Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38895208

ABSTRACT

A defined number of hematopoietic stem cell (HSC) clones are born during development and expand to form the pool of adult stem cells. An intricate balance between self-renewal and differentiation of these HSCs supports hematopoiesis for life. HSC fate is determined by complex transcription factor networks that drive cell-type specific gene programs. The transcription factor RUNX1 is required for definitive hematopoiesis, and mutations in Runx1 have been shown to reduce clonal diversity. The RUNX1 cofactor, CBFý, stabilizes RUNX1 binding to DNA, and disruption of their interaction alters downstream gene expression. Chemical screening for modulators of Runx1 and HSC expansion in zebrafish led us to identify a new mechanism for the RUNX1 inhibitor, Ro5-3335. We found that Ro5-3335 increased HSC divisions in zebrafish, and animals transplanted with Ro5-3335 treated cells had enhanced chimerism compared to untreated cells. Using human CD34+ cells, we show that Ro5-3335 remodels the RUNX1 transcription complex by binding to ELF1, independent of CBFý. This allows specific expression of cell cycle and hematopoietic genes that enhance HSC self-renewal and prevent differentiation. Furthermore, we provide the first evidence to show that it is possible to pharmacologically increase the number of stem cell clones in vivo , revealing a previously unknown mechanism for enhancing clonal diversity. Our studies have revealed a mechanism by which binding partners of RUNX1 determine cell fate, with ELF transcription factors guiding cell division. This information could lead to treatments that enhance clonal diversity for blood diseases.

2.
Cell Stem Cell ; 29(8): 1181-1196.e6, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35931029

ABSTRACT

Human induced pluripotent stem cells (iPSCs) provide a potentially unlimited resource for cell therapies, but the derivation of mature cell types remains challenging. The histone methyltransferase EZH1 is a negative regulator of lymphoid potential during embryonic hematopoiesis. Here, we demonstrate that EZH1 repression facilitates in vitro differentiation and maturation of T cells from iPSCs. Coupling a stroma-free T cell differentiation system with EZH1-knockdown-mediated epigenetic reprogramming, we generated iPSC-derived T cells, termed EZ-T cells, which display a highly diverse T cell receptor (TCR) repertoire and mature molecular signatures similar to those of TCRαß T cells from peripheral blood. Upon activation, EZ-T cells give rise to effector and memory T cell subsets. When transduced with chimeric antigen receptors (CARs), EZ-T cells exhibit potent antitumor activities in vitro and in xenograft models. Epigenetic remodeling via EZH1 repression allows efficient production of developmentally mature T cells from iPSCs for applications in adoptive cell therapy.


Subject(s)
Induced Pluripotent Stem Cells , Receptors, Chimeric Antigen , Cell Differentiation , Humans , Immunotherapy, Adoptive , Induced Pluripotent Stem Cells/metabolism , Polycomb Repressive Complex 2/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes
3.
Nat Cell Biol ; 24(4): 579-589, 2022 04.
Article in English | MEDLINE | ID: mdl-35414020

ABSTRACT

Intercellular communication orchestrates a multitude of physiologic and pathologic conditions. Algorithms to infer cell-cell communication and predict downstream signalling and regulatory networks are needed to illuminate mechanisms of stem cell differentiation and tissue development. Here, to fill this gap, we developed and applied CellComm to investigate how the aorta-gonad-mesonephros microenvironment dictates haematopoietic stem and progenitor cell emergence. We identified key microenvironmental signals and transcriptional networks that regulate haematopoietic development, including Stat3, Nr0b2, Ybx1 and App, and confirmed their roles using zebrafish, mouse and human models. Notably, CellComm revealed extensive crosstalk among signalling pathways and convergence on common transcriptional regulators, indicating a resilient developmental programme that ensures dynamic adaptation to changes in the embryonic environment. Our work provides an algorithm and data resource for the scientific community.


Subject(s)
Hematopoietic Stem Cells , Zebrafish , Animals , Cell Differentiation , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Mesonephros/metabolism , Mice , Zebrafish/genetics
4.
Nat Commun ; 12(1): 7322, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34916498

ABSTRACT

Blastocyst-derived stem cell lines were shown to self-organize into embryo-like structures in 3D cell culture environments. Here, we provide evidence that embryo-like structures can be generated solely based on transcription factor-mediated reprogramming of embryonic stem cells in a simple 3D co-culture system. Embryonic stem cells in these cultures self-organize into elongated, compartmentalized embryo-like structures reflecting aspects of the inner regions of the early post-implantation embryo. Single-cell RNA-sequencing reveals transcriptional profiles resembling epiblast, primitive-/visceral endoderm, and extraembryonic ectoderm of early murine embryos around E4.5-E5.5. In this stem cell-based embryo model, progression from rosette formation to lumenogenesis accompanied by progression from naïve- to primed pluripotency was observed within Epi-like cells. Additionally, lineage specification of primordial germ cells and distal/anterior visceral endoderm-like cells was observed in epiblast- or visceral endoderm-like compartments, respectively. The system presented in this study allows for fast and reproducible generation of embryo-like structures, providing an additional tool to study aspects of early embryogenesis.


Subject(s)
Embryoid Bodies/cytology , Embryonic Development , Embryonic Stem Cells/cytology , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Culture Techniques, Three Dimensional , Cellular Reprogramming , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryoid Bodies/metabolism , Embryonic Stem Cells/metabolism , Endoderm/embryology , Endoderm/metabolism , Gene Expression Regulation, Developmental , Mice , RNA-Seq
5.
J Clin Invest ; 131(22)2021 11 15.
Article in English | MEDLINE | ID: mdl-34779407

ABSTRACT

High expression of LIN28B is associated with aggressive malignancy and poor survival. Here, probing MYCN-amplified neuroblastoma as a model system, we showed that LIN28B expression was associated with enhanced cell migration in vitro and invasive and metastatic behavior in murine xenografts. Sequence analysis of the polyribosome fraction of LIN28B-expressing neuroblastoma cells revealed let-7-independent enrichment of transcripts encoding components of the translational and ribosomal apparatus and depletion of transcripts of neuronal developmental programs. We further observed that LIN28B utilizes both its cold shock and zinc finger RNA binding domains to preferentially interact with MYCN-induced transcripts of the ribosomal complex, enhancing their translation. These data demonstrated that LIN28B couples the MYCN-driven transcriptional program to enhanced ribosomal translation, thereby implicating LIN28B as a posttranscriptional driver of the metastatic phenotype.


Subject(s)
N-Myc Proto-Oncogene Protein/physiology , Neoplasm Metastasis , Neuroblastoma/pathology , RNA-Binding Proteins/physiology , Ribosomes/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Neuroblastoma/etiology
6.
Blood Adv ; 4(19): 4679-4692, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33002135

ABSTRACT

Fanconi anemia (FA) is a disorder of DNA repair that manifests as bone marrow (BM) failure. The lack of accurate murine models of FA has refocused efforts toward differentiation of patient-derived induced pluripotent stem cells (IPSCs) to hematopoietic progenitor cells (HPCs). However, an intact FA DNA repair pathway is required for efficient IPSC derivation, hindering these efforts. To overcome this barrier, we used inducible complementation of FANCA-deficient IPSCs, which permitted robust maintenance of IPSCs. Modulation of FANCA during directed differentiation to HPCs enabled the production of FANCA-deficient human HPCs that recapitulated FA genotoxicity and hematopoietic phenotypes relative to isogenic FANCA-expressing HPCs. FANCA-deficient human HPCs underwent accelerated terminal differentiation driven by activation of p53/p21. We identified growth arrest specific 6 (GAS6) as a novel target of activated p53 in FANCA-deficient HPCs and modulate GAS6 signaling to rescue hematopoiesis in FANCA-deficient cells. This study validates our strategy to derive a sustainable, highly faithful human model of FA, uncovers a mechanism of HPC exhaustion in FA, and advances toward future cell therapy in FA.


Subject(s)
Fanconi Anemia , Induced Pluripotent Stem Cells , Animals , Cell Differentiation , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group A Protein/genetics , Humans , Mice , Tumor Suppressor Protein p53/genetics
7.
Dev Cell ; 55(2): 133-149.e6, 2020 10 26.
Article in English | MEDLINE | ID: mdl-32810442

ABSTRACT

Embryonic hematopoietic stem and progenitor cells (HSPCs) robustly proliferate while maintaining multilineage potential in vivo; however, an incomplete understanding of spatiotemporal cues governing their generation has impeded robust production from human induced pluripotent stem cells (iPSCs) in vitro. Using the zebrafish model, we demonstrate that NLRP3 inflammasome-mediated interleukin-1-beta (IL1ß) signaling drives HSPC production in response to metabolic activity. Genetic induction of active IL1ß or pharmacologic inflammasome stimulation increased HSPC number as assessed by in situ hybridization for runx1/cmyb and flow cytometry. Loss of inflammasome components, including il1b, reduced CD41+ HSPCs and prevented their expansion in response to metabolic cues. Cell ablation studies indicated that macrophages were essential for initial inflammasome stimulation of Il1rl1+ HSPCs. Significantly, in human iPSC-derived hemogenic precursors, transient inflammasome stimulation increased multilineage hematopoietic colony-forming units and T cell progenitors. This work establishes the inflammasome as a conserved metabolic sensor that expands HSPC production in vivo and in vitro.


Subject(s)
Embryonic Stem Cells/metabolism , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Inflammasomes/metabolism , Animals , Cell Differentiation/physiology , Core Binding Factor Alpha 2 Subunit/metabolism , Embryo, Nonmammalian/metabolism , Embryonic Development/physiology , Hematopoiesis/physiology , Humans , Zebrafish/embryology
8.
Int J Mol Sci ; 21(15)2020 Jul 31.
Article in English | MEDLINE | ID: mdl-32752102

ABSTRACT

Activating mutations in the human KIT receptor is known to drive severe hematopoietic disorders and tumor formation spanning various entities. The most common mutation is the substitution of aspartic acid at position 816 to valine (D816V), rendering the receptor constitutively active independent of ligand binding. As the role of the KIT receptor in placental signaling cascades is poorly understood, we analyzed the impact of KITD816V expression on placental development using a humanized mouse model. Placentas from KITD816V animals present with a grossly changed morphology, displaying a reduction in labyrinth and spongiotrophoblast layer and an increase in the Parietal Trophoblast Giant Cell (P-TGC) layer. Elevated differentiation to P-TGCs was accompanied with reduced differentiation to other Trophoblast Giant Cell (TGC) subtypes and by severe decrease in proliferation. The embryos display growth retardation and die in utero. KITD816V-trophoblast stem cells (TSC) differentiate much faster compared to wild type (WT) controls. In undifferentiated KITD816V-TSCs, levels of Phosphorylated Extracellular-signal Regulated Kinase (P-ERK) and Phosphorylated Protein Kinase B (P-AKT) are comparable to wildtype cultures differentiating for 3-6 days. Accordingly, P-TGC markers Placental Lactogen 1 (PL1) and Proliferin (PLF) are upregulated as well. The results reveal that KIT signaling orchestrates the fine-tuned differentiation of the placenta, with special emphasis on P-TGC differentiation. Appropriate control of KIT receptor action is therefore essential for placental development and nourishment of the embryo.


Subject(s)
Homeobox A10 Proteins/genetics , Placentation/genetics , Prolactin/genetics , Proto-Oncogene Proteins c-kit/genetics , Trophoblasts/metabolism , Animals , Female , Gene Expression Regulation, Developmental/genetics , Humans , MAP Kinase Signaling System/genetics , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Placenta/metabolism , Placental Lactogen/genetics , Pregnancy , Proto-Oncogene Proteins c-akt/genetics , Trophoblasts/pathology
9.
Stem Cell Reports ; 14(5): 956-971, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32302558

ABSTRACT

Studies of hematopoietic stem cell (HSC) development from pre-HSC-producing hemogenic endothelial cells (HECs) are hampered by the rarity of these cells and the presence of other cell types with overlapping marker expression profiles. We generated a Tg(Runx1-mKO2; Ly6a-GFP) dual reporter mouse to visualize hematopoietic commitment and study pre-HSC emergence and maturation. Runx1-mKO2 marked all intra-arterial HECs and hematopoietic cluster cells (HCCs), including pre-HSCs, myeloid- and lymphoid progenitors, and HSCs themselves. However, HSC and lymphoid potential were almost exclusively found in reporter double-positive (DP) cells. Robust HSC activity was first detected in DP cells of the placenta, reflecting the importance of this niche for (pre-)HSC maturation and expansion before the fetal liver stage. A time course analysis by single-cell RNA sequencing revealed that as pre-HSCs mature into fetal liver stage HSCs, they show signs of interferon exposure, exhibit signatures of multi-lineage differentiation gene expression, and develop a prolonged cell cycle reminiscent of quiescent adult HSCs.


Subject(s)
Antigens, Ly/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Genes, Reporter , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Membrane Proteins/genetics , Transcriptome , Animals , Antigens, Ly/metabolism , Cells, Cultured , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/cytology , Membrane Proteins/metabolism , Mice , RNA-Seq , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Single-Cell Analysis
10.
Placenta ; 90: 128-137, 2020 01 15.
Article in English | MEDLINE | ID: mdl-32056544

ABSTRACT

INTRODUCTION: The first lineage separation in mammalian development occurs when totipotent cells of the zygote give rise to the inner cell mass and the trophectoderm. The lineages are strictly separated by an epigenetic barrier. In vitro derivatives of these lineages embryonic stem cells (ESC) and trophoblast stem cells (TSC) are used to study the requirements needed to overcome the barrier in ESC to TSC conversion approaches. METHODS: Different combinations of TSC transcription factors were induced in ESC for three days. Cells were kept in TS medium with fetal bovine serum (FBS) or the chemically defined TX medium. Obtained cells were analysed for OCT4 levels, TSC surface marker levels, expression of TSC markers and methylation status of Elf5, Oct4 and Nanog promoters. Further, long-term culture stability and in vitro and in vivo differentiation was tested. RESULTS: Overexpression of Gata3, Eomes, Tfap2c, Ets2 and Cdx2 in ESC resulted in induction of TSC fate. Overexpression of Cdx2 or four factors (Gata3, Eomes, Tfap2c and Ets2) resulted in complete conversion only when cells were cultured in TX medium. The obtained induced TSC (iTSC) display characteristics of bona fide TSC in terms of marker expression and promoter methylation patterns. The generated converted cells were shown to display self-renewal and to be capable to differentiate into TSC derivatives in vitro and in vivo. CONCLUSION: Gata3, Eomes, Tfap2c, Ets2 and Cdx2 overexpression in ESC resulted in stable iTSC fate independent of culture conditions. For four factors or Cdx2 alone, TX medium is required for complete TSC conversion.


Subject(s)
Cell Culture Techniques , Cell Differentiation/physiology , Culture Media , Embryonic Stem Cells/cytology , Trophoblasts/cytology , Animals , Cell Lineage/physiology , Mice
11.
Cell Res ; 27(8): 961-962, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28429768

ABSTRACT

Researchers at the University of Cambridge, UK have succeeded in reconstructing mouse embryos by combining pluripotent embryonic and multipotent trophoblast stem cells in a 3D scaffold; the study from the laboratory of Professor Zernicka-Goetz, recently published in Science, provides a break-through tool to probe early mammalian development outside the uterus. Achieving a similar feat with human cells might necessitate reconsideration of the 14-day rule as a limitation of such research.


Subject(s)
Mouse Embryonic Stem Cells/metabolism , Tissue Scaffolds/chemistry , Trophoblasts/metabolism , Animals , Embryo Culture Techniques/methods , Mice , Mouse Embryonic Stem Cells/cytology , Trophoblasts/cytology
12.
Placenta ; 60 Suppl 1: S52-S56, 2017 12.
Article in English | MEDLINE | ID: mdl-28043657

ABSTRACT

Recently, direct cell fate conversion attempts between the embryonic and extra-embryonic lineage gained new momentum. Two concomitant publications were published, describing the successful generation of transgene-independent, self-renewing trophoblast stem cells (TSCs) from murine fibroblasts. Cells were faithfully converted, displaying high similarity to blastocyst or extraembryonic ectoderm derived TSCs. Here, we summarize and compare published attempts aiming at the direct induction of trophoblast-fate from either mouse embryonic stem cells or fibroblasts.


Subject(s)
Cell Lineage , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Models, Biological , Trophoblasts/cytology , Animals , Blastocyst/cytology , Cell Dedifferentiation , Cell Differentiation , Cell Line , Cell Transdifferentiation , Cells, Cultured , Female , Mice , Placenta/cytology , Pregnancy , Stem Cell Research
13.
J Vis Exp ; (113)2016 07 25.
Article in English | MEDLINE | ID: mdl-27500445

ABSTRACT

Trophoblast stem cells (TSCs) arise as a consequence of the first cell fate decision in mammalian development. They can be cultured in vitro, retaining the ability to self-renew and to differentiate into all subtypes of the trophoblast lineage, equivalent to the in vivo stem cell population giving rise to the fetal portion of the placenta. Therefore, TSCs offer a unique model to study placental development and embryonic versus extra-embryonic cell fate decision in vitro. From the blastocyst stage onwards, a distinct epigenetic barrier consisting of DNA methylation and histone modifications tightly separates both lineages. Here, we describe a protocol to fully overcome this lineage barrier by transient over-expression of trophoblast key regulators Tfap2c, Gata3, Eomes and Ets2 in murine embryonic fibroblasts. The induced trophoblast stem cells are able to self-renew and are almost identical to blastocyst derived trophoblast stem cells in terms of morphology, marker gene expression and methylation pattern. Functional in vitro and in vivo assays confirm that these cells are able to differentiate along the trophoblast lineage generating polyploid trophoblast giant cells and chimerizing the placenta when injected into blastocysts. The induction of trophoblast stem cells from somatic tissue opens new avenues to study genetic and epigenetic characteristics of this extra-embryonic lineage and offers the possibility to generate trophoblast stem cell lines without destroying the respective embryo.


Subject(s)
Cell Culture Techniques , Cell Differentiation , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Trophoblasts/cytology , Animals , Cell Lineage , Female , Mice , Placenta , Pregnancy
14.
Development ; 143(5): 787-98, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26811378

ABSTRACT

Loss of TFAP2C in mouse leads to developmental defects in the extra-embryonic compartment with lethality at embryonic day (E)7.5. To investigate the requirement of TFAP2C in later placental development, deletion of TFAP2C was induced throughout extra-embryonic ectoderm at E6.5, leading to severe placental abnormalities caused by reduced trophoblast population and resulting in embryonic retardation by E8.5. Deletion of TFAP2C in TPBPA(+) progenitors at E8.5 results in growth arrest of the junctional zone. TFAP2C regulates its target genes Cdkn1a (previously p21) and Dusp6, which are involved in repression of MAPK signaling. Loss of TFAP2C reduces activation of ERK1/2 in the placenta. Downregulation of Akt1 and reduced activation of phosphorylated AKT in the mutant placenta are accompanied by impaired glycogen synthesis. Loss of TFAP2C led to upregulation of imprinted gene H19 and downregulation of Slc38a4 and Ascl2. The placental insufficiency post E16.5 causes fetal growth restriction, with 19% lighter mutant pups. Knockdown of TFAP2C in human trophoblast choriocarcinoma JAr cells inhibited MAPK and AKT signaling. Thus, we present a model where TFAP2C in trophoblasts controls proliferation by repressing Cdkn1a and activating the MAPK pathway, further supporting differentiation of glycogen cells by activating the AKT pathway.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Pregnancy Proteins/physiology , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor AP-2/physiology , Animals , Cell Differentiation , Cell Proliferation , Crosses, Genetic , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genotype , Glycogen/metabolism , In Situ Hybridization , Integrases/genetics , Integrases/physiology , Male , Mice , Phosphorylation , Placenta/metabolism , Pregnancy , Pregnancy Proteins/genetics , Transcription Factor AP-2/genetics , Transgenes , Trophoblasts/metabolism
15.
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
16.
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
17.
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
18.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...