gene Peg3, in germ cells from sex-reversed and control embryos. For these imprinted genes, we conclude that the female imprint is a response of the germ cells to undergoing oogenesis, rather than to their XX chromosome constitution. Similarly, both our XY and our sex-reversed XX male germ cells clearly showed a male rather than a female pattern of DNA methylation; here, however, the sex chromosome constitution had a significant effect, with XX male germ cells less methylated than the XY controls.


Assuntos
Impressão Genômica , Células Germinativas/citologia , Cromossomos Sexuais/genética , Animais , Sequência de Bases , Metilação de DNA , Feminino , Masculino , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , RNA Longo não Codificante , RNA não Traduzido/genética , Sulfitos/farmacologia , Transgenes
13.
Stem Cells ; 24(6): 1441-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16769760

RESUMO

The germ cell lineage is a specified cell population that passes through a series of differentiation steps before giving rise, eventually, to either eggs or sperm. We have investigated the manner in which primordial germ cells (PGCs) are reprogrammed in vitro to form pluripotent stem cells in response to exogenous fibroblast growth factor-2 (FGF-2). The response is dependent on time of exposure and concentration of FGF-2. PGCs isolated in culture show a motile phenotype and lose any expression of a characteristic germ cell marker, mouse vasa homolog. Subsequently, some but not all of the cells show further changes of phenotype, accompanied by changes in expression of endogenous FGF-2 and up-regulation of its receptor, fibroblast growth factor receptor-3, in the nucleus. We propose that it is from this reprogrammed component of the now heterogeneous PGC population that pluripotent stem cells arise.


Assuntos
Fator 2 de Crescimento de Fibroblastos/farmacologia , Células Germinativas/citologia , Células Germinativas/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células Germinativas/metabolismo , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Células-Tronco Pluripotentes/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais
14.
Dev Biol ; 268(1): 105-10, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15031108

RESUMO

Sex-specific differences are apparent in the methylation patterns of H19 and Igf2 imprinted genes in embryonic germ cells (EGCs) derived from 11.5 or 12.5 days post coitum (dpc) primordial germ cells (PGCs). Here we studied whether these differences are associated either with the sex chromosome constitution of the EGCs or with the sex of the genital ridge (testis versus ovary) from which the PGCs were isolated. For this purpose we derived pluripotent EGC lines from sex-reversed embryos, either XY embryos deleted for Sry (XY(Tdym1)) or XX embryos carrying an Sry transgene. Southern blotting of the EGC DNA was used to analyze the differentially methylated regions of Igf2 and H19. The analysis revealed that both genes were more methylated in EGCs with an XY sex chromosome constitution than in those with an XX sex chromosome constitution, irrespective of the phenotypic sex of the genital ridge from which the EGCs had been derived. We conclude that the sex-specific methylation is intrinsic and cell-autonomous, and is not due to any influence of the genital ridge somatic cells upon the PGCs.


Assuntos
Impressão Genômica , Células Germinativas/citologia , Fatores Sexuais , Animais , Southern Blotting , Feminino , Fator de Crescimento Insulin-Like II/genética , Masculino , Reação em Cadeia da Polimerase , RNA Longo não Codificante , RNA não Traduzido/genética
15.
Differentiation ; 71(2): 135-41, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12641567

RESUMO

Embryonic germ cells (EGCs) derived from mouse primordial germ cells (PGCs) are known both to colonize all cell lineages of the fetus and to make tumors in vivo. When aggregated with eight-cell embryos, EGCs from a new EGC line expressing green fluorescent protein (GFP) were found to contribute preferentially to the epiblast but unexpectedly were also capable of colonizing primary endoderm. When injected under the kidney capsule, EGCs derived from 12.5 days post coitum (dpc) PGCs formed differentiated tumors. The ability of EGCs to differentiate in an organ culture system depends upon their partners in cell culture. When EGCs, marked with a LacZ transgene, were mixed with disaggregated and reaggregated mouse fetal lung in an organ culture system, they remained undifferentiated. In urogenital ridge reaggregates on the other hand, some EGCs were capable of differentiating to form small epithelial cysts.


Assuntos
Células Germinativas/crescimento & desenvolvimento , Animais , Quimera/fisiologia , Embrião de Mamíferos/fisiologia , Genes Reporter/fisiologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...