Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Dev Cell ; 57(24): 2714-2730.e8, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36538894

ABSTRACT

Cancer stem cells (CSCs) may serve as the cellular seeds of tumor recurrence and metastasis, and they can be generated via epithelial-mesenchymal transitions (EMTs). Isolating pure populations of CSCs is difficult because EMT programs generate multiple alternative cell states, and phenotypic plasticity permits frequent interconversions between these states. Here, we used cell-surface expression of integrin ß4 (ITGB4) to isolate highly enriched populations of human breast CSCs, and we identified the gene regulatory network operating in ITGB4+ CSCs. Specifically, we identified ΔNp63 and p73, the latter of which transactivates ΔNp63, as centrally important transcriptional regulators of quasi-mesenchymal CSCs that reside in an intermediate EMT state. We found that the transcriptional program controlled by ΔNp63 in CSCs is largely distinct from the one that it orchestrates in normal basal mammary stem cells and, instead, it more closely resembles a regenerative epithelial stem cell response to wounding. Moreover, quasi-mesenchymal CSCs repurpose this program to drive metastatic colonization via autocrine EGFR signaling.


Subject(s)
Mesenchymal Stem Cells , Neoplasms , Humans , Cell Line, Tumor , Neoplastic Stem Cells/metabolism , Signal Transduction , Epithelial-Mesenchymal Transition , Neoplasms/pathology
2.
Sci Transl Med ; 11(484)2019 03 20.
Article in English | MEDLINE | ID: mdl-30894502

ABSTRACT

Inhibiting MYC has long been considered unfeasible, although its key role in human cancers makes it a desirable target for therapeutic intervention. One reason for its perceived undruggability was the fear of catastrophic side effects in normal tissues. However, we previously designed a dominant-negative form of MYC called Omomyc and used its conditional transgenic expression to inhibit MYC function both in vitro and in vivo. MYC inhibition by Omomyc exerted a potent therapeutic impact in various mouse models of cancer, causing only mild, well-tolerated, and reversible side effects. Nevertheless, Omomyc has been so far considered only a proof of principle. In contrast with that preconceived notion, here, we show that the purified Omomyc mini-protein itself spontaneously penetrates into cancer cells and effectively interferes with MYC transcriptional activity therein. Efficacy of the Omomyc mini-protein in various experimental models of non-small cell lung cancer harboring different oncogenic mutation profiles establishes its therapeutic potential after both direct tissue delivery and systemic administration, providing evidence that the Omomyc mini-protein is an effective MYC inhibitor worthy of clinical development.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell-Penetrating Peptides/pharmacokinetics , Cell-Penetrating Peptides/therapeutic use , DNA/metabolism , Disease Models, Animal , E-Box Elements/genetics , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice, Inbred C57BL , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacokinetics , Peptide Fragments/therapeutic use , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-myc/administration & dosage , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/pharmacokinetics , Proto-Oncogene Proteins c-myc/pharmacology , Proto-Oncogene Proteins c-myc/therapeutic use
3.
Cancer Discov ; 7(10): 1136-1153, 2017 10.
Article in English | MEDLINE | ID: mdl-28729405

ABSTRACT

We characterized the enhancer landscape of 66 patients with acute myeloid leukemia (AML), identifying 6 novel subgroups and their associated regulatory loci. These subgroups are defined by their superenhancer (SE) maps, orthogonal to somatic mutations, and are associated with distinct leukemic cell states. Examination of transcriptional drivers for these epigenomic subtypes uncovers a subset of patients with a particularly strong SE at the retinoic acid receptor alpha (RARA) gene locus. The presence of a RARA SE and concomitant high levels of RARA mRNA predisposes cell lines and ex vivo models to exquisite sensitivity to a selective agonist of RARα, SY-1425 (tamibarotene). Furthermore, only AML patient-derived xenograft (PDX) models with high RARA mRNA were found to respond to SY-1425. Mechanistically, we show that the response to SY-1425 in RARA-high AML cells is similar to that of acute promyelocytic leukemia treated with retinoids, characterized by the induction of known retinoic acid response genes, increased differentiation, and loss of proliferation.Significance: We use the SE landscape of primary human AML to elucidate transcriptional circuitry and identify novel cancer vulnerabilities. A subset of patients were found to have an SE at RARA, which is predictive for response to SY-1425, a potent and selective RARα agonist, in preclinical models, forming the rationale for its clinical investigation in biomarker-selected patients. Cancer Discov; 7(10); 1136-53. ©2017 AACR.See related commentary by Wang and Aifantis, p. 1065.This article is highlighted in the In This Issue feature, p. 1047.


Subject(s)
Benzoates/administration & dosage , Enhancer Elements, Genetic , Epigenomics/methods , Leukemia, Myeloid, Acute/drug therapy , Retinoic Acid Receptor alpha/genetics , Tetrahydronaphthalenes/administration & dosage , Aged , Animals , Benzoates/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing/methods , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Retinoic Acid Receptor alpha/agonists , Tetrahydronaphthalenes/pharmacology , Up-Regulation , Xenograft Model Antitumor Assays
4.
Cell Rep ; 9(3): 1163-70, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25437568

ABSTRACT

Epigenomic profiling by chromatin immunoprecipitation coupled with massively parallel DNA sequencing (ChIP-seq) is a prevailing methodology used to investigate chromatin-based regulation in biological systems such as human disease, but the lack of an empirical methodology to enable normalization among experiments has limited the precision and usefulness of this technique. Here, we describe a method called ChIP with reference exogenous genome (ChIP-Rx) that allows one to perform genome-wide quantitative comparisons of histone modification status across cell populations using defined quantities of a reference epigenome. ChIP-Rx enables the discovery and quantification of dynamic epigenomic profiles across mammalian cells that would otherwise remain hidden using traditional normalization methods. We demonstrate the utility of this method for measuring epigenomic changes following chemical perturbations and show how reference normalization of ChIP-seq experiments enables the discovery of disease-relevant changes in histone modification occupancy.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin Immunoprecipitation/standards , Epigenesis, Genetic , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Animals , Benzimidazoles/pharmacology , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Epigenesis, Genetic/drug effects , Histones/metabolism , Humans , Jurkat Cells , Lysine/metabolism , Methylation/drug effects , Reference Standards
5.
Nat Biotechnol ; 32(1): 92-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24336317

ABSTRACT

A vast number of small-molecule ligands, including therapeutic drugs under development and in clinical use, elicit their effects by binding specific proteins associated with the genome. An ability to map the direct interactions of a chemical entity with chromatin genome-wide could provide important insights into chemical perturbation of cellular function. Here we describe a method that couples ligand-affinity capture and massively parallel DNA sequencing (Chem-seq) to identify the sites bound by small chemical molecules throughout the human genome. We show how Chem-seq can be combined with ChIP-seq to gain unique insights into the interaction of drugs with their target proteins throughout the genome of tumor cells. These methods will be broadly useful to enhance understanding of therapeutic action and to characterize the specificity of chemical entities that interact with DNA or genome-associated proteins.


Subject(s)
Chromatin/genetics , DNA/genetics , Proteins/genetics , Transcription Factors/genetics , Binding Sites/genetics , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Ligands , Protein Binding/genetics
6.
Proc Natl Acad Sci U S A ; 110(8): 2876-81, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23382218

ABSTRACT

Many long noncoding RNA (lncRNA) species have been identified in mammalian cells, but the genomic origin and regulation of these molecules in individual cell types is poorly understood. We have generated catalogs of lncRNA species expressed in human and murine embryonic stem cells and mapped their genomic origin. A surprisingly large fraction of these transcripts (>60%) originate from divergent transcription at promoters of active protein-coding genes. The divergently transcribed lncRNA/mRNA gene pairs exhibit coordinated changes in transcription when embryonic stem cells are differentiated into endoderm. Our results reveal that transcription of most lncRNA genes is coordinated with transcription of protein-coding genes.


Subject(s)
Embryonic Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Untranslated/genetics , Transcription, Genetic , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Humans , Mice
7.
Genomics ; 100(5): 320-6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22819920

ABSTRACT

TrxG and PcG complexes play key roles in the epigenetic regulation of development through H3K4me3 and H3K27me3 modification at specific sites throughout the human genome, but how these sites are selected is poorly understood. We find that in pluripotent cells, clustered CpG-islands at genes predict occupancy of H3K4me3 and H3K27me3, and these "bivalent" chromatin domains precisely span the boundaries of CpG-island clusters. These relationships are specific to pluripotent stem cells and are not retained at H3K4me3 and H3K27me3 sites unique to differentiated cells. We show that putative transcripts from clustered CpG-islands predict stem-loop structures characteristic of those bound by PcG complexes, consistent with the possibility that RNA facilitates PcG recruitment or maintenance at these sites. These studies suggest that CpG-island structure plays a fundamental role in establishing developmentally important chromatin structures in the pluripotent genome, and a subordinate role in establishing TrxG/PcG chromatin structure at sites unique to differentiated cells.


Subject(s)
Chromatin/genetics , CpG Islands/genetics , Epigenesis, Genetic/genetics , Histones/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Pluripotent Stem Cells/metabolism , Polycomb-Group Proteins/genetics , Chromatin Immunoprecipitation , Histone-Lysine N-Methyltransferase , Humans , Inverted Repeat Sequences/genetics , Nucleic Acid Conformation
8.
Cell Stem Cell ; 10(5): 484-5, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22560069

ABSTRACT

Growing mouse pluripotent stem cells in defined media with signaling inhibitors establishes a naive ground state different from that of cells grown in serum. Recently in Cell, Marks et al. (2012) reported differences in the transcriptional and epigenomic landscapes of naive and serum-exposed mouse stem cells that improve our understanding of the pluripotent ground state.

9.
Cell ; 147(3): 565-76, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22036565

ABSTRACT

Transforming growth factor beta (TGF-ß) signaling, mediated through the transcription factors Smad2 and Smad3 (Smad2/3), directs different responses in different cell types. Here we report that Smad3 co-occupies the genome with cell-type-specific master transcription factors. Thus, Smad3 occupies the genome with Oct4 in embryonic stem cells (ESCs), Myod1 in myotubes, and PU.1 in pro-B cells. We find that these master transcription factors are required for Smad3 occupancy and that TGF-ß signaling largely affects the genes bound by the master transcription factors. Furthermore, we show that induction of Myod1 in nonmuscle cells is sufficient to redirect Smad3 to Myod1 sites. We conclude that cell-type-specific master transcription factors determine the genes bound by Smad2/3 and are thus responsible for orchestrating the cell-type-specific effects of TGF-ß signaling.


Subject(s)
Signal Transduction , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Embryonic Stem Cells , Enhancer Elements, Genetic , Humans , Mice , MyoD Protein/metabolism , Octamer Transcription Factor-3/metabolism , Smad3 Protein/metabolism
10.
Epigenomics ; 3(3): 323-43, 2011 Jun.
Article in English | MEDLINE | ID: mdl-22122341

ABSTRACT

Embryonic stem cells (ESCs) have the potential to generate virtually any cell type or tissue type in the body. This remarkable plasticity has yielded great interest in using these cells to understand early development and in treating human disease. In an effort to understand the basis of ESC pluripotency, genetic and genomic studies have revealed transcriptional regulatory circuitry that maintains the pluripotent cell state and poises the genome for downstream activation. Critical components of this circuitry include ESC transcription factors, chromatin regulators, histone modifications, signaling molecules and regulatory RNAs. This article will focus on our current understanding of these components and how they influence ESC and induced pluripotent stem cell states. Emerging themes include regulation of the pluripotent genome by a core set of transcription factors, transcriptional poising of developmental genes by chromatin regulatory complexes and the establishment of multiple layers of repression at key genomic loci.


Subject(s)
Chromatin/metabolism , Embryonic Stem Cells/physiology , Epigenesis, Genetic/physiology , Models, Biological , Pluripotent Stem Cells/physiology , Signal Transduction/physiology , Transcription Factors/metabolism , Embryonic Stem Cells/cytology , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Humans , Pluripotent Stem Cells/cytology , Regulatory Sequences, Ribonucleic Acid/genetics
11.
PLoS One ; 6(8): e22416, 2011.
Article in English | MEDLINE | ID: mdl-21886766

ABSTRACT

A surprising portion of both mammalian and Drosophila genomes are transcriptionally paused, undergoing initiation without elongation. We tested the hypothesis that transcriptional pausing is an obligate transition state between definitive activation and silencing as human embryonic stem cells (hESCs) change state from pluripotency to mesoderm. Chromatin immunoprecipitation for trimethyl lysine 4 on histone H3 (ChIP-Chip) was used to analyze transcriptional initiation, and 3' transcript arrays were used to determine transcript elongation. Pluripotent and mesodermal cells had equivalent fractions of the genome in active and paused transcriptional states (∼48% each), with ∼4% definitively silenced (neither initiation nor elongation). Differentiation to mesoderm changed the transcriptional state of 12% of the genome, with roughly equal numbers of genes moving toward activation or silencing. Interestingly, almost all loci (98-99%) changing transcriptional state do so either by entering or exiting the paused state. A majority of these transitions involve either loss of initiation, as genes specifying alternate lineages are archived, or gain of initiation, in anticipation of future full-length expression. The addition of chromatin dynamics permitted much earlier predictions of final cell fate compared to sole use of conventional transcript arrays. These findings indicate that the paused state may be the major transition state for genes changing expression during differentiation, and implicate control of transcriptional elongation as a key checkpoint in lineage specification.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Gene Silencing , Transcription, Genetic , Transcriptional Activation/genetics , Animals , Cell Line , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Genetic Loci/genetics , Genome, Human/genetics , Humans , Mice , Models, Genetic , Open Reading Frames/genetics
12.
Cell Stem Cell ; 7(2): 249-57, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20682450

ABSTRACT

Knowledge of both the global chromatin structure and the gene expression programs of human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) should provide a robust means to assess whether the genomes of these cells have similar pluripotent states. Recent studies have suggested that ESCs and iPSCs represent different pluripotent states with substantially different gene expression profiles. We describe here a comparison of global chromatin structure and gene expression data for a panel of human ESCs and iPSCs. Genome-wide maps of nucleosomes with histone H3K4me3 and H3K27me3 modifications indicate that there is little difference between ESCs and iPSCs with respect to these marks. Gene expression profiles confirm that the transcriptional programs of ESCs and iPSCs show very few consistent differences. Although some variation in chromatin structure and gene expression was observed in these cell lines, these variations did not serve to distinguish ESCs from iPSCs.


Subject(s)
Chromatin/chemistry , Chromatin/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Induced Pluripotent Stem Cells/metabolism , Cell Line , Chromatin Assembly and Disassembly/genetics , Cluster Analysis , Genome, Human/genetics , Histones/metabolism , Humans
14.
Cell ; 141(5): 872-83, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20471072

ABSTRACT

The presence of two active X chromosomes (XaXa) is a hallmark of the ground state of pluripotency specific to murine embryonic stem cells (ESCs). Human ESCs (hESCs) invariably exhibit signs of X chromosome inactivation (XCI) and are considered developmentally more advanced than their murine counterparts. We describe the establishment of XaXa hESCs derived under physiological oxygen concentrations. Using these cell lines, we demonstrate that (1) differentiation of hESCs induces random XCI in a manner similar to murine ESCs, (2) chronic exposure to atmospheric oxygen is sufficient to induce irreversible XCI with minor changes of the transcriptome, (3) the Xa exhibits heavy methylation of the XIST promoter region, and (4) XCI is associated with demethylation and transcriptional activation of XIST along with H3K27-me3 deposition across the Xi. These findings indicate that the human blastocyst contains pre-X-inactivation cells and that this state is preserved in vitro through culture under physiological oxygen.


Subject(s)
Chromosomes, Human, X/metabolism , Embryonic Stem Cells/metabolism , Oxygen/metabolism , X Chromosome Inactivation , Animals , Cell Differentiation , Female , Histones/metabolism , Humans , Karyotyping , Male , Mice , Oxidative Stress , Pluripotent Stem Cells/metabolism
15.
Cell ; 134(3): 521-33, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18692474

ABSTRACT

MicroRNAs (miRNAs) are crucial for normal embryonic stem (ES) cell self-renewal and cellular differentiation, but how miRNA gene expression is controlled by the key transcriptional regulators of ES cells has not been established. We describe here the transcriptional regulatory circuitry of ES cells that incorporates protein-coding and miRNA genes based on high-resolution ChIP-seq data, systematic identification of miRNA promoters, and quantitative sequencing of short transcripts in multiple cell types. We find that the key ES cell transcription factors are associated with promoters for miRNAs that are preferentially expressed in ES cells and with promoters for a set of silent miRNA genes. This silent set of miRNA genes is co-occupied by Polycomb group proteins in ES cells and shows tissue-specific expression in differentiated cells. These data reveal how key ES cell transcription factors promote the ES cell miRNA expression program and integrate miRNAs into the regulatory circuitry controlling ES cell identity.


Subject(s)
Embryonic Stem Cells/metabolism , MicroRNAs/genetics , Transcription, Genetic , Animals , Mice , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Transcription Factors/metabolism
16.
Genes Dev ; 22(24): 3403-8, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19141473

ABSTRACT

Mixed-lineage leukemia (MLL) fusion proteins are potent inducers of leukemia, but how these proteins generate aberrant gene expression programs is poorly understood. Here we show that the MLL-AF4 fusion protein occupies developmental regulatory genes important for hematopoietic stem cell identity and self-renewal in human leukemia cells. These MLL-AF4-bound regions have grossly altered chromatin structure, with histone modifications catalyzed by trithorax group proteins and DOT1 extending across large domains. Our results define direct targets of the MLL fusion protein, reveal the global role of epigenetic misregulation in leukemia, and identify new targets for therapeutic intervention in cancer.


Subject(s)
Cell Differentiation/genetics , Chromatin/genetics , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/physiology , Leukemia/genetics , Cell Line , Hematopoietic Stem Cells/cytology , Humans , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
17.
Cell ; 130(1): 77-88, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17632057

ABSTRACT

We describe the results of a genome-wide analysis of human cells that suggests that most protein-coding genes, including most genes thought to be transcriptionally inactive, experience transcription initiation. We found that nucleosomes with H3K4me3 and H3K9,14Ac modifications, together with RNA polymerase II, occupy the promoters of most protein-coding genes in human embryonic stem cells. Only a subset of these genes produce detectable full-length transcripts and are occupied by nucleosomes with H3K36me3 modifications, a hallmark of elongation. The other genes experience transcription initiation but show no evidence of elongation, suggesting that they are predominantly regulated at postinitiation steps. Genes encoding most developmental regulators fall into this group. Our results also identify a class of genes that are excluded from experiencing transcription initiation, at which mechanisms that prevent initiation must predominate. These observations extend to differentiated cells, suggesting that transcription initiation at most genes is a general phenomenon in human cells.


Subject(s)
Chromatin/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Transcription Initiation Site , Transcription, Genetic , Cell Differentiation , Cells, Cultured , Chromatin/genetics , Chromatin Immunoprecipitation , DNA Methylation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Humans , Lysine/metabolism , Multigene Family , Nucleosomes/metabolism , Oligonucleotide Array Sequence Analysis
18.
Cell ; 125(2): 301-13, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16630818

ABSTRACT

Polycomb group proteins are essential for early development in metazoans, but their contributions to human development are not well understood. We have mapped the Polycomb Repressive Complex 2 (PRC2) subunit SUZ12 across the entire nonrepeat portion of the genome in human embryonic stem (ES) cells. We found that SUZ12 is distributed across large portions of over two hundred genes encoding key developmental regulators. These genes are occupied by nucleosomes trimethylated at histone H3K27, are transcriptionally repressed, and contain some of the most highly conserved noncoding elements in the genome. We found that PRC2 target genes are preferentially activated during ES cell differentiation and that the ES cell regulators OCT4, SOX2, and NANOG cooccupy a significant subset of these genes. These results indicate that PRC2 occupies a special set of developmental genes in ES cells that must be repressed to maintain pluripotency and that are poised for activation during ES cell differentiation.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation, Developmental , Stem Cells/physiology , Animals , Carrier Proteins/genetics , Cells, Cultured , Gene Expression Profiling , Humans , Multiprotein Complexes , Neoplasm Proteins , Nuclear Proteins , Oligonucleotide Array Sequence Analysis , Polycomb Repressive Complex 2 , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Signal Transduction/physiology , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
19.
Cell ; 122(6): 947-56, 2005 Sep 23.
Article in English | MEDLINE | ID: mdl-16153702

ABSTRACT

The transcription factors OCT4, SOX2, and NANOG have essential roles in early development and are required for the propagation of undifferentiated embryonic stem (ES) cells in culture. To gain insights into transcriptional regulation of human ES cells, we have identified OCT4, SOX2, and NANOG target genes using genome-scale location analysis. We found, surprisingly, that OCT4, SOX2, and NANOG co-occupy a substantial portion of their target genes. These target genes frequently encode transcription factors, many of which are developmentally important homeodomain proteins. Our data also indicate that OCT4, SOX2, and NANOG collaborate to form regulatory circuitry consisting of autoregulatory and feedforward loops. These results provide new insights into the transcriptional regulation of stem cells and reveal how OCT4, SOX2, and NANOG contribute to pluripotency and self-renewal.


Subject(s)
Cell Transplantation/physiology , Embryo, Mammalian/cytology , Gene Expression Regulation, Developmental/physiology , Genes, Regulator/physiology , Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , DNA-Binding Proteins/metabolism , Genes, Regulator/genetics , HMGB Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Oligonucleotide Array Sequence Analysis/methods , Promoter Regions, Genetic , Protein Binding , SOXB1 Transcription Factors , Signal Transduction/physiology , Stem Cells/cytology , Transcription Factors/metabolism
20.
Proc Natl Acad Sci U S A ; 102(24): 8603-8, 2005 Jun 14.
Article in English | MEDLINE | ID: mdl-15941828

ABSTRACT

The mixed-lineage leukemia (MLL1/ALL-1/HRX) histone methyltransferase is involved in the epigenetic maintenance of transcriptional memory and the pathogenesis of human leukemias. To understand its role in cell type specification, we determined the human genomic binding sites of MLL1. We found that MLL1 functions as a human equivalent of yeast Set1. Like Set1, MLL1 localizes with RNA polymerase II (Pol II) to the 5' end of actively transcribed genes, where histone H3 lysine 4 trimethylation occurs. Consistent with this global role in transcription, MLL1 also localizes to microRNA (miRNA) loci that are involved in leukemia and hematopoiesis. In contrast to the 5' proximal binding behavior at most protein-coding genes, MLL1 occupies an extensive domain within a transcriptionally active region of the HoxA cluster. The ability of MLL1 to serve as a start site-specific global transcriptional regulator and to participate in larger chromatin domains at the Hox genes reveals dual roles for MLL1 in maintenance of cellular identity.


Subject(s)
Chromosomes, Human, Pair 7/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/genetics , Genes, Regulator/genetics , Genome, Human , Histone-Lysine N-Methyltransferase/metabolism , Transcription Factors/metabolism , Chromatin Immunoprecipitation , Chromosomes, Human, Pair 7/genetics , DNA-Binding Proteins/genetics , Genes, Homeobox/genetics , Genomics/methods , Histone Methyltransferases , Humans , MicroRNAs/metabolism , Myeloid-Lymphoid Leukemia Protein , Oligonucleotide Array Sequence Analysis , Protein Methyltransferases , Proto-Oncogenes/genetics , RNA Polymerase II/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...