Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Mol Cell ; 53(2): 290-300, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24374312

RESUMEN

JARID2 is an accessory component of Polycomb repressive complex-2 (PRC2) required for the differentiation of embryonic stem cells (ESCs). A role for JARID2 in the recruitment of PRC2 to target genes silenced during differentiation has been put forward, but the molecular details remain unclear. We identified a 30-amino-acid region of JARID2 that mediates interactions with long noncoding RNAs (lncRNAs) and found that the presence of lncRNAs stimulated JARID2-EZH2 interactions in vitro and JARID2-mediated recruitment of PRC2 to chromatin in vivo. Native and crosslinked RNA immunoprecipitations of JARID2 revealed that Meg3 and other lncRNAs from the imprinted Dlk1-Dio3 locus, an important regulator of development, interacted with PRC2 via JARID2. Lack of MEG3 expression in human induced pluripotent cells altered the chromatin distribution of JARID2, PRC2, and H3K27me3. Our findings show that lncRNAs facilitate JARID2-PRC2 interactions on chromatin and suggest a mechanism by which lncRNAs contribute to PRC2 recruitment.


Asunto(s)
Cromatina/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Complejo Represivo Polycomb 2/fisiología , ARN no Traducido/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Proteína Potenciadora del Homólogo Zeste 2 , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas , Ratones , Complejo Represivo Polycomb 2/química , ARN Largo no Codificante/metabolismo
2.
PLoS Genet ; 7(5): e1002085, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21637780

RESUMEN

Epigenetic reprogramming is a critical event in the generation of induced pluripotent stem cells (iPSCs). Here, we determined the DNA methylation profiles of 22 human iPSC lines derived from five different cell types (human endometrium, placental artery endothelium, amnion, fetal lung fibroblast, and menstrual blood cell) and five human embryonic stem cell (ESC) lines, and we followed the aberrant methylation sites in iPSCs for up to 42 weeks. The iPSCs exhibited distinct epigenetic differences from ESCs, which were caused by aberrant methylation at early passages. Multiple appearances and then disappearances of random aberrant methylation were detected throughout iPSC reprogramming. Continuous passaging of the iPSCs diminished the differences between iPSCs and ESCs, implying that iPSCs lose the characteristics inherited from the parent cells and adapt to very closely resemble ESCs over time. Human iPSCs were gradually reprogrammed through the "convergence" of aberrant hyper-methylation events that continuously appeared in a de novo manner. This iPS reprogramming consisted of stochastic de novo methylation and selection/fixation of methylation in an environment suitable for ESCs. Taken together, random methylation and convergence are driving forces for long-term reprogramming of iPSCs to ESCs.


Asunto(s)
Diferenciación Celular , Metilación de ADN , Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas/metabolismo , Línea Celular , Cromosomas Humanos/genética , Cromosomas Humanos/metabolismo , Células Madre Embrionarias/citología , Epigénesis Genética , Expresión Génica , Silenciador del Gen , Marcadores Genéticos , Genoma Humano , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/citología , Cariotipificación , Proteínas/genética , ARN Largo no Codificante , Transgenes
3.
BMC Genet ; 14: 32, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23631808

RESUMEN

BACKGROUND: hiPSCs are generated through epigenetic reprogramming of somatic tissue. Genomic imprinting is an epigenetic phenomenon through which monoallelic gene expression is regulated in a parent-of-origin-specific manner. Reprogramming relies on the successful erasure of marks of differentiation while maintaining those required for genomic imprinting. Loss of imprinting (LOI), which occurs in many types of malignant tumors, would hinder the clinical application of hiPSCs. RESULTS: We examined the imprinting status, expression levels and DNA methylation status of eight imprinted genes in five independently generated hiPSCs. We found a low frequency of LOI in some lines. Where LOI was identified in an early passage cell line, we found that this was maintained through subsequent passages of the cells. Just as normal imprints are maintained in long-term culture, this work suggests that abnormal imprints are also stable in culture. CONCLUSIONS: Analysis of genomic imprints in hiPSCs is a necessary safety step in regenerative medicine, with relevance both to the differentiation potential of these stem cells and also their potential tumorigenic properties.


Asunto(s)
Impresión Genómica , Inestabilidad Genómica , Células Madre Pluripotentes Inducidas/metabolismo , Metilación de ADN , Regulación de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Pérdida de Heterocigocidad
4.
J Toxicol Sci ; 48(11): 571-583, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37914285

RESUMEN

Various chemicals, including pesticides, heavy metals, and metabolites of tobacco, have been detected in fetal environment. Fetuses are exposed to these chemicals at relatively low concentrations; however, their risk of developing neurological and behavioral disorders increases after birth. We aimed to evaluate the effects of five chemicals (diethylphosphate, cotinine, octachlorodipropyl ether, mercury, and selenium) detected in the serum of pregnant mothers on neural development using human neurospheres (NSphs) differentiated from induced pluripotent stem cells. Exposure to each chemical at serum concentrations revealed no effects on NSph development. However, combined exposure to the five chemicals caused a significant decrease in NSph size and altered gene expression and neural differentiation. Thus, we next focused on DNA methylation to investigate changes in NSph properties caused by chemical exposure. Combined exposure to chemicals had extremely small effects on the DNA methylation status of NSphs at individual gene loci. However, stochastic changes in methylation status caused by chemical exposure were significantly accumulated throughout the entire genome. These results suggest that the five chemicals acted as epimutagens that alter the epigenetic status during human neural development at the biological level. Taken together, we showed for the first time, the epimutagen-induced alterations in neural differentiation at serum concentrations using an in vitro human neuronal model.


Asunto(s)
Epigénesis Genética , Células Madre Pluripotentes Inducidas , Embarazo , Femenino , Humanos , Mutágenos/metabolismo , Metilación de ADN , Diferenciación Celular/genética
5.
Mol Cancer ; 11: 89, 2012 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-23231822

RESUMEN

BACKGROUND: Transgenes introduced into cancer cell lines serve as powerful tools for identification of genes involved in cancer. However, the random nature of genomic integration site of a transgene highly influences the fidelity, reliability and level of its expression. In order to alleviate this bottleneck, we characterized the potential utility of a novel PhiC31 integrase-mediated site-specific insertion system (PhiC31-IMSI) for introduction of transgenes into a pre-inserted docking site in the genome of cancer cells. METHODS: According to this system, a "docking-site" was first randomly inserted into human cancer cell lines and clones with a single copy were selected. Subsequently, an "incoming" vector containing the gene of interest was specifically inserted in the docking-site using PhiC31. RESULTS: Using the Pc-3 and SKOV-3 cancer cell lines, we showed that transgene insertion is reproducible and reliable. Furthermore, the selection system ensured that all surviving stable transgenic lines harbored the correct integration site. We demonstrated that the expression levels of reporter genes, such as green fluorescent protein and luciferase, from the same locus were comparable among sister, isogenic clones. Using in vivo xenograft studies, we showed that the genetically altered cancer cell lines retain the properties of the parental line. To achieve temporal control of transgene expression, we coupled our insertion strategy with the doxycycline inducible system and demonstrated tight regulation of the expression of the antiangiogenic molecule sFlt-1-Fc in Pc-3 cells. Furthermore, we introduced the luciferase gene into the insertion cassette allowing for possible live imaging of cancer cells in transplantation assays. We also generated a series of Gateway cloning-compatible intermediate cassettes ready for high-throughput cloning of transgenes and demonstrated that PhiC31-IMSI can be achieved in a high throughput 96-well plate format. CONCLUSIONS: The novel PhiC31-IMSI system described in this study represents a powerful tool that can facilitate the characterization of cancer-related genes.


Asunto(s)
Técnicas de Transferencia de Gen , Recombinación Homóloga , Neoplasias/genética , Transgenes , Animales , Línea Celular Tumoral , Doxiciclina/farmacología , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Orden Génico , Genes Reporteros , Vectores Genéticos , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Reproducibilidad de los Resultados , Trasplante Heterólogo
6.
Methods ; 53(4): 380-5, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21185379

RESUMEN

Homologous and site-specific DNA recombination has revolutionized genetic engineering. The reliability of recombinases such as Cre and FLP has allowed scientists to design complex strategies to study gene function in mammals. However, the retention of recombination sites in the genome limits the use of Cre and FLP recombinases in subsequent modifications. Access to additional recombinases in the ES cell toolbox would enormously widen the number of possibilities to manipulate the genome. In the method presented here, we combine the use of PhiC31, a site-specific integrase, with FLP to obtain site-specific insertion and replacement in pre-inserted docking sites in the genome of mouse ES cells. This method allows for the integration of any sequence of interest in a pre-defined locus, leaving Cre recombinase available for downstream applications. The selection strategy is based on a silent selection marker activated by a plasmid-delivered promoter, making the integration system highly reliable and reducing the need for extensive molecular screens. This article describes how to create "dockable" mouse embryonic stem (ES) cell lines, integrate incoming vectors, and analyze the resulting clones. Current applications of this technology are also discussed.


Asunto(s)
Ingeniería Genética/métodos , Integrasas/genética , Recombinasas/genética , Animales , Southern Blotting , Técnicas de Cultivo de Célula , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Componentes del Gen , Vectores Genéticos , Humanos , Ratones , Análisis de Secuencia de ADN , Transfección/métodos
7.
Regen Ther ; 21: 620-630, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36514370

RESUMEN

Introduction: Human induced pluripotent stem cells (hiPSCs) are useful tools for reproducing neural development in vitro. However, each hiPSC line has a different ability to differentiate into specific lineages, known as differentiation propensity, resulting in reduced reproducibility and increased time and funding requirements for research. To overcome this issue, we searched for predictive signatures of neural differentiation propensity of hiPSCs focusing on DNA methylation, which is the main modulator of cellular properties. Methods: We obtained 32 hiPSC lines and their comprehensive DNA methylation data using the Infinium MethylationEPIC BeadChip. To assess the neural differentiation efficiency of these hiPSCs, we measured the percentage of neural stem cells on day 7 of induction. Using the DNA methylation data of undifferentiated hiPSCs and their measured differentiation efficiency into neural stem cells as the set of data, and HSIC Lasso, a machine learning-based nonlinear feature selection method, we attempted to identify neural differentiation-associated differentially methylated sites. Results: Epigenome-wide unsupervised clustering cannot distinguish hiPSCs with varying differentiation efficiencies. In contrast, HSIC Lasso identified 62 CpG sites that could explain the neural differentiation efficiency of hiPSCs. Features selected by HSIC Lasso were particularly enriched within 3 Mbp of chromosome 5, harboring IRX1, IRX2, and C5orf38 genes. Within this region, DNA methylation rates were correlated with neural differentiation efficiency and were negatively correlated with gene expression of the IRX1/2 genes, particularly in female hiPSCs. In addition, forced expression of the IRX1/2 impaired the neural differentiation ability of hiPSCs in both sexes. Conclusion: We for the first time showed that the DNA methylation state of the IRX1/2 genes of hiPSCs is a predictive biomarker of their potential for neural differentiation. The predictive markers for neural differentiation efficiency identified in this study may be useful for the selection of suitable undifferentiated hiPSCs prior to differentiation induction.

8.
Int J Biol Macromol ; 208: 333-342, 2022 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-35339495

RESUMEN

Cartilage in the head of sturgeon or salmon has been gaining attention as a rich source of functional chondroitin sulfate (CS) or proteoglycans. Although the cartilage was found in the heads of other bony fishes, the structure of CS and its core protein, especially aggrecan, was not fully investigated. In this study, comprehensive analysis of CS and aggrecan in the head cartilage of 10 bony fishes including sturgeon and salmon was performed. The 4-O-sulfation to 6-O-sulfation ratio (4S/6S ratio; S: sulfate residue) of CS in Perciformes was ≧1.0, while the 4S/6S ratios of CS from sturgeons and salmon were less than 0.5. Dot blotting and proteomic analysis revealed that aggrecan was a major core protein in head cartilage of all bony fishes. These results suggest that the head cartilage of bony fishes is a promising source for the preparation of CS or proteoglycans as a health food ingredient.


Asunto(s)
Sulfatos de Condroitina , Proteoglicanos , Agrecanos/análisis , Animales , Cartílago/metabolismo , Sulfatos de Condroitina/química , Peces/metabolismo , Proteoglicanos/química , Proteómica , Salmón/metabolismo
9.
J Reprod Dev ; 57(5): 586-93, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21636956

RESUMEN

The Sry (sex determining region on Y chromosome) gene is a master gene for sex determination. We previously reported that the Sry gene has tissue-dependent and differentially methylated regions (T-DMRs) by analyzing the DNA methylation states at CpG sites in the promoter regions. In this study, we found unique non-CpG methylation at the internal cytosine in the 5'-CCTGG-3' pentanucleotide sequence in the Sry T-DMR. This non-CpG methylation was detected in four mouse strains (ICR, BALB/c, DBA2 and C3H), but not in two strains (C57BL/6 and 129S1), suggesting that the CCTGG methylation is tentative and unstable. Interestingly, this CCTGG methylation was associated with demethylation of the CpG sites in the Sry T-DMR in the developmental process. A methylation-mediated promoter assay showed that the CCTGG methylation promotes gene expression. Our finding shows that non-CpG methylation has unique characteristic and is still conserved in mammals.


Asunto(s)
Metilación de ADN/fisiología , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteína de la Región Y Determinante del Sexo/genética , Animales , Islas de CpG/genética , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos ICR , Embarazo , Análisis de Secuencia de ADN , Proteína de la Región Y Determinante del Sexo/metabolismo
10.
Hum Cell ; 34(1): 99-110, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33047283

RESUMEN

The use of human induced pluripotent stem cells (iPSCs), used as an alternative to human embryonic stem cells (ESCs), is a potential solution to challenges, such as immune rejection, and does not involve the ethical issues concerning the use of ESCs in regenerative medicine, thereby enabling developments in biological research. However, comparative analyses from previous studies have not indicated any specific feature that distinguishes iPSCs from ESCs. Therefore, in this study, we established a linear classification-based learning model to distinguish among ESCs, iPSCs, embryonal carcinoma cells (ECCs), and somatic cells on the basis of their DNA methylation profiles. The highest accuracy achieved by the learned models in identifying the cell type was 94.23%. In addition, the epigenetic signature of iPSCs, which is distinct from that of ESCs, was identified by component analysis of the learned models. The iPSC-specific regions with methylation fluctuations were abundant on chromosomes 7, 8, 12, and 22. The method developed in this study can be utilized with comprehensive data and widely applied to many aspects of molecular biology research.


Asunto(s)
Células Madre Pluripotentes Inducidas , Aprendizaje Automático , Células Cultivadas , Cromosomas Humanos/genética , Metilación de ADN , Células Madre Embrionarias , Epigénesis Genética , Humanos , Biología Molecular/métodos , Medicina Regenerativa/métodos
11.
Genes Cells ; 14(12): 1395-404, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19912344

RESUMEN

Practical clinical applications for current induced pluripotent stem cell (iPSC) technologies are hindered by very low generation efficiencies. Here, we demonstrate that newborn human (h) and mouse (m) extra-embryonic amnion (AM) and yolk-sac (YS) cells, in which endogenous KLF4/Klf4, c-MYC/c-Myc and RONIN/Ronin are expressed, can be reprogrammed to hiPSCs and miPSCs with efficiencies for AM cells of 0.02% and 0.1%, respectively. Both hiPSC and miPSCs are indistinguishable from embryonic stem cells in colony morphology, expression of pluripotency markers, global gene expression profile, DNA methylation status of OCT4 and NANOG, teratoma formation and, in the case of miPSCs, generation of germline transmissible chimeric mice. As copious amounts of human AM cells can be collected without invasion, and stored long term by conventional means without requirement for in vitro culture, they represent an ideal source for cell banking and subsequent 'on demand' generation of hiPSCs for personal regenerative and pharmaceutical applications.


Asunto(s)
Amnios/citología , Reprogramación Celular/fisiología , Células Madre Embrionarias/fisiología , Células Madre Pluripotentes/fisiología , Saco Vitelino/citología , Amnios/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Quimera , Metilación de ADN , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Recién Nacido , Factor 4 Similar a Kruppel , Masculino , Ratones , Ratones Transgénicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Teratoma/metabolismo , Teratoma/patología , Saco Vitelino/metabolismo
12.
Exp Cell Res ; 315(16): 2727-40, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19559696

RESUMEN

POU5F1 (more commonly known as OCT4/3) is one of the stem cell markers, and affects direction of differentiation in embryonic stem cells. To investigate whether cells of mesenchymal origin acquire embryonic phenotypes, we generated human cells of mesodermal origin with overexpression of the chimeric OCT4/3 gene with physiological co-activator EWS (product of the EWSR1 gene), which is driven by the potent EWS promoter by translocation. The cells expressed embryonic stem cell genes such as NANOG, lost mesenchymal phenotypes, and exhibited embryonal stem cell-like alveolar structures when implanted into the subcutaneous tissue of immunodeficient mice. Hierarchical analysis by microchip analysis and cell surface analysis revealed that the cells are subcategorized into the group of human embryonic stem cells and embryonal carcinoma cells. These results imply that cells of mesenchymal origin can be traced back to cells of embryonic phenotype by the OCT4/3 gene in collaboration with the potent cis-regulatory element and the fused co-activator. The cells generated in this study with overexpression of chimeric OCT4/3 provide us with insight into cell plasticity involving OCT4/3 that is essential for embryonic cell maintenance, and the complexity required for changing cellular identity.


Asunto(s)
Embrión de Mamíferos , Mesodermo/fisiología , Morfogénesis/fisiología , Factor 3 de Transcripción de Unión a Octámeros , Proteína EWS de Unión a ARN/metabolismo , Proteínas Recombinantes de Fusión , Animales , Biomarcadores/metabolismo , Linaje de la Célula , Células Cultivadas , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Cariotipificación , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Análisis de Componente Principal , Proteína EWS de Unión a ARN/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Teratoma/metabolismo , Teratoma/patología
13.
J Vet Med Sci ; 82(6): 681-689, 2020 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-32238671

RESUMEN

Steroidogenic factor 1 (SF-1) is a nuclear receptor that is important in steroid hormone production, and adrenal and gonad development. The SF-1 gene is highly conserved among most vertebrates. However, dog SF-1 registered in public databases, such as CanFam3.1, lacks the 5' end compared to other mammals including mouse, human, bovine, and cat. Whether this defect is due to species differences or database error is unclear. Here, we determined the full-length dog SF-1 cDNA sequence and identified the missing 5' end sequence in the databases. The coding region of the dog SF-1 gene has 1,386 base pairs, and the protein has 461 amino acid residues. Sequence alignment analysis among vertebrates revealed that the 5' end sequence of dog SF-1 cDNA is highly conserved compared to other vertebrates. The genomic position of the first exon was determined, and its promoter region sequence was analyzed. The DNA methylation state at the basal promoter and the expression of dog SF-1 in steroidogenic tissues and non-steroidogenic cells were examined. CpG sites at the basal promoter displayed methylation kinetics inversely correlated with gene expression. The promoter was hypomethylated and hypermethylated in SF-1 expressing and non-SF-1 expressing tissues, respectively. In conclusion, we identified the true full sequence of dog SF-1 cDNA and determined the genome sequence around the first exon. The gene is under the control of epigenetic regulation, such as DNA methylation, at the promoter.


Asunto(s)
Perros/genética , Epigénesis Genética , Análisis de Secuencia de ADN , Factor Esteroidogénico 1/genética , Tejido Adiposo/metabolismo , Glándulas Suprarrenales/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , ADN Complementario , Femenino , Regulación de la Expresión Génica , Masculino , Ovario/metabolismo , Alineación de Secuencia , Factor Esteroidogénico 1/metabolismo , Testículo/metabolismo
14.
Sci Rep ; 10(1): 5287, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32210272

RESUMEN

Fibrillin-1 (FBN1) is responsible for haploinsufficient and autosomal dominant Marfan syndrome. Even in the same Marfan pedigree, penetrance and expressivity in heterozygous individuals can differ and result in variable disease onset and severity. Thus, other factors in addition to mutations in FBN1 are likely to contribute to the disease. In this study, we examined the regulation of FBN1 in porcine Marfan syndrome model, focusing on DNA methylation patterns distinguishable as wild-type (WT) and FBN1 null (KO) alleles in heterozygous cells. Most importantly, the ratio of the transcriptionally active hypomethylated WT allele was altered during cellular passage and highly correlated with FBN1 mRNA level compared with that in the KO allele. Transcribed FBN1 RNA from the KO allele was abolished after splicing coupled with translational initiation, suggesting that the functional FBN1 mRNA levels were affected by DNA methylation of the WT allele.


Asunto(s)
Islas de CpG , Metilación de ADN , Fibrilina-1/genética , Fibroblastos/patología , Regulación de la Expresión Génica , Síndrome de Marfan/patología , Mutación , Animales , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Penetrancia , Porcinos
15.
Hum Cell ; 31(1): 78-86, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29103143

RESUMEN

During reprogramming into human induced pluripotent stem cells (iPSCs), several stem cell marker genes are induced, such as OCT-4, NANOG, SALL4, and TERT. OCT-4, NANOG, and SALL4 gene expression can be regulated by DNA methylation. Their promoters become hypomethylated in iPSCs during reprogramming, leading to their induced expression. However, epigenetic regulation of the TERT gene remains unclear. In this study, we focused on epigenetic regulation of the human TERT gene and identified a differentially methylated region (DMR) at a distal region in the TERT promoter between human iPSCs and their parental somatic cells. Interestingly, the TERT-DMR was highly methylated in iPSCs, but low-level methylation was observed in their parental somatic cells. Region-specific, methylated-promoter assays showed that the methylated TERT-DMR up-regulated the promoter activity in iPSCs. In addition, Lamin B1 accumulated at the TERT-DMR in iPSCs, but not in their parent somatic cells. These results suggested that the TERT transcription was enhanced by DNA methylation at the TERT-DMR via binding to nuclear lamina during reprogramming. Our findings shed light on a new functional aspect of DNA methylation in gene expression.


Asunto(s)
Reprogramación Celular/genética , Metilación de ADN/fisiología , Expresión Génica/genética , Células Madre Pluripotentes Inducidas/enzimología , Telomerasa/genética , Telomerasa/metabolismo , Células Cultivadas , Epigénesis Genética , Humanos , Transcripción Genética/genética
16.
Regen Ther ; 9: 71-78, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30525077

RESUMEN

Human induced pluripotent stem cells (iPSCs) are established by introducing several reprogramming factors, such as OCT3/4, SOX2, KLF4, c-MYC. Because of their pluripotency and immortality, iPSCs are considered to be a powerful tool for regenerative medicine. To date, iPSCs have been established all over the world by various gene delivery methods. All methods induced high-quality iPSCs, but epigenetic analysis of abnormalities derived from differences in the gene delivery methods has not yet been performed. Here, we generated genetically matched human iPSCs from menstrual blood cells by using three kinds of vectors, i.e., retrovirus, Sendai virus, and episomal vectors, and compared genome-wide DNA methylation profiles among them. Although comparison of aberrant methylation revealed that iPSCs generated by Sendai virus vector have lowest number of aberrant methylation sites among the three vectors, the iPSCs generated by non-integrating methods did not show vector-specific aberrant methylation. However, the differences between the iPSC lines were determined to be the number of random aberrant hypermethylated regions compared with embryonic stem cells. These random aberrant hypermethylations might be a cause of the differences in the properties of each of the iPSC lines.

17.
J Vet Med Sci ; 79(2): 230-237, 2017 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-27916785

RESUMEN

Runt-related transcription factor 2 (Runx2) is essential for osteogenesis. This study aimes at identification of the genomic region differentially methylated in DNA for regulation of Runx2 expression. In the proximal promoter of mouse Runx2, DNA methylation was frequent at the region further than 3 kb relative to the transcription start site, in contrast to lower methylation status of the closer locus within 2 kb from the transcription start site. At the intermediate part, we identified a novel differentially methylated region in the Runx2 promoter region (Runx2-DMR): from -2.7 to -2.2 kb relative to the start site of Runx2 transcription in mice. In this region, the DNA methylation rate correlated negatively with Runx2 expression among mouse organs as well as among primary cultures of bone marrow from different dogs. Induction of mouse and dog mesenchymal-like cells into osteoblastic differentiation decreased the methylation rate of Runx2-DMR. Thus, in this study, we identified a novel genomic region in which DNA methylation status is related to Runx2 expression and detected demethylation of Runx2-DMR during osteoblastic differentiation in mouse and dog.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Metilación de ADN , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Perros , Regulación de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis , Regiones Promotoras Genéticas , Especificidad de la Especie
18.
Methods Mol Biol ; 329: 421-45, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16846008

RESUMEN

Stem cells raise the possibility of regenerating failing body parts with new tissue. Before stem cells can safely fulfill their promise, many technical problems, including understanding the stem cell phenotype, must be overcome. DNA methylation, which is responsible for gene silencing and is associated with chromatin remodeling, is an epigenetic system that determines the specific characteristic of a variety of cells, including stem cells. Each cell type has a unique DNA methylation profile produced by varied loci-specific methylation. Investigation of such DNA methylation profiles provides a way of identifying pluripotent stem cells. Further, it is likely that analysis of the epigenetic status of stem cells may provide novel information regarding "sternness" within these populations.


Asunto(s)
Metilación de ADN , Embrión de Mamíferos/citología , Células Madre Pluripotentes/metabolismo , Animales , Secuencia de Bases , Southern Blotting , Técnicas de Cultivo de Célula/métodos , Línea Celular , ADN/química , ADN/genética , ADN/aislamiento & purificación , Epigénesis Genética , Ratones , Células Madre Pluripotentes/citología , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Mapeo Restrictivo , Teratoma/química , Teratoma/genética
19.
Hum Cell ; 29(3): 97-100, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27083573

RESUMEN

Indeed human induced pluripotent stem cells (hiPSCs) are considered to be powerful tools in regenerative medicine. To enable the use of hiPSCs in the field of regenerative medicine, it is necessary to understand the mechanisms of reprogramming during the transformation of somatic cells into hiPSCs. Genome-wide epigenetic modification constitutes a critical event in the generation of iPSCs. In other words, to analyze epigenetic changes in iPSCs means to elucidate reprogramming processes. We have established a large number of hiPSCs derived from various human tissues and have obtained their DNA methylation profiles. Comparison analyses indicated that the epigenetic patterns of various hiPSCs, irrespective of their source tissue, were very similar to one another and were similar to those of human embryonic stem cells (hESCs). However, the profiles of hiPSCs and hESCs exhibited epigenetic differences, which were caused by random aberrant hypermethylation at early passages. Interestingly, continuous passaging of the hiPSCs diminished the differences between DNA methylation profiles of hiPSCs and hESCs. The number of aberrant DNA methylation regions may thus represent a useful epigenetic index for evaluating hiPSCs in the context of therapeutic applications.


Asunto(s)
Reprogramación Celular/genética , Metilación de ADN , Células Madre Pluripotentes Inducidas/citología , Metilación de ADN/genética , Epigénesis Genética , Humanos , Medicina Regenerativa
20.
PLoS One ; 10(6): e0130585, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26091100

RESUMEN

Polyploid amphibians and fishes occur naturally in nature, while polyploid mammals do not. For example, tetraploid mouse embryos normally develop into blastocysts, but exhibit abnormalities and die soon after implantation. Thus, polyploidization is thought to be harmful during early mammalian development. However, the mechanisms through which polyploidization disrupts development are still poorly understood. In this study, we aimed to elucidate how genome duplication affects early mammalian development. To this end, we established tetraploid embryonic stem cells (TESCs) produced from the inner cell masses of tetraploid blastocysts using electrofusion of two-cell embryos in mice and studied the developmental potential of TESCs. We demonstrated that TESCs possessed essential pluripotency and differentiation potency to form teratomas, which differentiated into the three germ layers, including diploid embryonic stem cells. TESCs also contributed to the inner cell masses in aggregated chimeric blastocysts, despite the observation that tetraploid embryos fail in normal development soon after implantation in mice. In TESCs, stability after several passages, colony morphology, and alkaline phosphatase activity were similar to those of diploid ESCs. TESCs also exhibited sufficient expression and localization of pluripotent markers and retained the normal epigenetic status of relevant reprogramming factors. TESCs proliferated at a slower rate than ESCs, indicating that the difference in genomic dosage was responsible for the different growth rates. Thus, our findings suggested that mouse ESCs maintained intrinsic pluripotency and differentiation potential despite tetraploidization, providing insights into our understanding of developmental elimination in polyploid mammals.


Asunto(s)
Células Madre Embrionarias/citología , Estratos Germinativos/metabolismo , Animales , Blastocisto/citología , Diferenciación Celular , Proliferación Celular , Metilación de ADN , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Femenino , Genes Reporteros , Ratones , Tetraploidía , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda