Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Stem Cell ; 17(5): 557-68, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26412560

ABSTRACT

Trophoblast stem cells (TSCs) arise from the first cell fate decision in the developing embryo and generate extra-embryonic lineages, giving rise to the fetal portion of the placenta. Mouse embryonic and extra-embryonic lineages are strictly separated by a distinct epigenetic barrier, which is not fully overcome following expression of TSC-determining factors in embryonic stem cells. Here, we show that transient expression of Tfap2c, Gata3, Eomes, and Ets2 is sufficient to reprogram mouse embryonic fibroblasts and post-natal tail-tip-derived fibroblasts into induced TSCs (iTSCs) and surmount the epigenetic barrier separating somatic from extra-embryonic lineages. iTSCs share nearly identical morphological characteristics, gene expression profiles, and DNA methylation patterns with blastocyst-derived TSCs. Furthermore, iTSCs display transgene-independent self-renewal, differentiate along extra-embryonic lineages, and chimerize host placentas following blastocyst injection. These findings provide insights into the transcription factor networks governing TSC identity and opportunities for studying the epigenetic barriers underlying embryonic and extra-embryonic lineage segregation.


Subject(s)
Cell Transdifferentiation , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Trophoblasts/cytology , Animals , Cell Cycle , Cell Lineage , Cells, Cultured , Mice
2.
Biol Reprod ; 93(2): 31, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26063869

ABSTRACT

Tfap2c is required for placental development and trophoblast stem cell maintenance. Deletion of Tfap2c results in early embryonic loss because of failure in placental development. We evaluated the effect of reduced Tfap2c expression on fetal outcome and placental development. Sixty percent of the heterozygous mice were lost directly after birth. Labyrinthine differentiation was impaired, as indicated by enhanced proliferation and inclusions of cobblestone-shaped cell clusters characterized by expression of Tfap2c and glycogen stores. Moreover, expression of marker genes such as Cdx2, Eomes, Gata3, and Ascl2 are decreased in the spongiotrophoblast and indicate a lowered stem cell potential. On Day 18.5 postcoitum, the labyrinth layer of Tfap2c(+/-) placentas exhibited massive hemorrhages in the maternal blood spaces; these hemorrhages might have contributed to the significantly reduced number of live-born pups. These morphological alterations were accompanied by a shift toward sinusoidal trophoblast giant cells as the cell subpopulation lining the maternal sinusoids and toward reduction in expression of the prolactin gene family member Prl2c2, a finding characteristic of the spiral arteries lining trophoblast cells. The trophoblast stem cells heterozygous for Tfap2c exhibited a reduction in the expression level of stem cell markers and in their proliferation and differentiation capacity but did not exhibit changes in marker genes of the trophoblast giant cell lineage. Taken together, these findings indicate that a reduction in the gene dosage of placental Tfap2c leads to morphological changes in the labyrinth at midgestation and in the maternal blood spaces during late pregnancy.


Subject(s)
Placenta/pathology , Transcription Factor AP-2/genetics , Trophoblasts/physiology , Animals , Cell Differentiation , Cell Lineage , Female , Genetic Markers/genetics , Genotype , Giant Cells , Mice , Mice, Knockout , Mice, Transgenic , Pregnancy , Prolactin/genetics , Stem Cells
3.
Stem Cell Reports ; 2(2): 232-42, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24527396

ABSTRACT

Trophoblast stem cells (TSCs) are in vitro equivalents to the precursor cells of the placenta. TSCs are cultured in serum-rich medium with fibroblast growth factor 4, heparin, and embryonic-fibroblast-conditioned medium. Here, we developed a simple medium consisting of ten chemically defined ingredients for culture of TSCs on Matrigel or synthetic substrates, named TX medium. Gene expression and DNA methylation profiling demonstrated the faithful propagation of expression profiles and epigenomic characteristics of TSCs cultured in TX. Further, TX medium supported the de novo derivation of TSC lines. Finally, TSCs cultured in TX differentiate into all derivatives of the trophectodermal lineage in vitro, give rise to hemorrhagic lesions in nude mice, and chimerize the placenta, indicating that they retained all hallmarks of TSCs. TX media formulation no longer requires fetal bovine serum and conditioned medium, which facilitates and standardizes the culture of this extraembryonic lineage.


Subject(s)
Cell Culture Techniques , Cell Differentiation , Stem Cells/cytology , Trophoblasts/cytology , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Chimera , CpG Islands , Culture Media, Conditioned , Culture Media, Serum-Free , DNA Methylation , Embryo, Mammalian , Female , Fibroblasts/metabolism , Gene Expression , Gene Expression Profiling , Karyotype , Male , Mice , Stem Cells/drug effects , Stem Cells/metabolism , Transcriptome , Trophoblasts/metabolism
4.
Reprod Biomed Online ; 25(1): 12-20, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22560121

ABSTRACT

In recent years, knowledge regarding the genetic and epigenetic programmes governing specification, maintenance and differentiation of the extraembryonic lineage has advanced substantially. Establishment and analysis of mice deficient in genes implicated in trophoblast lineage and the option to generate and manipulate murine stem cell lines from the inner cell mass and the trophectoderm in vitro represent major advances. The activating enhancer binding protein 2 (AP2) family of transcription factors is expressed during mammalian development and in certain malignant diseases. This article summarizes the data regarding expression and function of murine Tcfap2 and human TFAP2 in extraembryonic development and differentiation. It also presents a model integrating Tcfap2c into the framework of trophoblast development and highlights the requirement of Tcfap2c to maintain trophoblast stem cells. With regard to human trophoblast cell-lineage restriction, the role of TFAP2C in lineage specification and maintenance is speculated upon. Furthermore, an overview of target genes of AP2 in mouse and human affecting placenta development and function is provided and the evidence suggesting that defects in regulating TFAP2 members might contribute to placental defects is discussed.


Subject(s)
Transcription Factor AP-2/physiology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Ectoderm/physiology , Female , Humans , Mice , Mice, Knockout , Pregnancy , Stem Cells/physiology , Trophoblasts/cytology , Trophoblasts/physiology
5.
PLoS One ; 6(7): e22034, 2011.
Article in English | MEDLINE | ID: mdl-21779369

ABSTRACT

BACKGROUND: The transcription factor Tcfap2c has been demonstrated to be essential for various processes during mammalian development. It has been found to be upregulated in various undifferentiated tumors and is implicated with poor prognosis. Tcfap2c is reported to impinge on cellular proliferation, differentiation and apoptosis. However, the physiological consequences of Tcfap2c-expression remain largely unknown. METHODOLOGY/PRINCIPAL FINDINGS: Therefore we established a gain of function model to analyze the role of Tcfap2c in development and disease. Induction of the transgene led to robust expression in all tissues (except brain and testis) and lead to rapid mortality within 3-7 days. In the liver cellular proliferation and apoptosis was detected. Accumulation of microvesicular lipid droplets and breakdown of major hepatic metabolism pathways resulted in steatosis. Serum analysis showed a dramatic increase of enzymes indicative for hepatic failure. After induction of Tcfap2c we identified a set of 447 common genes, which are deregulated in both liver and primary hepatocyte culture. Further analysis showed a prominent repression of the cytochrome p450 system, PPARA, Lipin1 and Lipin2. These data indicate that in the liver Tcfap2c represses pathways, which are responsible for fatty acid metabolism. In the intestine, Tcfap2c expression resulted in expansion of Sox9 positive and proliferative active epithelial progenitor cells resulting in dysplastic growth of mucosal crypt cells and loss of differentiated mucosa. CONCLUSIONS: The transgenic mice show that ectopic expression of Tcfap2c is not tolerated. Due to the phenotype observed, iTcfap2c-mice represent a model system to study liver failure. In intestine, Tcfap2c induced cellular hyperplasia and suppressed terminal differentiation indicating that Tcfap2c serves as a repressor of differentiation and inducer of proliferation. This might be achieved by the Tcfap2c mediated activation of Sox9 known to be expressed in intestinal and hepatic stem/progenitor cell populations.


Subject(s)
Intestinal Diseases/metabolism , Liver Failure/metabolism , Transcription Factor AP-2/metabolism , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Intestinal Diseases/etiology , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Liver Failure/etiology , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , PPAR alpha/metabolism , Phosphatidate Phosphatase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor AP-2/genetics
6.
Mol Cell Biol ; 31(8): 1748-56, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21300784

ABSTRACT

In mammals, the first cell fate decision is initialized by cell polarization at the 8- to 16-cell stage of the preimplantation embryo. At this stage, outside cells adopt a trophectoderm (TE) fate, whereas the inside cell population gives rise to the inner cell mass (ICM). Prior to implantation, transcriptional interaction networks and epigenetic modifications divide the extraembryonic and embryonic fate irrevocably. Here, we report that extraembryonic trophoblast stem cell (TSC) lines are converted to induced pluripotent stem cells (TSC-iPSCs) by overexpressing Oct4, Sox2, Klf4, and cMyc. Methylation studies and gene array analyses indicated that TSC-iPSCs had adopted a pluripotent potential. The rate of conversion was lower than those of somatic reprogramming experiments, probably due to the unique genetic network controlling extraembryonic lineage fixation. Both in vitro and in vivo, TSC-iPSCs differentiated into tissues representing all three embryonic germ layers, indicating that somatic cell fate could be induced. Finally, TSC-iPSCs chimerized the embryo proper and contributed to the germ line of mice, indicating that these cells had acquired full somatic differentiation potential. These results lead to a better understanding of the molecular processes that govern the first lineage decision in mammals.


Subject(s)
Cell Lineage , Pluripotent Stem Cells/cytology , Stem Cells/cytology , Trophoblasts/cytology , Animals , Cell Differentiation , Epigenesis, Genetic , Kruppel-Like Factor 4 , Mice , Mice, Inbred C57BL , Pluripotent Stem Cells/metabolism , Stem Cells/metabolism , Trophoblasts/metabolism
7.
Mol Cell Biol ; 30(13): 3310-20, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20404091

ABSTRACT

In mammals, cell lineage specification is established at the blastocyst stage. At this stage, transcription factor Cdx2 represses pluripotency genes, thus promoting extraembryonic trophoblast fate. Recently, transcription factor Gata3 was shown to act in a parallel pathway in promoting trophoblast cell fate, suggesting that there are more factors working in the trophoblast lineage. Here, we report that the transcription factor Tcfap2c is expressed at a high level in the trophectoderm and is able to induce trophoblast fate in embryonic stem cells. Trophoblast fate induced by Tcfap2c does not require Cdx2 and vice versa, suggesting that the molecules act in alternative pathways. However, both Tcfap2c and Cdx2 are required for the upregulation of Elf5, a marker of trophoblast stem cell maintenance, suggesting that both factors are required for stable trophoblast induction. Tcfap2c-induced trophoblast-like cells are stable in long-term culture, indicating that they are capable of self-renewal. Tcfap2c-controlled trophoblast maintenance involves the induction of Cdx2 and the repression of the pluripotency factor Nanog. Tcfap2c-induced trophoblast-like cells differentiate to trophoblast derivatives in vitro and contribute to the trophectoderm in blastocysts in vivo. Taken together, these observations suggest that Tcfap2c and Cdx2 cooperate to override the pluripotency program and establish the extraembryonic trophoblast maintenance program in murine embryos.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Homeodomain Proteins/metabolism , Transcription Factor AP-2/metabolism , Transcription Factors/metabolism , Trophoblasts , Animals , Biomarkers/metabolism , CDX2 Transcription Factor , Cell Lineage , Cells, Cultured , Embryonic Development/physiology , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Nanog Homeobox Protein , Transcription Factor AP-2/genetics , Transcription Factors/genetics , Trophoblasts/cytology , Trophoblasts/physiology
8.
Biol Reprod ; 82(1): 214-23, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19776388

ABSTRACT

Formation of the germ cell lineage involves multiple processes, including repression of somatic differentiation and reacquisition of pluripotency as well as a unique epigenetic constitution. The transcriptional regulator Prdm1 has been identified as a main coordinator of this process, controlling epigenetic modification and gene expression. Here we report on the expression pattern of the transcription factor Tcfap2c, a putative downstream target of Prdm1, during normal mouse embryogenesis and the consequences of its specific loss in primordial germ cells (PGCs) and their derivatives. Tcfap2c is expressed in PGCs from Embryonic Day 7.25 (E 7.25) up to E 12.5, and targeted disruption resulted in sterile animals, both male and female. In the mutant animals, PGCs were specified but were lost around E 8.0. PGCs generated in vitro from embryonic stem cells lacking TCFAP2C displayed induction of Prdm1 and Dppa3. Upregulation of Hoxa1, Hoxb1, and T together with lack of expression of germ cell markers such Nanos3, Dazl, and Mutyh suggested that the somatic gene program is induced in TCFAP2C-deficient PGCs. Repression of TCFAP2C in TCam-2, a human PGC-resembling seminoma cell line, resulted in specific upregulation of HOXA1, HOXB1, MYOD1, and HAND1, indicative of mesodermal differentiation. Expression of genes indicative of ectodermal, endodermal, or extraembryonic differentiation, as well as the finding of no change to epigenetic modifications, suggested control by other factors. Our results implicate Tcfap2c as an important effector of Prdm1 activity that is required for PGC maintenance, most likely mediating Prdm1-induced suppression of mesodermal differentiation.


Subject(s)
Germ Cells/growth & development , Transcription Factor AP-2/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Female , Germ Cells/metabolism , Male , Mesoderm/metabolism , Mice , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1 , Reproduction , Transcription Factors/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...