Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
EMBO Rep ; 18(2): 319-333, 2017 02.
Article in English | MEDLINE | ID: mdl-28007765

ABSTRACT

The transcription factor Oct4 is a core component of molecular cocktails inducing pluripotent stem cells (iPSCs), while other members of the POU family cannot replace Oct4 with comparable efficiency. Rather, group III POU factors such as Oct6 induce neural lineages. Here, we sought to identify molecular features determining the differential DNA-binding and reprogramming activity of Oct4 and Oct6. In enhancers of pluripotency genes, Oct4 cooperates with Sox2 on heterodimeric SoxOct elements. By re-analyzing ChIP-Seq data and performing dimerization assays, we found that Oct6 homodimerizes on palindromic OctOct more cooperatively and more stably than Oct4. Using structural and biochemical analyses, we identified a single amino acid directing binding to the respective DNA elements. A change in this amino acid decreases the ability of Oct4 to generate iPSCs, while the reverse mutation in Oct6 does not augment its reprogramming activity. Yet, with two additional amino acid exchanges, Oct6 acquires the ability to generate iPSCs and maintain pluripotency. Together, we demonstrate that cell type-specific POU factor function is determined by select residues that affect DNA-dependent dimerization.


Subject(s)
Cell Transdifferentiation/genetics , Cellular Reprogramming/genetics , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , POU Domain Factors/chemistry , POU Domain Factors/metabolism , Protein Multimerization , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Embryonic Stem Cells , Enhancer Elements, Genetic , Epigenesis, Genetic , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Models, Molecular , Nucleotide Motifs , Octamer Transcription Factors/chemistry , Octamer Transcription Factors/genetics , Octamer Transcription Factors/metabolism , POU Domain Factors/genetics , Promoter Regions, Genetic , Protein Binding , Protein Conformation , Protein Stability , Transcriptome
2.
Cell Rep ; 15(4): 787-800, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27149845

ABSTRACT

It has previously been reported that mouse epiblast stem cell (EpiSC) lines comprise heterogeneous cell populations that are functionally equivalent to cells of either early- or late-stage postimplantation development. So far, the establishment of the embryonic stem cell (ESC) pluripotency gene regulatory network through the widely known chemical inhibition of MEK and GSK3beta has been impractical in late-stage EpiSCs. Here, we show that chemical inhibition of casein kinase 1alpha (CK1alpha) induces the conversion of recalcitrant late-stage EpiSCs into ESC pluripotency. CK1alpha inhibition directly results in the simultaneous activation of the WNT signaling pathway, together with inhibition of the TGFbeta/SMAD2 signaling pathway, mediating the rewiring of the gene regulatory network in favor of an ESC-like state. Our findings uncover a molecular mechanism that links CK1alpha to ESC pluripotency through the direct modulation of WNT and TGFbeta signaling.

3.
Sci Rep ; 5: 13533, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26314899

ABSTRACT

The transcription factors OCT4 and SOX2 are required for generating induced pluripotent stem cells (iPSCs) and for maintaining embryonic stem cells (ESCs). OCT4 and SOX2 associate and bind to DNA in different configurations depending on the arrangement of their individual DNA binding elements. Here we have investigated the role of the different OCT4-SOX2-DNA assemblies in regulating and inducing pluripotency. To this end, we have generated SOX2 mutants that interfere with specific OCT4-SOX2 heterodimer configurations and assessed their ability to generate iPSCs and to rescue ESC self-renewal. Our results demonstrate that the OCT4-SOX2 configuration that dimerizes on a Hoxb1-like composite, a canonical element with juxtaposed individual binding sites, plays a more critical role in the induction and maintenance of pluripotency than any other OCT4-SOX2 configuration. Overall, the results of this study provide new insight into the protein interactions required to establish a de novo pluripotent network and to maintain a true pluripotent cell fate.


Subject(s)
Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Protein Multimerization , SOXB1 Transcription Factors/metabolism , Animals , Cell Differentiation , Cellular Reprogramming , Human Embryonic Stem Cells , Humans , Mice, Transgenic , Models, Molecular , Pluripotent Stem Cells/cytology
4.
Biores Open Access ; 3(1): 1-8, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24570840

ABSTRACT

BAF chromatin remodeling complexes containing the BRG1 protein have been shown to be not only essential for early embryonic development, but also paramount in enhancing the efficiency of reprogramming somatic cells to pluripotency mediated by four transcription factors. To investigate the role of BRG1 in regulating pluripotency, we found that Oct4 and Nanog levels were increased immediately after BRG1 knockdown. While Nanog levels remained elevated over the investigated time period, Oct4 levels decreased at later time points. Additionally, OCT4 target genes were also found to be upregulated upon Brg1 knockdown. SiRNA-mediated BRG1 knockdown in embryonic stem (ES) cells led to Oct4 and Nanog upregulation, whereas F9 cells showed primarily Oct4 upregulation. BRG1 knockdown upregulated the expression of differentiation markers in mouse ES cells as well as differentiated morphology under reduced leukemia inhibitory factor conditions. Our results show that BRG1 plays an important role in maintaining pluripotency by fine-tuning the expression level of Oct4 and other pluripotency-associated genes.

5.
Nature ; 495(7442): 516-9, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23503660

ABSTRACT

Embryonic stem (ES) cells are pluripotent and characterized by open chromatin and high transcription levels, achieved through auto-regulatory and feed-forward transcription factor loops. ES-cell identity is maintained by a core of factors including Oct4 (also known as Pou5f1), Sox2, Klf4, c-Myc (OSKM) and Nanog, and forced expression of the OSKM factors can reprogram somatic cells into induced pluripotent stem cells (iPSCs) resembling ES cells. These gene-specific factors for RNA-polymerase-II-mediated transcription recruit transcriptional cofactors and chromatin regulators that control access to and activity of the basal transcription machinery on gene promoters. How the basal transcription machinery is involved in setting and maintaining the pluripotent state is unclear. Here we show that knockdown of the transcription factor IID (TFIID) complex affects the pluripotent circuitry in mouse ES cells and inhibits reprogramming of fibroblasts. TFIID subunits and the OSKM factors form a feed-forward loop to induce and maintain a stable transcription state. Notably, transient expression of TFIID subunits greatly enhanced reprogramming. These results show that TFIID is critical for transcription-factor-mediated reprogramming. We anticipate that, by creating plasticity in gene expression programs, transcription complexes such as TFIID assist reprogramming into different cellular states.


Subject(s)
Pluripotent Stem Cells/metabolism , Transcription Factor TFIID/metabolism , Transcription, Genetic , Animals , Cellular Reprogramming/genetics , Chromatin/genetics , Chromatin/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Male , Mice , Pluripotent Stem Cells/cytology , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , TATA-Binding Protein Associated Factors/deficiency , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , TATA-Box Binding Protein/metabolism , Transcription Factor TFIID/deficiency , Transcription Factor TFIID/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Nat Cell Biol ; 15(3): 295-301, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23376973

ABSTRACT

Terminally differentiated cells can be reprogrammed to pluripotency by the forced expression of Oct4, Sox2, Klf4 and c-Myc. However, it remains unknown how this leads to the multitude of epigenetic changes observed during the reprogramming process. Interestingly, Oct4 is the only factor that cannot be replaced by other members of the same family to induce pluripotency. To understand the unique role of Oct4 in reprogramming, we determined the structure of its POU domain bound to DNA. We show that the linker between the two DNA-binding domains is structured as an α-helix and exposed to the protein's surface, in contrast to the unstructured linker of Oct1. Point mutations in this α-helix alter or abolish the reprogramming activity of Oct4, but do not affect its other fundamental properties. On the basis of mass spectrometry studies of the interactome of wild-type and mutant Oct4, we propose that the linker functions as a protein-protein interaction interface and plays a crucial role during reprogramming by recruiting key epigenetic players to Oct4 target genes. Thus, we provide molecular insights to explain how Oct4 contributes to the reprogramming process.


Subject(s)
Cell Differentiation , Cellular Reprogramming , DNA/metabolism , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Octamer Transcription Factor-3/chemistry , Pluripotent Stem Cells/cytology , Amino Acid Sequence , Animals , Blotting, Western , Cells, Cultured , Crystallography, X-Ray , DNA/genetics , Electrophoretic Mobility Shift Assay , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Fibroblasts/metabolism , Humans , Kruppel-Like Factor 4 , Luciferases/metabolism , Mice , Molecular Sequence Data , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.
Nat Commun ; 3: 1279, 2012.
Article in English | MEDLINE | ID: mdl-23232409

ABSTRACT

The evolutionary origins of the gene network underlying cellular pluripotency, a central theme in developmental biology, have yet to be elucidated. In mammals, Oct4 is a factor crucial in the reprogramming of differentiated cells into induced pluripotent stem cells. The Oct4 and Pou2 genes evolved from a POU class V gene ancestor, but it is unknown whether pluripotency induced by Oct4 gene activity is a feature specific to mammals or was already present in ancestral vertebrates. Here we report that different vertebrate Pou2 and Oct4 homologues can induce pluripotency in mouse and human fibroblasts and that the inability of zebrafish Pou2 to establish pluripotency is not representative of all Pou2 genes, as medaka Pou2 and axolotl Pou2 are able to reprogram somatic cells into pluripotent cells. Therefore, our results indicate that induction of pluripotency is not a feature specific to mammals, but existed in the Oct4/Pou2 common ancestral vertebrate.


Subject(s)
Cell Differentiation/physiology , Octamer Transcription Factor-3/physiology , Pluripotent Stem Cells/physiology , Vertebrates/physiology , Zebrafish Proteins/physiology , Adult , Ambystoma mexicanum/embryology , Ambystoma mexicanum/physiology , Animals , Biological Evolution , Cloning, Molecular , Female , Fibroblasts/physiology , Humans , In Situ Hybridization , Mice , Middle Aged , Oryzias/physiology , Phylogeny , Zebrafish/physiology
8.
PLoS One ; 7(4): e34645, 2012.
Article in English | MEDLINE | ID: mdl-22485183

ABSTRACT

Expression of the four transcription factors Oct4, Sox2, Klf4, and c-Myc (OSKM) is sufficient to reprogram somatic cells into induced pluripotent stem (iPSCs). However, this process is slow and inefficient compared with the fusion of somatic cells with embryonic stem cells (ESCs), indicating that ESCs express additional factors that can enhance the efficiency of reprogramming. We had previously developed a method to detect and isolate early neural induction intermediates during the differentiation of mouse ESCs. Using the gene expression profiles of these intermediates, we identified 23 ESC-specific transcripts and tested each for the ability to enhance iPSC formation. Of the tested factors, zinc finger protein 296 (Zfp296) led to the largest increase in mouse iPSC formation. We confirmed that Zfp296 was specifically expressed in pluripotent stem cells and germ cells. Zfp296 in combination with OSKM induced iPSC formation earlier and more efficiently than OSKM alone. Through mouse chimera and teratoma formation, we demonstrated that the resultant iPSCs were pluripotent. We showed that Zfp296 activates transcription of the Oct4 gene via the germ cell-specific conserved region 4 (CR4), and when overexpressed in mouse ESCs leads to upregulation of Nanog expression and downregulation of the expression of differentiation markers, including Sox17, Eomes, and T, which is consistent with the observation that Zfp296 enhances the efficiency of reprogramming. In contrast, knockdown of Zfp296 in ESCs leads to the expression of differentiation markers. Finally, we demonstrated that expression of Zfp296 in ESCs inhibits, but does not block, differentiation into neural cells.


Subject(s)
DNA-Binding Proteins/physiology , Induced Pluripotent Stem Cells/physiology , Animals , Antigens, Differentiation/metabolism , Cell Dedifferentiation , Cell Differentiation , Chimera , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Female , Gene Expression , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/transplantation , Kruppel-Like Factor 4 , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Octamer Transcription Factor-3/genetics , Organ Specificity , Promoter Regions, Genetic , Teratoma/pathology , Transcription Factors/genetics , Transcription Factors/physiology
9.
BMC Evol Biol ; 11: 267, 2011 Sep 24.
Article in English | MEDLINE | ID: mdl-21942985

ABSTRACT

BACKGROUND: The prediction of transcription factor binding sites is difficult for many reasons. Thus, filtering methods are needed to enrich for biologically relevant (true positive) matches in the large amount of computational predictions that are frequently generated from promoter sequences. RESULTS: ReXSpecies 2 filters predictions of transcription factor binding sites and generates a set of figures displaying them in evolutionary context. More specifically, it uses position specific scoring matrices to search for motifs that specify transcription factor binding sites. It removes redundant matches and filters the remaining matches by the phylogenetic group that the matrices belong to. It then identifies potential transcriptional modules, and generates figures that highlight such modules, taking evolution into consideration. Module formation, scoring by evolutionary criteria and visual clues reduce the amount of predictions to a manageable scale. Identification of transcription factor binding sites of particular functional importance is left to expert filtering. ReXSpecies 2 interacts with genome browsers to enable scientists to filter predictions together with other sequence-related data. CONCLUSIONS: Based on ReXSpecies 2, we derive plausible hypotheses about the regulation of pluripotency. Our tool is designed to analyze transcription factor binding site predictions considering their common pattern of occurrence, highlighting their evolutionary history.


Subject(s)
Binding Sites/genetics , Computational Biology/methods , Software Validation , Software , Transcription Factors/genetics , Luciferases , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism
10.
Development ; 137(21): 3551-60, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20876643

ABSTRACT

Oct1 (Pou2f1) is a transcription factor of the POU-homeodomain family that is unique in being ubiquitously expressed in both embryonic and adult mouse tissues. Although its expression profile suggests a crucial role in multiple regions of the developing organism, the only essential function demonstrated so far has been the regulation of cellular response to oxidative and metabolic stress. Here, we describe a loss-of-function mouse model for Oct1 that causes early embryonic lethality, with Oct1-null embryos failing to develop beyond the early streak stage. Molecular and morphological analyses of Oct1 mutant embryos revealed a failure in the establishment of a normal maternal-embryonic interface due to reduced extra-embryonic ectoderm formation and lack of the ectoplacental cone. Oct1(-/-) blastocysts display proper segregation of trophectoderm and inner cell mass lineages. However, Oct1 loss is not compatible with trophoblast stem cell derivation. Importantly, the early gastrulation defect caused by Oct1 disruption can be rescued in a tetraploid complementation assay. Oct1 is therefore primarily required for the maintenance and differentiation of the trophoblast stem cell compartment during early post-implantation development. We present evidence that Cdx2, which is expressed at high levels in trophoblast stem cells, is a direct transcriptional target of Oct1. Our data also suggest that Oct1 is required in the embryo proper from late gastrulation stages onwards.


Subject(s)
Embryonic Development/genetics , Organic Cation Transporter 1/physiology , Trophoblasts/physiology , Animals , Cell Differentiation , Cells, Cultured , Embryo Loss/genetics , Embryo, Mammalian , Female , Gestational Age , Mice , Mice, Inbred C57BL , Mice, Knockout , Organic Cation Transporter 1/genetics , Organic Cation Transporter 1/metabolism , Pregnancy , Time Factors , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...