Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(1)2021 Dec 21.
Article in English | MEDLINE | ID: mdl-35008480

ABSTRACT

The pluripotent transcription factor NANOG is essential for maintaining embryonic stem cells and driving tumorigenesis. We previously showed that PKC activity is involved in the regulation of NANOG expression. To explore the possible involvement of microRNAs in regulating the expression of key pluripotency factors, we performed a genome-wide analysis of microRNA expression in the embryonal carcinoma cell line NT2/D1 in the presence of the PKC activator, PMA. We found that MIR630 was significantly upregulated in PMA-treated cells. Experimentally, we showed that transfection of MIR630 mimic into embryonal carcinoma cell lines directly targeted the 3'UTR of OCT4, SOX2, and NANOG and markedly suppressed their expression. RNAhybrid and RNA22 algorithms were used to predict miRNA target sites in the NANOG 3'UTR, four possible target sites of MIR630 were identified. To examine the functional interaction between MIR630 and NANOG mRNA, the predicted MIR630 target sites in the NANOG 3'UTR were deleted and the activity of the reporters were compared. After targeted mutation of the predicted MIR630 target sites, the MIR630 mimic inhibited NANOG significantly less than the wild-type reporters. It is worth noting that mutation of a single putative binding site in the 3'UTR of NANOG did not completely abolish MIR630-mediated suppression, suggesting that MIR630 in the NANOG 3'UTR may have multiple binding sites and act together to maximally repress NANOG expression. Interestingly, MIR630 mimics significantly downregulated NANOG gene transcription. Exogenous expression of OCT4, SOX2, and NANOG lacking the 3'UTR almost completely rescued the reduced transcriptional activity of MIR630. MIR630 mediated the expression of differentiation markers in NT2/D1 cells, suggesting that MIR630 leads to the differentiation of NT2/D1 cell. Our findings show that MIR630 represses NANOG through transcriptional and post-transcriptional regulation, suggesting a direct link between core pluripotency factors and MIR630.


Subject(s)
Carcinoma, Embryonal/genetics , Embryonal Carcinoma Stem Cells/physiology , MicroRNAs/genetics , Nanog Homeobox Protein/genetics , RNA Interference/physiology , Transcription, Genetic/genetics , 3' Untranslated Regions/genetics , Binding Sites/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Embryonic Stem Cells/physiology , Humans , Mutation/genetics , Up-Regulation/genetics
2.
J Ethnopharmacol ; 246: 112214, 2020 Jan 10.
Article in English | MEDLINE | ID: mdl-31491437

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Medicinal herb Cichorium intybus L. (chicory) has been used traditionally for the treatment of various diseases, including diabetes. One of the promising therapeutic options to treat diabetes is replacing the degenerative pancreatic ß cells by stem cell-derived IPCs (insulin-producing cells). AIM OF THE STUDY: By the combination of cell therapy as a modern approach and traditional medicine, the current study was designed to evaluate the effects of chicory leaf extract (LE) on the differentiation potential of P19 EC cells (an embryonal carcinoma stem cell line) into IPCs. MATERIALS AND METHODS: The plant (voucher no. 4567) were collected and deposited in the herbarium of Shahrekord University. In vitro experiments were designed to compare the effects of various concentrations of LE on the differentiation potential of P19 EC cells. RESULTS: The differentiated cells showed morphological characteristics of pancreatic ß cells. They could also synthesized and secreted insulin when exposed to glucose. Moreover, the cells expressed specific proteins and genes of mature pancreatic ß cells. CONCLUSIONS: In conclusion, LE as a natural herbal extract was efficiently able to induce the differentiation of P19 EC cells into the clusters similar to pancreatic islets with the molecular, cellular and functional characteristics of mature ß cells.


Subject(s)
Cell Differentiation/drug effects , Cichorium intybus , Embryonal Carcinoma Stem Cells/drug effects , Insulin/metabolism , Plant Extracts/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Embryonal Carcinoma Stem Cells/physiology , Insulin-Secreting Cells/metabolism , Mice , Phytochemicals/analysis , Phytochemicals/pharmacology , Plant Extracts/chemistry , Plant Leaves
3.
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
4.
Cell Death Dis ; 6: e1960, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26539909

ABSTRACT

Long non-coding RNAs (lncRNAs), which are extensively transcribed from the genome, have been proposed to be key regulators of diverse biological processes. However, little is known about the role of lncRNAs in regulating spermatogenesis in human males. Here, using microarray technology, we show altered expression of lncRNAs in the testes of infertile men with maturation arrest (MA) or hypospermatogenesis (Hypo), with 757 and 2370 differentially down-regulated and 475 and 163 up-regulated lncRNAs in MA and Hypo, respectively. These findings were confirmed by quantitative real-time PCR (qRT-PCR) assays on select lncRNAs, including HOTTIP, imsrna320, imsrna292 and NLC1-C (narcolepsy candidate-region 1 genes). Interestingly, NLC1-C, also known as long intergenic non-protein-coding RNA162 (LINC00162), was down-regulated in the cytoplasm and accumulated in the nucleus of spermatogonia and primary spermatocytes in the testes of infertile men with mixed patterns of MA compared with normal control. The accumulation of NLC1-C in the nucleus repressed miR-320a and miR-383 transcript and promoted testicular embryonal carcinoma cell proliferation by binding to Nucleolin. Here, we define a novel mechanism by which lncRNAs modulate miRNA expression at the transcriptional level by binding to RNA-binding proteins to regulate human spermatogenesis.


Subject(s)
Carcinoma, Embryonal/metabolism , Embryonal Carcinoma Stem Cells/physiology , Infertility, Male/metabolism , MicroRNAs/metabolism , Neoplasms, Germ Cell and Embryonal/metabolism , RNA, Long Noncoding/metabolism , Testicular Neoplasms/metabolism , Adult , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Case-Control Studies , Cell Proliferation/genetics , Down-Regulation , Embryonal Carcinoma Stem Cells/metabolism , Humans , Infertility, Male/genetics , Infertility, Male/pathology , Male , MicroRNAs/genetics , Middle Aged , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Phosphoproteins/metabolism , RNA, Long Noncoding/genetics , RNA-Binding Proteins/metabolism , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology , Young Adult , Nucleolin
5.
Altern Lab Anim ; 43(1): 9-18, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25802994

ABSTRACT

Serum is generally regarded as an essential component of many eukaryotic cell culture media, despite the fact that serum composition varies greatly and may be the source of a wide range of artefacts. The objective of this study was to assess serum-free growth conditions for the human embryonal carcinoma cell line, NT2/D1. These cells greatly resemble embryonic stem cells. In the presence of retinoic acid (RA), NT2/D1 cells irreversibly differentiate along the neuronal lineage. We have previously shown that the early phases of neural induction of these cells by RA involve the up-regulation of SOX3 gene expression. Our goal was to compare RA-induced differentiation of NT2/D1 cells in serum-containing and serum-free media, by using SOX3 protein levels as a marker of differentiation. We found that NT2/D1 cells can be successfully grown under serum-free conditions, and that the presence or absence of serum does not affect the level of SOX3 protein after a 48-hour RA induction. However, six days of RA treatment resulted in a marked increase in SOX3 protein levels in serum-free media compared to serum-containing media, indicating that serum might have an inhibitory effect on the expression of this neural differentiation marker. This finding is important for both basic and translational studies that hope to exploit cell culture conditions that are free of animal-derived products.


Subject(s)
Cell Differentiation/drug effects , Culture Media, Serum-Free , Embryonal Carcinoma Stem Cells/physiology , Neurons/cytology , Neurons/physiology , Cell Line , Embryonal Carcinoma Stem Cells/drug effects , Gene Expression Regulation , Humans , Neurons/drug effects , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Tretinoin/pharmacology
6.
Invest New Drugs ; 33(1): 64-74, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25404486

ABSTRACT

Cancer stem cells (CSCs) are potential targets for innovative anticancer therapies that involve natural products with potential chemopreventive effects. We therefore analyzed the antineoplastic activity of rooperol, the aglycone of the plant-derived compound hypoxoside, on a model of Oct4-expressing cancer stem-like cell, i.e. the human embryonal carcinoma (EC) cell NT2/D1. Rooperol selectively inhibited the proliferation of NT2/D1 cells in a concentration-dependent manner and had no effect on either normal embryonic fibroblasts which are more restrictive pluripotent stem cells or on NCCIT p53-mutant EC cells. Accordingly, rooperol only eliminates colon carcinoma cells expressing p53. Rooperol treatment triggered cell death on NT2/D1 cells through the alteration of mitochondrial membrane potential and production of reactive oxygen species (ROS). Rooperol-induced apoptosis was associated with activation of p53 and concentration-dependent changes of the expression levels of both caspase 3 and poly ADP ribose polymerase type 1 cleaved subunits. These modifications were accompanied by a downregulation of Oct4 and its two partners involved in the maintenance of cell pluripotency and self-renewal, Nanog and Sox2.Treatment with intracellular membrane permeant O2 (-) scavengers prevented rooperol-induced apoptosis and upregulation of the expression of p53 and active caspase-3. Our findings indicate that rooperol mediates its growth inhibitory effects on CSCs via a mitochondrial redox-sensitive mechanism. We propose that abrogating the expression of the stemness regulators is a prerequisite for rooperol to fully exert its pro-apoptotic properties on wild-type p53-bearing CSCs.


Subject(s)
Antineoplastic Agents/pharmacology , Catechols/pharmacology , Embryonal Carcinoma Stem Cells/drug effects , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Alkynes , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/physiology , Glucosides , Humans , Membrane Potential, Mitochondrial/drug effects , Teratocarcinoma
7.
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
8.
Mol Cell Biol ; 34(2): 158-79, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24190971

ABSTRACT

LSD1 is essential for the maintenance of pluripotency of embryonic stem (ES) or embryonic carcinoma/teratocarcinoma (EC) cells. We have previously developed novel LSD1 inhibitors that selectively inhibit ES/EC cells. However, the critical targets of LSD1 remain unclear. Here, we found that LSD1 interacts with histone deacetylase 1 (HDAC1) to regulate the proliferation of ES/EC cells through acetylation of histone H4 at lysine 16 (H4K16), which we show is a critical substrate of HDAC1. The LSD1 demethylase and HDAC1 deacetylase activities were both inactivated if one of them in the complex was chemically inhibited in ES/EC cells or in reconstituted protein complexes. Loss of HDAC1 phenocopied the selective growth-inhibitory effects and increased the levels of H3K4 methylation and H4K16 acetylation of LSD1 inactivation on ES/EC cells. Reduction of acetylated H4K16 by ablation of the acetyltransferase males absent on the first (MOF) is sufficient to rescue the growth inhibition induced by LSD1 inactivation. While LSD1 or HDAC1 inactivation caused the downregulation of Sox2 and Oct4 and induction of differentiation genes, such as FOXA2 or BMP2, depletion of MOF restored the levels of Sox2, Oct4, and FoxA2 in LSD1-deficient cells. Our studies reveal a novel mechanism by which LSD1 acts through the HDAC1- and MOF-mediated regulation of H4K16 acetylation to maintain the pluripotency of ES/EC cells.


Subject(s)
Embryonal Carcinoma Stem Cells/physiology , Histone Deacetylase 1/physiology , Histone Demethylases/physiology , Histones/metabolism , Protein Processing, Post-Translational , Acetylation , Animals , Cell Proliferation , Co-Repressor Proteins , Embryonic Stem Cells/physiology , Epigenesis, Genetic , G1 Phase Cell Cycle Checkpoints , Gene Expression , HCT116 Cells , HeLa Cells , Histone Acetyltransferases/metabolism , Humans , Lysine/metabolism , Methylation , Mice , NIH 3T3 Cells , Nerve Tissue Proteins/metabolism , Octamer Transcription Factor-3/metabolism , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Sirtuin 1/metabolism
9.
Stem Cells ; 32(1): 126-34, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24105929

ABSTRACT

Long noncoding RNAs (lncRNAs) have emerged as new regulators of stem cell pluripotency and tumorigenesis. The SOX2 gene, a master regulator of pluripotency, is embedded within the third intron of a lncRNA known as SOX2 overlapping transcript (SOX2OT). SOX2OT has been suspected to participate in regulation of SOX2 expression and/or other related processes; nevertheless, its potential involvement in tumor initiation and/or progression is unclear. Here, we have evaluated a possible correlation between expression patterns of SOX2OT and those of master regulators of pluripotency, SOX2 and OCT4, in esophageal squamous cell carcinoma (ESCC) tissue samples. We have also examined its potential function in the human embryonic carcinoma stem cell line, NTERA2 (NT2), which highly expresses SOX2OT, SOX2, and OCT4. Our data revealed a significant coupregulation of SOX2OT along with SOX2 and OCT4 in tumor samples, compared to the non-tumor tissues obtained from the margin of same tumors. We also identified two novel splice variants of SOX2OT (SOX2OT-S1 and SOX2OT-S2) which coupregulated with SOX2 and OCT4 in ESCCs. Suppressing SOX2OT variants caused a profound alteration in cell cycle distribution, including a 5.9 and 6.9 time increase in sub-G1 phase of cell cycle for SOX2OT-S1 and SOX2OT-S2, respectively. The expression of all variants was significantly diminished, upon the induction of neural differentiation in NT2 cells, suggesting their potential functional links to the undifferentiated state of the cells. Our data suggest a part for SOX2OT spliced variants in tumor initiation and/or progression as well as regulating pluripotent state of stem cells.


Subject(s)
Carcinoma, Squamous Cell/genetics , Embryonal Carcinoma Stem Cells/physiology , Esophageal Neoplasms/genetics , Octamer Transcription Factor-3/genetics , SOXB1 Transcription Factors/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Culture Techniques , Cell Differentiation/genetics , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Humans , Octamer Transcription Factor-3/biosynthesis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , Protein Isoforms , RNA Interference , RNA Splicing , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , SOXB1 Transcription Factors/biosynthesis , Up-Regulation
10.
Mol Cell Biochem ; 376(1-2): 145-50, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23361361

ABSTRACT

Members of the SOX (SRY box) family proteins play critical roles in multiple aspects of development. SRY, as a founder member of SOX family, has been long believed to be involved in the development of sexual gonads by triggering signaling cascades which lead to the formation of testis or ovary from bipotential gonads. However, less is known about other potential regulatory roles of SRY in the development and differentiation. In order to gain further insight into the possible roles of SRY during development, we looked into possible SRY-regulated genes and their levels of expression in a human embryonic carcinoma cell line, named NTera2, before and after induction of differentiation. For this respect, SRY incorporation on the regulatory regions of two groups of genes including OCT4, NANOG, and SOX2 as pluripotency marker genes, and NESTIN and PAX6 as differentiation marker genes were evaluated quantitatively. Chromatin immunoprecipitation using SRY antibody was performed on chromatin extract of a human embryonic carcinoma cell line, NT2/NTERA-2, before and after onset of differentiation. The results showed that incorporation of SRY in both groups of genes was increased after induction of differentiation. Besides, lower expression of OCT4, SOX2, and NANOG and higher expression of PAX6 and NESTIN genes in differentiated cells suggest that SRY may act as a transcription repressor for pluripotency-associated genes and as a transcription activator for differentiation-related genes.


Subject(s)
Cell Differentiation/genetics , Embryonal Carcinoma Stem Cells/physiology , Gene Expression Regulation, Developmental , Sex-Determining Region Y Protein/genetics , Tretinoin/pharmacology , Cell Differentiation/drug effects , Chromatin Immunoprecipitation , Embryonal Carcinoma Stem Cells/drug effects , Eye Proteins/genetics , Gene Expression Regulation, Developmental/drug effects , Genetic Markers , Homeodomain Proteins/genetics , Humans , Intermediate Filament Proteins/genetics , Nanog Homeobox Protein , Nerve Tissue Proteins/genetics , Nestin , Octamer Transcription Factor-3/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Pluripotent Stem Cells/physiology , Regulatory Sequences, Nucleic Acid , Repressor Proteins/genetics , SOXB1 Transcription Factors/genetics
11.
Int J Dev Neurosci ; 30(2): 91-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22248690

ABSTRACT

Embryonic carcinoma cells are widely used models for studying the mechanisms of proliferation and differentiation occurring during early embryogenesis. We have now investigated how down-regulation of P2X2 and P2X7 receptor expression by RNA interference (RNAi) affects neural differentiation and phenotype specification of P19 embryonal carcinoma cells. Wild-type P19 embryonal carcinoma cells or cells stably expressing shRNAs targeting P2X2 or P2X7 receptor expression were induced to differentiate into neurons and glial cells in the presence of retinoic acid. Silencing of P2X2 receptor expression along differentiation promoted cell proliferation and an increase in the percentage of cells expressing glial-specific GFAP, while the presence of beta-3 tubulin-positive cells diminished at the same time. Proliferation induction in the presence of stable anti-P2X2 receptor RNAi points at a mechanism where glial proliferation is favored over growth arrest of progenitor cells which would allow neuronal maturation. Differently from the P2X2 receptor, inhibition of P2X7 receptor expression during neural differentiation of P19 cells resulted in a decrease in cell proliferation and GFAP expression, suggesting the need of functional P2X7 receptors for the progress of gliogenesis. The results obtained in this study indicate the importance of purinergic signaling for cell fate determination during neural differentiation, with P2X2 and P2X7 receptors promoting neurogenesis and gliogenesis, respectively. The shRNAs down-regulating P2X2 or P2X7 receptor gene expression, developed during this work, present useful tools for studying mechanisms of neural differentiation in other stem cell models.


Subject(s)
Embryonal Carcinoma Stem Cells/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , Neuroglia/cytology , Neurons/cytology , Receptors, Purinergic P2X2/physiology , Receptors, Purinergic P2X7/physiology , Tretinoin/physiology , Animals , Cell Differentiation/genetics , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/physiology , Mice , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Neurogenesis/genetics , Neuroglia/metabolism , Neuroglia/physiology , Neurons/metabolism , Neurons/physiology , RNA Interference/physiology , Receptors, Purinergic P2X2/genetics , Receptors, Purinergic P2X7/genetics , Signal Transduction/genetics
12.
Stem Cells ; 30(3): 452-60, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22162396

ABSTRACT

Human embryonic stem cells (hESC) have a unique capacity to self-renew and differentiate into all the cell types found in human body. Although the transcriptional regulators of pluripotency are well studied, the role of cytoplasmic regulators is still poorly characterized. Here, we report a new stem cell-specific RNA-binding protein L1TD1 (ECAT11, FLJ10884) required for hESC self-renewal and cancer cell proliferation. Depletion of L1TD1 results in immediate downregulation of OCT4 and NANOG. Furthermore, we demonstrate that OCT4, SOX2, and NANOG all bind to the promoter of L1TD1. Moreover, L1TD1 is highly expressed in seminomas, and depletion of L1TD1 in these cancer cells influences self-renewal and proliferation. We show that L1TD1 colocalizes and interacts with LIN28 via RNA and directly with RNA helicase A (RHA). LIN28 has been reported to regulate translation of OCT4 in complex with RHA. Thus, we hypothesize that L1TD1 is part of the L1TD1-RHA-LIN28 complex that could influence levels of OCT4. Our results strongly suggest that L1TD1 has an important role in the regulation of stemness.


Subject(s)
Cell Proliferation , DEAD-box RNA Helicases/metabolism , Embryonal Carcinoma Stem Cells/physiology , Neoplasm Proteins/metabolism , Proteins/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Argonaute Proteins/metabolism , Cells, Cultured , Embryonal Carcinoma Stem Cells/metabolism , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Gene Expression Regulation , Humans , Promoter Regions, Genetic , Protein Binding , Proteins/genetics , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/genetics , Seminoma/metabolism , Seminoma/pathology
13.
Int Rev Cell Mol Biol ; 291: 155-90, 2011.
Article in English | MEDLINE | ID: mdl-22017976

ABSTRACT

This review is focused on recent advances in fusion-based reprogramming of cells of different pluripotent statuses or lineage origins. Recent findings are discussed from standpoints of both the developmental potency of hybrid cells generated by fusion of pluripotent embryonic stem (ES) cells, embryonal carcinoma (EC) cells, and somatic cells and epigenetic mechanisms and other aspects involved in the reprogramming process. Complete reprogramming occurs at least 5-7 days after fusion and includes at least two steps. (i) initiation at the heterokaryon stage and choice of the direction of reprogramming using an "all-or-none principle" to establish the dominance of one parental genome and (ii) "fixation" of the newly acquired expression profile by epigenetic mechanisms. The first step is realized without cell division, whereas the second requires cell proliferation. Reprogramming in hybrid cells is rapid and complete. Thus, cell fusion is a powerful tool for reprogramming.


Subject(s)
Cell Fusion , Cellular Reprogramming , Pluripotent Stem Cells/physiology , Animals , Cell Cycle , Cell Lineage , Cell Proliferation , Chromosomes , Embryonal Carcinoma Stem Cells/physiology , Epigenesis, Genetic , Gene Expression Profiling , Gene Silencing , Genome-Wide Association Study , Humans , Phenotype
14.
Toxicol Lett ; 207(2): 149-58, 2011 Nov 30.
Article in English | MEDLINE | ID: mdl-21925251

ABSTRACT

Due to their pluripotent nature, embryonic carcinoma (EC) cell lines are useful for the study of basic and applied aspects of medical therapeutics, disease management and environmental mutagenesis. We evaluated the teratogenic like effects of inorganic arsenic during the early stages of cellular development in NCCIT cells, a type of human EC cells. Using matrix assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) and MetaCore pathway analysis software (GeneGo), the proteomic expression profiles of NCCIT cells after either short- or long-term sodium arsenite (NaAsO(2)) treatment and of NCCIT cell-derived embryoid bodies (EBs) after short-term NaAsO(2) treatment were studied to determine the toxic effects on the process of normal growth and differentiation. The protein expression profiles of the NaAsO(2)-treated NCCIT cells and EBs were compared with that of untreated NCCIT cells. Short-term NaAsO(2) treatment resulted in the down-regulation of most proteins, with heat shock 90-kDa protein (HSP90) and valosin-containing protein (VCP) being significantly downregulated. Long-term NaAsO(2) treatment caused marked up-regulation of B23/nucleophosmin, glucose regulated protein 78-kDa (GRP78), unc-51-like kinase 3 (ULK3), toll-like receptor 6 precursor (TLR6) and mitogen-activated protein kinase 7 isoform A (MAP3K7). NaAsO(2) treatment of the NCCIT cell-derived EBs resulted in the down-regulation of several tumor-suppressor proteins. Taken together, these data suggest that a wide spectrum of cellular responses is induced to cope with chronic non-lethal toxic stresses in NCCIT cells.


Subject(s)
Arsenites/pharmacology , Embryonal Carcinoma Stem Cells/drug effects , Protein Biosynthesis/drug effects , Sodium Compounds/pharmacology , Arsenites/administration & dosage , Biosynthetic Pathways/drug effects , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Embryoid Bodies/drug effects , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/physiology , Endoplasmic Reticulum Chaperone BiP , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Sodium Compounds/administration & dosage , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
Prague Med Rep ; 111(4): 289-99, 2010.
Article in English | MEDLINE | ID: mdl-21189168

ABSTRACT

The aim of our study was to characterize mouse embryonal carcinoma (EC) cells P19 in different stages of retinoic acid induced neurodifferentiation by two methods, immunocytochemistry and RT qPCR. The characterization of the cells is crucial before any transplantation into any model, e.g. in our case into the mouse brain with the aim to treat a neurodegenerative disease. Specific protein markers (MAP-2, OCT-4, FORSE-1) were detected by immunocytochemistry in the cell cultures. The mRNA expression levels of PAX-6, MASH-1, Brachyury, GATA-4 and AFP were determined by RT qPCR method. HPRT was used as a housekeeping gene. The degree of differentiation can be characterized by expression of analyzed genes. The presence of OCT-4 and FORSE-1 proteins in undifferentiated pluripotent cells and the presence of dendrite specific MAP-2 in neuroprogenitors was detected. The expression levels of PAX-6 and MASH-1 increased and expression of Brachyury decreased during the neurodifferentiation process. The expression levels of GATA-4 and AFP were the highest after induction of differentiation with retinoic acid. Detailed characterization of cells before transplantation experiments can contribute to better understanding of their effect.


Subject(s)
Cell Differentiation/drug effects , Cell Differentiation/genetics , Tretinoin/pharmacology , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Embryonal Carcinoma Stem Cells/physiology , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
16.
Stem Cells Dev ; 19(9): 1365-74, 2010 Sep.
Article in English | MEDLINE | ID: mdl-19916800

ABSTRACT

Transcription factor FoxA1 plays a critical role during embryonic development and is activated during retinoic acid (RA)-induced neural differentiation of pluripotent P19 embryonal carcinoma cells at the early stage, which is marked by decreased expression of Nanog and increased expression of neural stem cell marker Nestin. To further understand how FoxA1 mediates neural differentiation, we have overexpressed FoxA1 through an adenovirus vector in P19 cells and identified that early neurogenesis-related sonic hedgehog (Shh) gene is activated directly by FoxA1. Knockdown of FoxA1 expression during P19 cell neural differentiation results in prevention of Shh and Nestin induction. FoxA1 binds to Shh promoter at -486 to -462 bp region and activates the promoter in cotransfection assays. Furthermore, overexpression of FoxA1 alone in P19 cells stimulates expression of Nestin and results in decreased protein levels of Nanog. During RA-induced P19 cell differentiation, elevated levels of FoxA1 increase the population of neurons, evidenced by stimulated expression of neuron-specific Neurofilament-1 and Tubulin betaIII. Together, our results suggest a critical involvement of FoxA1 in stimulating neural differentiation of pluripotent stem cells at early stages.


Subject(s)
Cell Differentiation/genetics , Embryonal Carcinoma Stem Cells/metabolism , Hepatocyte Nuclear Factor 3-alpha/physiology , Neurogenesis/genetics , Animals , BALB 3T3 Cells , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Embryonal Carcinoma Stem Cells/pathology , Embryonal Carcinoma Stem Cells/physiology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Mice , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , Promoter Regions, Genetic/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , Tretinoin/pharmacology , Up-Regulation/physiology
17.
PLoS Biol ; 7(7): e1000149, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19582141

ABSTRACT

There is evidence that pluripotency of mouse embryonic stem (ES) cells is associated with the activity of a network of transcription factors with Sox2, Oct4, and Nanog at the core. Using fluorescent reporters for the expression of Nanog, we observed that a population of ES cells is best described by a dynamic distribution of Nanog expression characterized by two peaks defined by high (HN) and low (LN) Nanog expression. Typically, the LN state is 5%-20% of the total population, depending on the culture conditions. Modelling of the activity of Nanog reveals that a simple network of Oct4/Sox2 and Nanog activity can account for the observed distribution and its properties as long as the transcriptional activity is tuned by transcriptional noise. The model also predicts that the LN state is unstable, something that is born out experimentally. While in this state, cells can differentiate. We suggest that transcriptional fluctuations in Nanog expression are an essential element of the pluripotent state and that the function of Sox2, Oct4, and Nanog is to act as a network that promotes and maintains transcriptional noise to interfere with the differentiation signals.


Subject(s)
Embryonal Carcinoma Stem Cells/physiology , Embryonic Stem Cells/physiology , Gene Expression Regulation , Homeodomain Proteins/genetics , Pluripotent Stem Cells/physiology , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Cell Lineage , Embryonal Carcinoma Stem Cells/cytology , Embryonic Stem Cells/cytology , Flow Cytometry , Gene Expression Profiling , Gene Regulatory Networks , Genes, Reporter , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Mice , Models, Biological , Nanog Homeobox Protein , Octamer Transcription Factor-3/chemistry , Octamer Transcription Factor-3/metabolism , Phenotype , Pluripotent Stem Cells/cytology , SOXB1 Transcription Factors/chemistry , SOXB1 Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription, Genetic , Transgenes , Tumor Cells, Cultured
18.
J Proteome Res ; 8(4): 1765-81, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19275242

ABSTRACT

Cell-cell interactions play a crucial role during embryogenesis and are enhanced during cell aggregation. P19 mouse embryonic carcinoma cells can differentiate into neural cells by the addition of retinoic acid (RA) or by overexpression of the Wnt1 gene, with both processes dependent on cell aggregation. To identify molecules involved in the cell aggregation process, two-dimensional gel electrophoresis (2DE) was used to establish the cell aggregation-associated protein profiles. MALDI-TOF/TOF was used to identify 71 protein spots with differential expression patterns. Among these spots, 54 were differentially expressed in both P19 and Wnt1-overexpressing P19 (Wnt1/P19) cell aggregates, with 42 proteins up-regulated and 12 proteins down-regulated. The other 17 spots were differentially expressed only in Wnt1/P19 cells. The expression patterns of 5 cell aggregation-associated proteins, N-myc downstream-regulated gene 1 (NDRG1), 14-3-3 epsilon, 14-3-3 gamma, acid calponin and cell division control protein 2 homologue (Cdc2), were confirmed by immunoblot and RT-PCR. To further investigate the relationship between cell aggregation and neural differentiation, NDRG1 expression was inhibited by RNA interference during P19 cell aggregation. Silencing of NDRG1 reduced the size of cell aggregates and the expression of N-cadherin, and it also impaired the RA-induced P19 cell neural differentiation. In conclusion, this study provides new clues for the possible mechanism underlying cell aggregation during pluripotent stem cell neural differentiation.


Subject(s)
Cell Differentiation/physiology , Embryonal Carcinoma Stem Cells/physiology , Neurogenesis/physiology , Proteome/metabolism , Amino Acid Sequence , Animals , Cell Aggregation/drug effects , Cell Aggregation/physiology , Cell Differentiation/drug effects , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/drug effects , Mice , Molecular Sequence Data , Neurogenesis/drug effects , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tretinoin/pharmacology
19.
J Cell Biochem ; 106(6): 1079-89, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19229867

ABSTRACT

The core embryonic stem cell transcription factors Oct4, Sox2, and Nanog are expressed in germ cell tumors (GCTs) and have been proposed to play a regulatory role in tumorigenesis. However, little is known about the mechanism of regulation of tumorigenesis by the complicated network of these proteins. Nanog is a novel homeobox-containing transcription factor that is expressed in pluripotent cells as well as GCTs. To understand the molecular and functional role of human NANOG (hNANOG) in germ cells, mutagenesis of the C-terminal domain (CD) of hNANOG and transient transfection assays in NCCIT human embryonic carcinoma cells were carried out to identify critical transactivation motifs. We divided the CD into three putative functional subdomains, CD1, tryptophan-repeat (WR) subdomain, and CD2. WR subdomain and CD2 independently contained transcriptional potential and, in combination, had a synergistic effect on transcriptional activity, while CD1 was transcriptionally inactive. The glutamine (Q) motif in WR subdomain, and multiple acidic residues in CD2 were required for maximal and synergistic transcriptional activation by the hNANOG CD. The results of the current study contribute to a better understanding of the complicated molecular machinery of stem cell transcription factors and their role in unregulated proliferation in germ cell tumorigenesis.


Subject(s)
Embryonal Carcinoma Stem Cells/physiology , Homeodomain Proteins/metabolism , Transcriptional Activation , Amino Acid Sequence , Animals , Cell Line , Embryonal Carcinoma Stem Cells/cytology , Genes, Reporter , Homeodomain Proteins/genetics , Humans , Mice , Molecular Sequence Data , Nanog Homeobox Protein , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment
20.
Int J Cardiol ; 134(1): 75-81, 2009 May 01.
Article in English | MEDLINE | ID: mdl-18538425

ABSTRACT

BACKGROUND: It has been reported that P19 embryonal carcinoma (EC) cells differentiate into beating cardiomyocytes under the action of oxytocin (OT). It has been suggested that dimethylsulfoxide (DMSO) acts via the oxytocin/oxytocin receptor pathway because an oxytocin receptor antagonist not only blocks oxytocin-induced cardiomyocyte differentiation, but also blocks DMSO-induced differentiation. In this study, the differentiation ability of OT was tested using P19CL6 cells. METHODS: P19CL6 cells were cultured as a confluent monolayer and aggregated cells. OT was then added to culture media as an inducing agent. The cells treated with 1% DMSO were used as a positive control group. Differentiated cells were evaluated morphologically and immunocytochemically, as well as by RT-PCR. In addition, a stable line of green fluorescent protein (GFP)-expressing P19CL6 cells were differentiated into beating cardiomyocytes by OT. RESULTS: Aggregated P19CL6 cells could be differentiated into cardiomyocytes, whereas monolayer cells could not differentiate and express specific cardiac muscle marker genes. In the control group, both aggregates and monolayer cells could be differentiated into cardiomyocytes by DMSO. In addition, GFP-expressing P19CL6 cells differentiated efficiently into beating cardiomyocytes when treated with OT. The results of all evaluations confirmed that the differentiated cells were cardiomyocytes. CONCLUSIONS: We concluded that embryoid body formation (cell aggregation) is necessary for the differentiation of P19CL6 cells into cardiomyocytes when using OT as an inducer agent. Furthermore, because of the high rate of differentiation efficiency, GFP-expressing cardiomyocytes derived from P19CL6 cells have the potential to be used for regenerative therapies in experimental models.


Subject(s)
Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/drug effects , Myocytes, Cardiac/cytology , Oxytocin/pharmacology , Animals , Cell Aggregation/drug effects , Cell Differentiation/drug effects , Dimethyl Sulfoxide/pharmacology , Embryonal Carcinoma Stem Cells/physiology , Free Radical Scavengers/pharmacology , Green Fluorescent Proteins/genetics , Mice , Myocardial Contraction , Myocytes, Cardiac/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...