Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Stem Cell ; 9(6): 588-98, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-22136932

ABSTRACT

We compared two genetically highly defined transgenic systems to identify parameters affecting reprogramming of somatic cells to a pluripotent state. Our results demonstrate that the level and stoichiometry of reprogramming factors during the reprogramming process strongly influence the resulting pluripotency of iPS cells. High expression of Oct4 and Klf4 combined with lower expression of c-Myc and Sox2 produced iPS cells that efficiently generated "all-iPSC mice" by tetraploid (4n) complementation, maintained normal imprinting at the Dlk1-Dio3 locus, and did not create mice with tumors. Loss of imprinting (LOI) at the Dlk1-Dio3 locus did not strictly correlate with reduced pluripotency though the efficiency of generating "all-iPSC mice" was diminished. Our data indicate that stoichiometry of reprogramming factors can influence epigenetic and biological properties of iPS cells. This concept complicates efforts to define a "generic" epigenetic state of iPSCs and ESCs and should be considered when comparing different iPS and ES cell lines.


Subject(s)
Cellular Reprogramming/physiology , Epigenesis, Genetic , Induced Pluripotent Stem Cells/physiology , Animals , Biomarkers/metabolism , Cell Line , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Transgenic , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Transgenes
2.
Proc Natl Acad Sci U S A ; 108(44): 18061-6, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22011581

ABSTRACT

Alterations in DNA methylation have been associated with genome-wide hypomethylation and regional de novo methylation in numerous cancers. De novo methylation is mediated by the de novo methyltransferases Dnmt3a and 3b, but only Dnmt3b has been implicated in promoting cancer by silencing of tumor-suppressor genes. In this study, we have analyzed the role of Dnmt3a in lung cancer by using a conditional mouse tumor model. We show that Dnmt3a deficiency significantly promotes tumor growth and progression but not initiation. Changes in gene expression show that Dnmt3a deficiency affects key steps in cancer progression, such as angiogenesis, cell adhesion, and cell motion, consistent with accelerated and more malignant growth. Our results suggest that Dnmt3a may act like a tumor-suppressor gene in lung tumor progression and may be a critical determinant of lung cancer malignancy.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Lung Neoplasms/pathology , Animals , Base Sequence , Cell Division , DNA Methylation , DNA Methyltransferase 3A , DNA Primers , Disease Models, Animal , Disease Progression , Gene Silencing , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
3.
Stem Cells ; 29(6): 992-1000, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21563275

ABSTRACT

Pluripotent cells can be derived from different types of somatic cells by nuclear reprogramming through the ectopic expression of four transcription factors, Oct3/4, Sox2, Klf4, and c-Myc. However, it is unclear whether postmitotic neurons are susceptible to direct reprogramming. Here, we show that postnatal cortical neurons, the vast majority of which are postmitotic, are amenable to epigenetic reprogramming. However, ectopic expression of the four canonical reprogramming factors is not sufficient to reprogram postnatal neurons. Efficient reprogramming was only achieved after forced cell proliferation by p53 suppression. Additionally, overexpression of repressor element-1 silencing transcription, a suppressor of neuronal gene activity, increased reprogramming efficiencies in combination with the reprogramming factors. Our findings indicate that terminally differentiated postnatal neurons are able to acquire the pluripotent state by direct epigenetic reprogramming, and this process is made more efficient through the suppression of lineage specific gene expression.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Repressor Proteins/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Blastocyst/cytology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Coculture Techniques , Embryo Transfer , Fibroblasts/cytology , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Kruppel-Like Factor 4 , Lewis X Antigen/metabolism , Mice , Nanog Homeobox Protein , Neurons/metabolism , Octamer Transcription Factor-3/metabolism , Promoter Regions, Genetic , Teratoma/pathology , Transplantation Chimera
4.
J Clin Invest ; 121(5): 1748-52, 2011 May.
Article in English | MEDLINE | ID: mdl-21490393

ABSTRACT

Human cancer cells frequently have regions of their DNA hypermethylated, which results in transcriptional silencing of affected genes and promotion of tumor formation. However, it is still unknown whether cancer-associated aberrant DNA methylation is targeted to specific genomic regions, whether this methylation also occurs in noncancerous cells, and whether these epigenetic events are maintained in the absence of the initiating cause. Here we have addressed some of these issues by demonstrating that transgenic expression of DNA methyltransferase 3b (Dnmt3b) in the mouse colon initiates de novo DNA methylation of genes that are similar to genes that become methylated in human colon cancer. This is consistent with the notion that aberrant methylation in cancer may be attributable to targeting of specific sequences by Dnmt3b rather than to random methylation followed by clonal selection. We also showed that Dnmt3b-induced aberrant DNA methylation was maintained in regenerating tissue, even in the absence of continuous Dnmt3b expression. This supports the concept that transient stressors can cause permanent epigenetic changes in somatic stem cells and that these accumulate over the lifetime of an organism in analogy to DNA mutations.


Subject(s)
Colonic Neoplasms/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Intestinal Mucosa/metabolism , Animals , Colon/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Transgenic , Mutation , Receptors, G-Protein-Coupled/metabolism , DNA Methyltransferase 3B
5.
Proc Natl Acad Sci U S A ; 107(50): 21931-6, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21106759

ABSTRACT

Developmental programs are controlled by transcription factors and chromatin regulators, which maintain specific gene expression programs through epigenetic modification of the genome. These regulatory events at enhancers contribute to the specific gene expression programs that determine cell state and the potential for differentiation into new cell types. Although enhancer elements are known to be associated with certain histone modifications and transcription factors, the relationship of these modifications to gene expression and developmental state has not been clearly defined. Here we interrogate the epigenetic landscape of enhancer elements in embryonic stem cells and several adult tissues in the mouse. We find that histone H3K27ac distinguishes active enhancers from inactive/poised enhancer elements containing H3K4me1 alone. This indicates that the amount of actively used enhancers is lower than previously anticipated. Furthermore, poised enhancer networks provide clues to unrealized developmental programs. Finally, we show that enhancers are reset during nuclear reprogramming.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Histones/metabolism , Acetylation , Animals , Cell Differentiation/genetics , Cell Line , Histones/genetics , Mice , Mice, Inbred C57BL
6.
Genome Res ; 20(8): 1133-42, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20508142

ABSTRACT

Isogenic settings are routine in model organisms, yet remain elusive for genetic experiments on human cells. We describe the use of designed zinc finger nucleases (ZFNs) for efficient transgenesis without drug selection into the PPP1R12C gene, a "safe harbor" locus known as AAVS1. ZFNs enable targeted transgenesis at a frequency of up to 15% following transient transfection of both transformed and primary human cells, including fibroblasts and hES cells. When added to this locus, transgenes such as expression cassettes for shRNAs, small-molecule-responsive cDNA expression cassettes, and reporter constructs, exhibit consistent expression and sustained function over 50 cell generations. By avoiding random integration and drug selection, this method allows bona fide isogenic settings for high-throughput functional genomics, proteomics, and regulatory DNA analysis in essentially any transformed human cell type and in primary cells.


Subject(s)
Gene Transfer Techniques , Genome, Human , Genomics/methods , Regulatory Sequences, Nucleic Acid/genetics , Zinc Fingers/genetics , Base Sequence , Cell Line , Endonucleases/genetics , Genetic Loci , Humans , Molecular Sequence Data , Proteomics/methods
7.
Cell ; 133(2): 250-64, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18423197

ABSTRACT

Pluripotent cells can be derived from fibroblasts by ectopic expression of defined transcription factors. A fundamental unresolved question is whether terminally differentiated cells can be reprogrammed to pluripotency. We utilized transgenic and inducible expression of four transcription factors (Oct4, Sox2, Klf4, and c-Myc) to reprogram mouse B lymphocytes. These factors were sufficient to convert nonterminally differentiated B cells to a pluripotent state. However, reprogramming of mature B cells required additional interruption with the transcriptional state maintaining B cell identity by either ectopic expression of the myeloid transcription factor CCAAT/enhancer-binding-protein-alpha (C/EBPalpha) or specific knockdown of the B cell transcription factor Pax5. Multiple iPS lines were clonally derived from both nonfully and fully differentiated B lymphocytes, which gave rise to adult chimeras with germline contribution, and to late-term embryos when injected into tetraploid blastocysts. Our study provides definite proof for the direct nuclear reprogramming of terminally differentiated adult cells to pluripotency.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation , Pluripotent Stem Cells/cytology , Animals , Cell Nucleus/genetics , Embryonic Stem Cells/cytology , Humans , Kruppel-Like Factor 4 , Mice , Transcription Factors/metabolism
8.
Genes Dev ; 21(23): 3110-22, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18056424

ABSTRACT

Increased methylation of CpG islands and silencing of affected target genes is frequently found in human cancer; however, in vivo the question of causality has only been addressed by loss-of-function studies. To directly evaluate the role and mechanism of de novo methylation in tumor development, we overexpressed the de novo DNA methyltransferases Dnmt3a1 and Dnmt3b1 in Apc Min/+ mice. We found that Dnmt3b1 enhanced the number of colon tumors in Apc Min/+ mice approximately twofold and increased the average size of colonic microadenomas, whereas Dnmt3a1 had no effect. The overexpression of Dnmt3b1 caused loss of imprinting and increased expression of Igf2 as well as methylation and transcriptional silencing of the tumor suppressor genes Sfrp2, Sfrp4, and Sfrp5. Importantly, we found that Dnmt3b1 but not Dnmt3a1 efficiently methylates the same set of genes in tumors and in nontumor tissues, demonstrating that de novo methyltransferases can initiate methylation and silencing of specific genes in phenotypically normal cells. This suggests that DNA methylation patterns in cancer are the result of specific targeting of at least some tumor suppressor genes rather than of random, stochastic methylation followed by clonal selection due to a proliferative advantage caused by tumor suppressor gene silencing.


Subject(s)
Colonic Neoplasms/etiology , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Gene Silencing , Adenoma/etiology , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Carcinogens/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA Methyltransferase 3A , Down-Regulation , Genes, APC , Genomic Imprinting , Humans , Insulin-Like Growth Factor II/genetics , Loss of Heterozygosity , Mice , Mice, Inbred C57BL , Mice, Transgenic , DNA Methyltransferase 3B
SELECTION OF CITATIONS
SEARCH DETAIL
...