Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
J Mol Biol ; 432(7): 2271-2288, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32105733

ABSTRACT

R-loops are a prevalent class of non-B DNA structures that have been associated with both positive and negative cellular outcomes. DNA:RNA immunoprecipitation (DRIP) approaches based on the anti-DNA:RNA hybrid S9.6 antibody revealed that R-loops form dynamically over conserved genic hotspots. We have developed an orthogonal approach that queries R-loops via the presence of long stretches of single-stranded DNA on their looped-out strand. Nondenaturing sodium bisulfite treatment catalyzes the conversion of unpaired cytosines to uracils, creating permanent genetic tags for the position of an R-loop. Long-read, single-molecule PacBio sequencing allows the identification of R-loop 'footprints' at near nucleotide resolution in a strand-specific manner on long single DNA molecules and at ultra-deep coverage. Single-molecule R-loop footprinting coupled with PacBio sequencing (SMRF-seq) revealed a strong agreement between S9.6-based and bisulfite-based R-loop mapping and confirmed that R-loops form over genic hotspots, including gene bodies and terminal gene regions. Based on the largest single-molecule R-loop dataset to date, we show that individual R-loops form nonrandomly, defining discrete sets of overlapping molecular clusters that pileup through larger R-loop zones. R-loops most often map to intronic regions and their individual start and stop positions do not match with intron-exon boundaries, reinforcing the model that they form cotranscriptionally from unspliced transcripts. SMRF-seq further established that R-loop distribution patterns are not simply driven by intrinsic DNA sequence features but most likely also reflect DNA topological constraints. Overall, DRIP-based and SMRF-based approaches independently provide a complementary and congruent view of R-loop distribution, consolidating our understanding of the principles underlying R-loop formation.


Subject(s)
DNA/chemistry , Embryonal Carcinoma Stem Cells/metabolism , R-Loop Structures , RNA/chemistry , Single-Cell Analysis/methods , Transcription, Genetic , Embryonal Carcinoma Stem Cells/cytology , Humans
2.
Biochem Biophys Res Commun ; 509(2): 603-610, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30606481

ABSTRACT

MicroRNA-135a-5p has been reported to play a potential role in the generation of new neurons. However, the underlying targets of miR-135a-5p in regulating neuronal differentiation have been poorly understood. Our study recently has uncovered that Sox6 and CD44 genes were significantly downregulated during neuronal differentiation of P19 cells, a multipotent cell type. We then found that Sox6 directly bound to the promoter of CD44. Importantly, we identified Sox6 as a direct target of miR-135a-5p. Additionally, we demonstrated that miR-135a-5p is crucial for the neuronal differentiation of P19 cells. More significantly, we found that Sox6 overexpression could overturn miR-135a-5p-mediated neuronal differentiation and dendrite development. In conclusion, these findings indicated that miR-135a-5p/Sox6/CD44 axis provides an important molecular target mechanism for neurodifferentiation.


Subject(s)
Embryonal Carcinoma Stem Cells/pathology , Hyaluronan Receptors/genetics , MicroRNAs/genetics , Neurogenesis , SOXD Transcription Factors/genetics , Animals , Cell Line, Tumor , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Hyaluronan Receptors/metabolism , Mice , SOXD Transcription Factors/metabolism , Signal Transduction
3.
In Vitro Cell Dev Biol Anim ; 54(8): 600-609, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30112697

ABSTRACT

Uc.40 is a long noncoding RNA that is highly conserved among different species, although its function is unknown. It is highly expressed in abnormal human embryonic heart. We previously reported that overexpression of uc.40 promoted apoptosis and inhibited proliferation of P19 cells, and downregulated PBX1, which was identified as a potential target gene of uc.40. The current study evaluated the effects of uc40-siRNA-44 (siRNA against uc.40) on the differentiation, proliferation, apoptosis, and mitochondrial function in P19 cells, and investigated the relationship between uc.40 and PBX1 in cardiomyocytes. The uc.40 silencing expression was confirmed by quantitative real-time polymerase chain reaction (RT-PCR). Observation of morphological changes in transfected P19 cells during different stages of differentiation revealed that uc40-siRNA-44 increased the number of cardiomyocyes. There was no significant difference in the morphology or time of differentiation between the uc40-siRNA-44 group and the control group. uc40-siRNA-44 significantly promoted proliferation of P19 cells and inhibited serum starvation-induced apoptosis. There was no significant difference in mitochondrial DNA copy number or cellular ATP level between the two groups, and ROS levels were significantly decreased in uc40-siRNA-44-transfected cells. The levels of PBX1 and myocardial markers of differentiation were examined in transfected P19 cells; uc40-siRNA-44 downregulated myocardial markers and upregulated PBX1 expression. These results suggest that uc.40 may play an important role during the differentiation of P19 cells by regulation of PBX1 to promote proliferation and inhibit apoptosis. These studies provide a foundation for further study of uc.40/PBX1 in cardiac development.


Subject(s)
Cell Differentiation , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Gene Silencing , Myocytes, Cardiac/cytology , Pre-B-Cell Leukemia Transcription Factor 1/genetics , RNA, Long Noncoding/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/genetics , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Proliferation , DNA, Mitochondrial/genetics , Down-Regulation/genetics , Matrix Metalloproteinases/metabolism , Mice , Mitochondria/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Organ Specificity , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Transfection , Up-Regulation/genetics
4.
In Vitro Cell Dev Biol Anim ; 54(8): 567-579, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30030768

ABSTRACT

P19 embryonal carcinoma cells (EC-cells) provide a simple and robust culture system for studying neural development. Most protocols developed so far for directing neural differentiation of P19 cells depend on the use of culture medium supplemented with retinoic acid (RA) and serum, which has an undefined composition. Hence, such protocols are not suitable for many molecular studies. In this study, we achieved neural differentiation of P19 cells in a serum- and RA-free culture medium by employing the knockout serum replacement (KSR) supplement. In the KSR-containing medium, P19 cells underwent predominant differentiation into neural lineage and by day 12 of culture, neural cells were present in 100% of P19-derived embryoid bodies (EBs). This was consistently accompanied by the increased expression of various neural lineage-associated markers during the course of differentiation. P19-derived neural cells comprised of NES+ neural progenitors (~ 46%), TUBB3+ immature neurons (~ 6%), MAP2+ mature neurons (~ 2%), and GFAP+ astrocytes (~ 50%). A heterogeneous neuronal population consisting of glutamatergic, GABAergic, serotonergic, and dopaminergic neurons was generated. Taken together, our study shows that the KSR medium is suitable for the differentiation of P19 cells to neural lineage without requiring additional (serum and RA) supplements. This stem cell differentiation system could be utilized for gaining mechanistic insights into neural differentiation and for identifying potential neuroactive compounds.


Subject(s)
Cell Differentiation , Embryonal Carcinoma Stem Cells/cytology , Neurons/cytology , Tretinoin/pharmacology , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Culture Media, Serum-Free , Endoderm/cytology , Gene Expression Regulation/drug effects , Mesoderm/cytology , Mice , Neurons/drug effects , Neurons/metabolism
5.
Stem Cell Res ; 31: 83-94, 2018 08.
Article in English | MEDLINE | ID: mdl-30031967

ABSTRACT

Developmental pluripotency associated factor 4 (Dppa4) is a highly specific marker of pluripotent cells, and is also overexpressed in certain cancers, but its function in either of these contexts is poorly understood. In this study, we use ChIP-Seq to identify Dppa4 binding genome-wide in three distinct cell types: mouse embryonic stem cells (mESC), embryonal carcinoma cells, and 3T3 fibroblasts ectopically expressing Dppa4. We find a core set of Dppa4 binding sites shared across cell types, and also a substantial number of sites unique to each cell type. Across cell types Dppa4 shows a preference for binding to regions with active chromatin signatures, and can influence chromatin modifications at target genes. In 3T3 fibroblasts with enforced Dppa4 expression, Dppa4 represses the cell cycle inhibitor Cdkn2c and activates Ets family transcription factor Etv4, leading to alterations in the cell cycle that likely contribute to the oncogenic phenotype. Dppa4 also directly regulates Etv4 in mESC but represses it in this context, and binds with Oct4 to a set of shared targets that are largely independent of Sox2 and Nanog, indicating that Dppa4 functions independently of the core pluripotency network in stem cells. Together these data provide novel insights into Dppa4 function in both pluripotent and oncogenic contexts.


Subject(s)
Embryonal Carcinoma Stem Cells/physiology , Nuclear Proteins/genetics , Pluripotent Stem Cells/physiology , 3T3 Cells , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Cell Proliferation/physiology , Chromatin/genetics , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Gene Expression Regulation , Genomics/methods , Humans , Mice , Nuclear Proteins/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Transfection
6.
Sci Rep ; 8(1): 4625, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29545540

ABSTRACT

The proneural transcription factor Ascl1 is a master regulator of neurogenesis, coordinating proliferation and differentiation in the central nervous system. While its expression is well characterised, post-translational regulation is much less well understood. Here we demonstrate that a population of chromatin-bound Ascl1 can be found associated with short chains of ubiquitin while cytoplasmic Ascl1 harbours much longer ubiquitin chains. Only cytoplasmic ubiquitylation targets Ascl1 for destruction, which occurs by conjugation of ubiquitin to lysines in the basic helix-loop-helix domain of Ascl1 and requires the E3 ligase Huwe1. In contrast, chromatin-bound Ascl1 associated with short ubiquitin-chains, which can occur on lysines within the N-terminal region or the bHLH domain and is not mediated by Huwe1, is not targeted for ubiquitin-mediated destruction. We therefore offer further insights into post-translational regulation of Ascl1, highlighting complex regulation of ubiquitylation and degradation in the cytoplasm and on chromatin.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin/metabolism , Embryonal Carcinoma Stem Cells/metabolism , Neural Stem Cells/metabolism , Subcellular Fractions/metabolism , Ubiquitin/metabolism , Animals , Cell Differentiation , Cells, Cultured , Embryonal Carcinoma Stem Cells/cytology , Mice , Neural Stem Cells/cytology , Neurogenesis , Proteolysis , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
7.
Cell Death Dis ; 9(2): 246, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445146

ABSTRACT

Oxidative stress generates reactive oxygen species (ROS) that can promote or inhibit cardiac differentiation of stem cells dependent on the intensity of stimuli as well as cellular context in redox and differentiation status. In the current study, we confirmed that suitable intensity of hydrogen peroxide at the formation stage of embryoid bodies (EBs) effectively favored the formation of spontaneously beating cardiomyocytes from P19 embryonal carcinoma cells. Mechanistic studies implicated that extrinsic ROS enhanced the Caspase-mediated degradation of Oct4 and Nanog, two factors that governing pluripotent property. Further experiments suggested that a cohort of Nanog together with histone deacetylase 4 (Hdac4) played a critical role in establishing and maintaining the silent transcriptional status of Gata4 and Nkx2.5 in undifferentiated cells. Thus, an impulse of hydrogen peroxide depleted Nanog and Hdac4 via a caspase-dependent manner to ameliorate the repression on Gata4 and Nkx2.5 promoters, thereby generating a persistent activation on cardiac differentiation program. Meanwhile, we found that excessive ROS-activated JNK cascade to facilitate the ubiquitination and subsequent degradation of Gata4 protein. Overall, our results indicate that suitable ROS promotes the activation of Gata4 in transcription, while excessive ROS targets Gata4 protein for proteasome-dependent degradation. Gata4 is an important modulator balancing the promoting and inhibitory effects of oxidative stress on differentiation program of cardiomyogenesis.


Subject(s)
Embryonal Carcinoma Stem Cells/drug effects , GATA4 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Hydrogen Peroxide/pharmacology , Myocytes, Cardiac/drug effects , Transcription, Genetic/drug effects , Animals , Caspases/genetics , Caspases/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Embryo, Mammalian , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , GATA4 Transcription Factor/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Signal Transduction , Ubiquitination/drug effects
8.
Gene ; 627: 369-372, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28633916

ABSTRACT

POU domain proteins are an important family of transcription factors that regulates cell type-specific gene expression. One of the most crucial members of this family that maintains pluripotency and self-renewal of embryonic stem cells is POU5F1/OCT4. The OCT4 gene can generate several variants under different situations/cell types includes OCT4A that is the major factor sustains pluripotency in embryonic stem and embryonic carcinoma cells, and also OCT4B and OCT4B1, which are transcribed from a different potential promoter located in intron1 and are expressed in various tissues and cell types. In present study, during expression check of OCT4B1 in embryonic carcinoma cells (NT2), we discovered a novel OCT4 transcript for the first time and designated it as OCT4B4. This variant is expressed in various human pluripotent cells and its expression is down-regulated upon induction of differentiation. Moreover, knocking down of OCT4B4 by shRNA resulted in increased accumulation of transfected cells in G0/G1 phase compared to the mock-transfected control cells.


Subject(s)
Embryonal Carcinoma Stem Cells/metabolism , Gene Expression Regulation, Neoplastic , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/metabolism , Cell Differentiation , Cell Line , Cell Line, Tumor , Embryonal Carcinoma Stem Cells/cytology , G1 Phase Cell Cycle Checkpoints , Humans , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism
9.
Methods Mol Biol ; 1601: 205-214, 2017.
Article in English | MEDLINE | ID: mdl-28470528

ABSTRACT

Glycine receptor chloride channels (GlyRs) are attractive drug targets for therapeutic intervention and are also more and more recognized in the context of in vitro neurotoxicity and developmental neurotoxicity testing. Assaying the functional properties of GlyR can serve as an indicator of cellular viability and the integrity of the developing and mature central nervous system. Human pluripotent NTERA-2 (NT2) stem cells undergo neuronal differentiation upon stimulation with retinoic acid and express a large variety of neuronal proteins-including GlyR. YFP-I152L, a halide-sensitive variant of yellow fluorescent protein, allows high-throughput fluorescence-based functional analysis of GlyRs in NT2 cells. Here we describe a protocol for phenotyping of cellular viability by functional analysis of GlyR in neuronally differentiated NT2 (NT2-N) cells using YFP-I152L as a reporter of functional integrity of GlyRs. The protocol describes neuronal differentiation of NT2 stem cells, transient transfection of NT2-N cells with YFP-I152L as well as functional imaging and analysis of data from high-content imaging.


Subject(s)
Cell Survival , Embryonal Carcinoma Stem Cells/cytology , Neurogenesis , Neurons/cytology , Pluripotent Stem Cells/cytology , Receptors, Glycine/metabolism , Cell Differentiation , Drug Evaluation, Preclinical , Embryonal Carcinoma Stem Cells/drug effects , Humans , Luminescent Proteins/metabolism , Neurogenesis/drug effects , Optical Imaging/methods , Tretinoin/pharmacology
10.
Genome Res ; 27(6): 947-958, 2017 06.
Article in English | MEDLINE | ID: mdl-28396520

ABSTRACT

Epigenetic mechanisms are believed to play key roles in the establishment of cell-specific transcription programs. Accordingly, the modified bases 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) have been observed in DNA of genomic regulatory regions such as enhancers, and oxidation of 5mC into 5hmC by Ten-eleven translocation (TET) proteins correlates with enhancer activation. However, the functional relationship between cytosine modifications and the chromatin architecture of enhancers remains elusive. To gain insights into their function, 5mC and 5hmC levels were perturbed by inhibiting DNA methyltransferases and TETs during differentiation of mouse embryonal carcinoma cells into neural progenitors, and chromatin characteristics of enhancers bound by the pioneer transcription factors FOXA1, MEIS1, and PBX1 were interrogated. In a large fraction of the tested enhancers, inhibition of DNA methylation was associated with a significant increase in monomethylation of H3K4, a characteristic mark of enhancer priming. In addition, at some specific enhancers, 5mC oxidation by TETs facilitated chromatin opening, a process that may stabilize MEIS1 binding to these genomic regions.


Subject(s)
5-Methylcytosine/metabolism , Chromatin/metabolism , Embryonal Carcinoma Stem Cells/metabolism , Enhancer Elements, Genetic , Epigenesis, Genetic , 5-Methylcytosine/analogs & derivatives , Animals , Cell Differentiation , Chromatin/ultrastructure , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Embryonal Carcinoma Stem Cells/cytology , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Histones/genetics , Histones/metabolism , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription, Genetic , Tumor Cells, Cultured
11.
Mol Cell Proteomics ; 15(11): 3450-3460, 2016 11.
Article in English | MEDLINE | ID: mdl-27634302

ABSTRACT

Polycomb proteins assemble to form complexes with important roles in epigenetic regulation. The Polycomb Repressive Complex 2 (PRC2) modulates the di- and tri-methylation of lysine 27 on histone H3, each of which are associated with gene repression. Although three subunits, EZH1/2, SUZ12, and EED, form the catalytic core of PRC2, a wider group of proteins associate with low stoichiometry. This raises the question of whether dynamic variation of the PRC2 interactome results in alternative forms of the complex during differentiation. Here we compared the physical interactions of PRC2 in undifferentiated and differentiated states of NTERA2 pluripotent embryonic carcinoma cells. Label-free quantitative proteomics was used to assess endogenous immunoprecipitation of the EZH2 and SUZ12 subunits of PRC2. A high stringency data set reflecting the endogenous state of PRC2 was produced that included all previously reported core and associated PRC2 components, and several novel interacting proteins. Comparison of the interactomes obtained in undifferentiated and differentiated cells revealed candidate proteins that were enriched in complexes isolated from one of the two states. For example, SALL4 and ZNF281 associate with PRC2 in pluripotent cells, whereas PCL1 and SMAD3 preferentially associate with PRC2 in differentiating cells. Analysis of the mRNA and protein levels of these factors revealed that their association with PRC2 correlated with their cell state-specific expression. Taken together, we propose that dynamic changes to the PRC2 interactome during differentiation may contribute to directing its activity during cell fate transitions.


Subject(s)
Embryonal Carcinoma Stem Cells/cytology , Pluripotent Stem Cells/cytology , Polycomb Repressive Complex 2/metabolism , Proteomics/methods , Cell Differentiation , Cell Line, Tumor , Embryonal Carcinoma Stem Cells/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Histones/metabolism , Humans , Neoplasm Proteins , Pluripotent Stem Cells/metabolism , Protein Interaction Maps , Transcription Factors
12.
PLoS One ; 10(7): e0132363, 2015.
Article in English | MEDLINE | ID: mdl-26177297

ABSTRACT

Botulinum neurotoxins produced by Clostridium botulinum cause flaccid paralysis by inhibiting neurotransmitter release at peripheral nerve terminals. Previously, we found that neurons derived from the murine P19 embryonal carcinoma cell line exhibited high sensitivity to botulinum neurotoxin type C. In order to prove the utility of P19 cells for the study of the intracellular mechanism of botulinum neurotoxins, ganglioside-knockout neurons were generated by deletion of the gene encoding beta-1,4 N-acetylgalactosaminyltransferase 1 in P19 cells using the clustered regularly interspaced short palindromic repeats combined with Cas9 (CRISPR/Cas9) system. By using this system, knockout cells could be generated more easily than with previous methods. The sensitivity of the generated beta-1,4 N-acetylgalactosaminyltransferase 1-depleted P19 neurons to botulinum neurotoxin type C was decreased considerably, and the exogenous addition of the gangliosides GD1a, GD1b, and GT1b restored the susceptibility of P19 cells to botulinum neurotoxin type C. In particular, addition of a mixture of these three ganglioside more effectively recovered the sensitivity of knockout cells compared to independent addition of GD1a, GD1b, or GT1b. Consequently, the genome-edited P19 cells generated by the CRISPR/Cas9 system were useful for identifying and defining the intracellular molecules involved in the toxic action of botulinum neurotoxins.


Subject(s)
Botulinum Toxins/toxicity , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , N-Acetylgalactosaminyltransferases/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Gangliosides/pharmacology , Mice , Microscopy, Confocal , N-Acetylgalactosaminyltransferases/antagonists & inhibitors , N-Acetylgalactosaminyltransferases/deficiency , N-Acetylgalactosaminyltransferases/metabolism , Proteolysis/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Sialyltransferases/antagonists & inhibitors , Sialyltransferases/genetics , Sialyltransferases/metabolism , Surface Plasmon Resonance , beta-D-Galactoside alpha 2-6-Sialyltransferase , Polypeptide N-acetylgalactosaminyltransferase
13.
J Biochem ; 158(5): 373-84, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26002960

ABSTRACT

The Mediator complex (Mediator) plays key roles in transcription and functions as the nexus for integration of various transcriptional signals. Previously, we screened for Mediator cyclin-dependent kinase (CDK)-interacting factors and identified three proteins related to chromatin regulation. One of them, SUZ12 is required for both stability and activity of Polycomb Repressive Complex 2 (PRC2). PRC2 primarily suppresses gene expression through histone H3 lysine 27 trimethylation, resulting in stem cell maintenance and differentiation; perturbation of this process leads to oncogenesis. Recent work showed that Mediator contributes to the embryonic stem cell state through DNA loop formation, which is strongly associated with chromatin architecture; however, it remains unclear how Mediator regulates gene expression in cooperation with chromatin regulators (i.e. writers, readers and remodelers). We found that Mediator CDKs interact directly with the PRC2 subunit EZH2, as well as SUZ12. Known PRC2 target genes were deregulated by Mediator CDK knockdown during neuronal differentiation, and both Mediator and PRC2 complexes co-occupied the promoters of developmental genes regulated by retinoic acid. Our results provide a mechanistic link between Mediator and PRC2 during neuronal differentiation.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/metabolism , Neurogenesis , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , Transcription, Genetic , Tretinoin/metabolism , Animals , Cell Line , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/enzymology , Embryonal Carcinoma Stem Cells/metabolism , Enhancer of Zeste Homolog 2 Protein , Gene Deletion , Humans , Mice , Neoplasm Proteins , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Polycomb Repressive Complex 2/genetics , Protein Kinases/metabolism , Protein Processing, Post-Translational , RNA Interference , RNA Polymerase II/metabolism , Recombinant Fusion Proteins/metabolism , Transcription Factors
14.
J Mol Neurosci ; 55(3): 684-705, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25189318

ABSTRACT

As members of the proneural basic-helix-loop-helix (bHLH) family of transcription factors, Ascl1 and Neurog2 direct the differentiation of specific populations of neurons at various times and locations within the developing nervous system. In order to characterize the mechanisms employed by these two bHLH factors, we generated stable, doxycycline-inducible lines of P19 embryonic carcinoma cells that express comparable levels of Ascl1 and Neurog2. Upon induction, both Ascl1 and Neurog2 directed morphological and immunocytochemical changes consistent with initiation of neuronal differentiation. Comparison of Ascl1- and Neurog2-regulated genes by microarray analyses showed both shared and distinct transcriptional changes for each bHLH protein. In both Ascl1- and Neurog2-differentiating cells, repression of Oct4 mRNA levels was accompanied by increased Oct4 promoter methylation. However, DNA demethylation was not detected for genes induced by either bHLH protein. Neurog2-induced genes included glutamatergic marker genes while Ascl1-induced genes included GABAergic marker genes. The Neurog2-specific induction of a gene encoding a protein phosphatase inhibitor, Ppp1r14a, was dependent on distinct, canonical E-box sequences within the Ppp1r14a promoter and the nucleotide sequences within these E-boxes were partially responsible for Neurog2-specific regulation. Our results illustrate multiple novel mechanisms by which Ascl1 and Neurog2 regulate gene repression during neuronal differentiation in P19 cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis , Promoter Regions, Genetic , Animals , Cell Line, Tumor , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Octamer Transcription Factor-3/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Transcription, Genetic
15.
Endocrinology ; 156(1): 346-59, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25380237

ABSTRACT

Neonatal gonocytes are direct precursors of spermatogonial stem cells, the cell pool that supports spermatogenesis. Although unipotent in vivo, gonocytes express pluripotency genes common with embryonic stem cells. Previously, we found that all-trans retinoic acid (RA) induced the expression of differentiation markers and a truncated form of platelet-derived growth factor receptor (PDGFR)ß in rat gonocytes, as well as in F9 mouse embryonal carcinoma cells, an embryonic stem cell-surrogate that expresses somatic lineage markers in response to RA. The present study is focused on identifying the signaling pathways involved in RA-induced gonocyte and F9 cell differentiation. Mitogen-activated protein kinase kinase (MEK) 1/2 activation was required during F9 cell differentiation towards somatic lineage, whereas its inhibition potentiated RA-induced Stra8 expression, suggesting that MEK1/2 acts as a lineage specification switch in F9 cells. In both cell types, RA increased the expression of the spermatogonial/premeiotic marker Stra8, which is in line with F9 cells being at a stage before somatic-germline lineage specification. Inhibiting PDGFR kinase activity reduced RA-induced Stra8 expression. Interestingly, RA increased the expression of PDGFRα variant forms in both cell types. Together, these results suggest a potential cross talk between RA and PDGFR signaling pathways in cell differentiation. RA receptor-α inhibition partially reduced RA effects on Stra8 in gonocytes, indicating that RA acts in part via RA receptor-α. RA-induced gonocyte differentiation was significantly reduced by inhibiting SRC (v-src avian sarcoma [Schmidt-Ruppin A-2] viral oncogene) and JAK2/STAT5 (Janus kinase 2/signal transducer and activator of transcription 5) activities, implying that these signaling molecules play a role in gonocyte differentiation. These results suggest that gonocyte and F9 cell differentiation is regulated via cross talk between RA and PDGFRs using different downstream pathways.


Subject(s)
Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Tretinoin/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/physiology , Animals , Animals, Newborn , Cell Differentiation , Cell Line , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Keratolytic Agents/pharmacology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Platelet-Derived Growth Factor/genetics , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor beta/genetics , Testis/cytology , Testis/growth & development
16.
Exp Cell Res ; 330(2): 442-450, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25447206

ABSTRACT

Transcription factor FOXM1 plays a critical role in maintenance of stem cell pluripotency through stimulating the transcription of pluripotency-related genes in mouse pluripotent stem cells. In this study, we have found that the repression of FOXM1 expression is mediated by FOXM1 3'UTR during retinoic acid-induced differentiation of human pluripotent NT2/D1 embryonal carcinoma cells. FOXM1 3'UTR contains a microRNA response element (MRE) for miR-134, which has been shown to attenuate the expression of pluripotency-related genes post-transcriptionally during mouse embryonic stem cell differentiation. We have determined that miR-134 is induced during RA-induced differentiation of NT2/D1 cells and the overexpression of miR-134 represses the expression of FOXM1 protein but not FOXM1 mRNA. Furthermore, the expression of OCT4 is diminished by FOXM1 knockdown and the OCT4 promoter is regulated directly by FOXM1, suggesting that FOXM1 is required for maintaining the expression of OCT4 in NT2/D1 cells. Together, our results suggest that FOXM1 is essential for human pluripotent stem cells and miR-134 attenuates its expression during differentiation.


Subject(s)
Carcinoma, Embryonal/pathology , Embryonal Carcinoma Stem Cells/cytology , Forkhead Transcription Factors/genetics , MicroRNAs/genetics , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/cytology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Embryonal/genetics , Cell Differentiation , Cell Line, Tumor , Forkhead Box Protein M1 , Forkhead Transcription Factors/biosynthesis , HEK293 Cells , Humans , Mice , Octamer Transcription Factor-3/biosynthesis , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Response Elements/genetics , Tretinoin/pharmacology
17.
PLoS One ; 9(11): e111637, 2014.
Article in English | MEDLINE | ID: mdl-25369332

ABSTRACT

Human embryonic stem cells and human embryonal carcinoma cells have been studied extensively with respect to the transcription factors (OCT4, SOX2 and NANOG), epigenetic modulators and associated signalling pathways that either promote self-renewal or induce differentiation in these cells. The ACTIVIN/NODAL axis (SMAD2/3) of the TGFß signalling pathway coupled with FGF signalling maintains self-renewal in these cells, whilst the BMP (SMAD1,5,8) axis promotes differentiation. Here we show that miR-27, a somatic-enriched miRNA, is activated upon RNAi-mediated suppression of OCT4 function in human embryonic stem cells. We further demonstrate that miR-27 negatively regulates the expression of the pluripotency-associated ACTIVIN/NODAL axis (SMAD2/3) of the TGFß signalling pathway by targeting ACVR2A, TGFßR1 and SMAD2. Additionally, we have identified a number of pluripotency-associated genes such as NANOG, LIN28, POLR3G and NR5A2 as novel miR-27 targets. Transcriptome analysis revealed that miR-27 over-expression in human embryonal carcinoma cells leads indeed to a significant up-regulation of genes involved in developmental pathways such as TGFß- and WNT-signalling.


Subject(s)
Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Activin Receptors, Type II/genetics , Cell Differentiation , Cell Line , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , MicroRNAs/metabolism , Octamer Transcription Factor-3/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Smad2 Protein/genetics , Up-Regulation
18.
Annu Rev Cell Dev Biol ; 30: 647-75, 2014.
Article in English | MEDLINE | ID: mdl-25288119

ABSTRACT

Mouse embryonic stem (ES) cells perpetuate in vitro the broad developmental potential of naïve founder cells in the preimplantation embryo. ES cells self-renew relentlessly in culture but can reenter embryonic development seamlessly, differentiating on schedule to form all elements of the fetus. Here we review the properties of these remarkable cells. Arising from the stability, homogeneity, and equipotency of ES cells, we consider the concept of a pluripotent ground state. We evaluate the authenticity of ES cells in relation to cells in the embryo and examine their utility for dissecting mechanisms that confer pluripotency and that execute fate choice. We summarize current knowledge of the transcription factor circuitry that governs the ES cell state and discuss the opportunity to expose molecular logic further through iterative computational modeling and experimentation. Finally, we present a perspective on unresolved questions, including the challenge of deriving ground state pluripotent stem cells from non-rodent species.


Subject(s)
Embryonic Stem Cells/cytology , Animals , Asymmetric Cell Division , Blastocyst/cytology , Cell Culture Techniques , Cell Lineage , Cells, Cultured , Cellular Reprogramming , Coculture Techniques , Culture Media , Culture Media, Serum-Free , Embryonal Carcinoma Stem Cells/cytology , Embryonic Stem Cells/physiology , Fibroblasts/cytology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, Reporter , Germ Layers/cytology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/physiology , Leukemia Inhibitory Factor/physiology , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Transcription Factors/pharmacology , Transcription Factors/physiology
19.
Dev Neurobiol ; 74(12): 1243-54, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24909558

ABSTRACT

Dickkopf-3 (Dkk-3) and Dkkl-1 (Soggy) are secreted proteins of poorly understood function that are highly expressed in subsets of neurons in the brain. To explore their potential roles during neuronal development, we examined their expression in Ntera-2 (NT2) human embryonal carcinoma cells, which differentiate into neurons upon treatment with retinoic acid (RA). RA treatment increased the mRNA and protein levels of Dkk-3 but not of Dkkl-1. Ectopic expression of both Dkk-3 and Dkkl-1 induced apoptosis in NT2 cells. Gene silencing of Dkk-3 did not affect NT2 cell growth or differentiation but altered their response to RA in suspension cultures. RA treatment of NT2 cells cultured in suspension resulted in morphological changes that led to cell attachment and flattening out of cell aggregates. Although there were no significant differences in the expression levels of cell adhesion molecules in control and Dkk-3-silenced cells, this morphological response was not observed in Dkk-3-silenced cells. These findings suggest that Dkk-3 plays a role in the regulation of cell interactions during RA-induced neuronal differentiation.


Subject(s)
Embryonal Carcinoma Stem Cells/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Mitosis Modulators/pharmacology , Neurogenesis/physiology , Tretinoin/pharmacology , Adaptor Proteins, Signal Transducing , Apoptosis/physiology , Blotting, Western , Caspase 3/metabolism , Caspase 7/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Tumor , Chemokines , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/drug effects , Gene Silencing , Humans , Intercellular Signaling Peptides and Proteins/genetics , Neurogenesis/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Polymerase Chain Reaction , RNA, Messenger/metabolism , Transfection
20.
Biochem Biophys Res Commun ; 450(2): 984-90, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24971534

ABSTRACT

The epithelium-specific ETS transcription factor-1 (ESE-1) is physiologically important in the pathogenesis of various diseases. Recently, OCT4, a transcription factor involved in stem cell pluripotency, has been implicated in tumorigenesis. In this study, we invested the molecular mechanism by which ESE-1 regulates transcription of OCT4 in NCCIT human embryonic carcinoma cells. Real-time PCR analysis revealed that OCT4 levels were high in undifferentiated NCCIT cells but significantly decreased upon retinoic acid-mediated differentiation, concomitant with up-regulation of ESE-1 expression. OCT4 mRNA level rose following shRNA-mediated knockdown of ESE-1, but declined when ESE-1 was overexpressed, suggesting that the expression levels of OCT4 and ESE-1 may be coordinated in an opposite manner. Promoter-reporter assays revealed that induced OCT4 promoter activity in NCCIT cells was significantly down-regulated by ESE-1 overexpression in a dose-dependent manner. The inhibitory effect of ESE-1 on OCT4 promoter activity was relieved by co-expression of an ESE-1 mutant lacking the transactivation domain, but not by mutants lacking other domains. Serial deletion and site-directed mutagenesis of the OCT4 promoter revealed that a potential ETS binding site (EBS) is present in the conserved region 2 (CR2). ESE-1 interacted with the EBS element in CR2 and enrichment of CR2 significantly increased upon RA-mediated differentiation of NCCIT cells, suggesting that this binding is likely to be involved in ESE-1-mediated repression of OCT4 promoter activity upon differentiation. Taken together, the results of this study reveal the molecular details of the mechanism by which the oncogenic factor ESE-1 regulates expression of the stem cell transcription factor OCT4 in pluripotent NCCIT cells.


Subject(s)
DNA-Binding Proteins/metabolism , Embryonal Carcinoma Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/metabolism , Cell Differentiation , DNA-Binding Proteins/genetics , Embryonal Carcinoma Stem Cells/cytology , Gene Knockdown Techniques , Humans , Mutation , Pluripotent Stem Cells/cytology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...