Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Struct Mol Biol ; 30(2): 148-158, 2023 02.
Article in English | MEDLINE | ID: mdl-36747093

ABSTRACT

Enhancer activation serves as the main mechanism regulating signal-dependent transcriptional programs, ensuring cellular plasticity, yet central questions persist regarding their mechanism of activation. Here, by successfully mapping topoisomerase I-DNA covalent complexes genome-wide, we find that most, if not all, acutely activated enhancers, including those induced by 17ß-estradiol, dihydrotestosterone, tumor necrosis factor alpha and neuronal depolarization, are hotspots for topoisomerase I-DNA covalent complexes, functioning as epigenomic signatures read by the classic DNA damage sensor protein, Ku70. Ku70 in turn nucleates a heterochromatin protein 1 gamma (HP1γ)-mediator subunit Med26 complex to facilitate acute, but not chronic, transcriptional activation programs. Together, our data uncover a broad, unappreciated transcriptional code, required for most, if not all, acute signal-dependent enhancer activation events in both mitotic and postmitotic cells.


Subject(s)
DNA Topoisomerases, Type I , Enhancer Elements, Genetic , DNA , DNA Topoisomerases, Type I/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Ku Autoantigen/metabolism
2.
Circulation ; 142(22): 2138-2154, 2020 12.
Article in English | MEDLINE | ID: mdl-32933333

ABSTRACT

BACKGROUND: Concentric and eccentric cardiac hypertrophy are associated with pressure and volume overload, respectively, in cardiovascular disease both conferring an increased risk of heart failure. These contrasting forms of hypertrophy are characterized by asymmetrical growth of the cardiac myocyte in mainly width or length, respectively. The molecular mechanisms determining myocyte preferential growth in width versus length remain poorly understood. Identification of the mechanisms governing asymmetrical myocyte growth could provide new therapeutic targets for the prevention or treatment of heart failure. METHODS: Primary adult rat ventricular myocytes, adeno-associated virus (AAV)-mediated gene delivery in mice, and human tissue samples were used to define a regulatory pathway controlling pathological myocyte hypertrophy. Chromatin immunoprecipitation assays with sequencing and precision nuclear run-on sequencing were used to define a transcriptional mechanism. RESULTS: We report that asymmetrical cardiac myocyte hypertrophy is modulated by SRF (serum response factor) phosphorylation, constituting an epigenomic switch balancing the growth in width versus length of adult ventricular myocytes in vitro and in vivo. SRF Ser103 phosphorylation is bidirectionally regulated by RSK3 (p90 ribosomal S6 kinase type 3) and PP2A (protein phosphatase 2A) at signalosomes organized by the scaffold protein mAKAPß (muscle A-kinase anchoring protein ß), such that increased SRF phosphorylation activates AP-1 (activator protein-1)-dependent enhancers that direct myocyte growth in width. AAV are used to express in vivo mAKAPß-derived RSK3 and PP2A anchoring disruptor peptides that block the association of the enzymes with the mAKAPß scaffold. Inhibition of RSK3 signaling prevents concentric cardiac remodeling induced by pressure overload, while inhibition of PP2A signaling prevents eccentric cardiac remodeling induced by myocardial infarction, in each case improving cardiac function. SRF Ser103 phosphorylation is significantly decreased in dilated human hearts, supporting the notion that modulation of the mAKAPß-SRF signalosome could be a new therapeutic approach for human heart failure. CONCLUSIONS: We have identified a new molecular switch, namely mAKAPß signalosome-regulated SRF phosphorylation, that controls a transcriptional program responsible for modulating changes in cardiac myocyte morphology that occur secondary to pathological stressors. Complementary AAV-based gene therapies constitute rationally-designed strategies for a new translational modality for heart failure.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Enlargement , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Serum Response Factor/metabolism , A Kinase Anchor Proteins/genetics , Adenoviridae/genetics , Animals , Animals, Newborn , Cells, Cultured , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Heart Failure/genetics , Heart Failure/pathology , Humans , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/pathology , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley
3.
Cell Rep ; 31(12): 107803, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32579929

ABSTRACT

The function of enhancer RNAs (eRNAs) in transcriptional regulation remains obscure. By analyzing the genome-wide nascent transcript profiles in breast cancer cells, we identify a special group of eRNAs that are essential for estrogen-induced transcriptional repression. Using eRNAs of TM4SF1 and EFEMP1 as the paradigms, we find that these RNA molecules not only stabilize promoter-enhancer interactions but also recruit liganded estrogen receptor α (ERα) to particular enhancer regions, facilitate the formation of a functional transcriptional complex, and cause gene silencing. Interestingly, ERα is shown to directly bind with eRNAs by its DNA-binding domain. These eRNAs help with the formation of a specific ERα-centered transcriptional complex and promote the association of the histone demethylase KDM2A, which dismisses RNA polymerase II from designated enhancers and suppresses the transcription of target genes. Our work demonstrates a complete mechanism underlying the action of eRNAs in modulating and refining the locus-specific transcriptional program.


Subject(s)
Enhancer Elements, Genetic , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , RNA/metabolism , Cell Line , Down-Regulation/genetics , Estrogen Receptor alpha/chemistry , F-Box Proteins/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Models, Biological , Open Reading Frames/genetics , Protein Binding , Protein Domains , RNA Polymerase II/metabolism , Transcription, Genetic
4.
Nat Struct Mol Biol ; 26(3): 193-203, 2019 03.
Article in English | MEDLINE | ID: mdl-30833784

ABSTRACT

A crucial feature of differentiated cells is the rapid activation of enhancer-driven transcriptional programs in response to signals. The potential contributions of physicochemical properties of enhancer assembly in signaling events remain poorly understood. Here we report that in human breast cancer cells, the acute 17ß-estradiol-dependent activation of functional enhancers requires assembly of an enhancer RNA-dependent ribonucleoprotein (eRNP) complex exhibiting properties of phase-separated condensates. Unexpectedly, while acute ligand-dependent assembly of eRNPs resulted in enhancer activation sensitive to chemical disruption of phase separation, chronically activated enhancers proved resistant to such disruption, with progressive maturation of eRNPs to a more gel-like state. Acute, but not chronic, stimulation resulted in ligand-induced, condensin-dependent changes in spatial chromatin conformation based on homotypic enhancer association, resulting in cooperative enhancer-activation events. Thus, distinct physicochemical properties of eRNP condensates on enhancers serve as determinants of rapid ligand-dependent alterations in chromosomal architecture and cooperative enhancer activation.


Subject(s)
Enhancer Elements, Genetic/genetics , Estradiol/metabolism , Ribonucleoproteins/metabolism , Transcriptional Activation/physiology , Cell Line, Tumor , Chromatin , Chromosomes/physiology , Humans , MCF-7 Cells , Protein Conformation , Transcription, Genetic/genetics , Transcriptional Activation/genetics
5.
Mol Cell ; 71(4): 526-539.e8, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30118678

ABSTRACT

Nuclear receptors induce both transcriptional activation and repression programs responsible for development, homeostasis, and disease. Here, we report a previously overlooked enhancer decommissioning strategy underlying a large estrogen receptor alpha (ERα)-dependent transcriptional repression program. The unexpected signature for this E2-induced program resides in indirect recruitment of ERα to a large cohort of pioneer factor basally active FOXA1-bound enhancers that lack cognate ERα DNA-binding elements. Surprisingly, these basally active estrogen-repressed (BAER) enhancers are decommissioned by ERα-dependent recruitment of the histone demethylase KDM2A, functioning independently of its demethylase activity. Rather, KDM2A tethers the E3 ubiquitin-protein ligase NEDD4 to ubiquitylate/dismiss Pol II to abrogate eRNA transcription, with consequent target gene downregulation. Thus, our data reveal that Pol II ubiquitylation/dismissal may serve as a potentially broad strategy utilized by indirectly bound nuclear receptors to abrogate large programs of pioneer factor-mediated, eRNA-producing enhancers.


Subject(s)
Enhancer Elements, Genetic , Estrogen Receptor alpha/genetics , F-Box Proteins/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Nedd4 Ubiquitin Protein Ligases/genetics , RNA Polymerase II/genetics , Base Sequence , Binding Sites , CRISPR-Cas Systems , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , F-Box Proteins/metabolism , Gene Editing/methods , Gene Expression Regulation/drug effects , HEK293 Cells , Hepatocyte Nuclear Factor 3-alpha/metabolism , Histones/genetics , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , MCF-7 Cells , Nedd4 Ubiquitin Protein Ligases/metabolism , Protein Binding , RNA/genetics , RNA/metabolism , RNA Polymerase II/metabolism , Signal Transduction , Transcription, Genetic/drug effects , Ubiquitination/drug effects
6.
Nature ; 556(7702): 510-514, 2018 04.
Article in English | MEDLINE | ID: mdl-29670286

ABSTRACT

Enhancers for embryonic stem (ES) cell-expressed genes and lineage-determining factors are characterized by conventional marks of enhancer activation in ES cells1-3, but it remains unclear whether enhancers destined to regulate cell-type-restricted transcription units might also have distinct signatures in ES cells. Here we show that cell-type-restricted enhancers are 'premarked' and activated as transcription units by the binding of one or two ES cell transcription factors, although they do not exhibit traditional enhancer epigenetic marks in ES cells, thus uncovering the initial temporal origins of cell-type-restricted enhancers. This premarking is required for future cell-type-restricted enhancer activity in the differentiated cells, with the strength of the ES cell signature being functionally important for the subsequent robustness of cell-type-restricted enhancer activation. We have experimentally validated this model in macrophage-restricted enhancers and neural precursor cell (NPC)-restricted enhancers using ES cell-derived macrophages or NPCs, edited to contain specific ES cell transcription factor motif deletions. DNA hydroxyl-methylation of enhancers in ES cells, determined by ES cell transcription factors, may serve as a potential molecular memory for subsequent enhancer activation in mature macrophages. These findings suggest that the massive repertoire of cell-type-restricted enhancers are essentially hierarchically and obligatorily premarked by binding of a defining ES cell transcription factor in ES cells, dictating the robustness of enhancer activation in mature cells.


Subject(s)
Cell Differentiation/genetics , Enhancer Elements, Genetic , Gene Expression Regulation/genetics , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Epigenesis, Genetic , Female , Macrophages/cytology , Macrophages/metabolism , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Organ Specificity , Pluripotent Stem Cells/cytology , Reproducibility of Results
7.
Cell Metab ; 25(5): 1160-1175.e11, 2017 May 02.
Article in English | MEDLINE | ID: mdl-28467932

ABSTRACT

Pancreatic ß cell mass for appropriate blood glucose control is established during early postnatal life. ß cell proliferative capacity declines postnatally, but the extrinsic cues and intracellular signals that cause this decline remain unknown. To obtain a high-resolution map of ß cell transcriptome dynamics after birth, we generated single-cell RNA-seq data of ß cells from multiple postnatal time points and ordered cells based on transcriptional similarity using a new analytical tool. This analysis captured signatures of immature, proliferative ß cells and established high expression of amino acid metabolic, mitochondrial, and Srf/Jun/Fos transcription factor genes as their hallmark feature. Experimental validation revealed high metabolic activity in immature ß cells and a role for reactive oxygen species and Srf/Jun/Fos transcription factors in driving postnatal ß cell proliferation and mass expansion. Our work provides the first high-resolution molecular characterization of state changes in postnatal ß cells and paves the way for the identification of novel therapeutic targets to stimulate ß cell regeneration.


Subject(s)
Cell Proliferation , Insulin-Secreting Cells/cytology , Metabolic Networks and Pathways , Transcriptome , Amino Acids/genetics , Amino Acids/metabolism , Animals , Cells, Cultured , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Insulin-Secreting Cells/metabolism , Male , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
8.
Mol Cell ; 66(3): 321-331.e6, 2017 May 04.
Article in English | MEDLINE | ID: mdl-28475868

ABSTRACT

The molecular mechanisms underlying the opposing functions of glucocorticoid receptors (GRs) and estrogen receptor α (ERα) in breast cancer development remain poorly understood. Here we report that, in breast cancer cells, liganded GR represses a large ERα-activated transcriptional program by binding, in trans, to ERα-occupied enhancers. This abolishes effective activation of these enhancers and their cognate target genes, and it leads to the inhibition of ERα-dependent binding of components of the MegaTrans complex. Consistent with the effects of SUMOylation on other classes of nuclear receptors, dexamethasone (Dex)-induced trans-repression of the estrogen E2 program appears to depend on GR SUMOylation, which leads to stable trans-recruitment of the GR-N-CoR/SMRT-HDAC3 corepressor complex on these enhancers. Together, these results uncover a mechanism by which competitive recruitment of DNA-binding nuclear receptors/transcription factors in trans to hot spot enhancers serves as an effective biological strategy for trans-repression, with clear implications for breast cancer and other diseases.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Receptor Cross-Talk , Receptors, Glucocorticoid/metabolism , Transcription, Genetic , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Dexamethasone/pharmacology , Down-Regulation , Enhancer Elements, Genetic , Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , MCF-7 Cells , Multiprotein Complexes , Mutation , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Protein Binding , RNA Interference , Receptor Cross-Talk/drug effects , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/genetics , Signal Transduction , Sumoylation , Transcription, Genetic/drug effects , Transcriptome , Transfection
9.
Nat Neurosci ; 18(9): 1256-64, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26214369

ABSTRACT

We found that a neuron-specific isoform of LSD1, LSD1n, which results from an alternative splicing event, acquires a new substrate specificity, targeting histone H4 Lys20 methylation, both in vitro and in vivo. Selective genetic ablation of LSD1n led to deficits in spatial learning and memory, revealing the functional importance of LSD1n in neuronal activity-regulated transcription that is necessary for long-term memory formation. LSD1n occupied neuronal gene enhancers, promoters and transcribed coding regions, and was required for transcription initiation and elongation steps in response to neuronal activity, indicating the crucial role of H4K20 methylation in coordinating gene transcription with neuronal function. Our results indicate that this alternative splicing of LSD1 in neurons, which was associated with altered substrate specificity, serves as a mechanism acquired by neurons to achieve more precise control of gene expression in the complex processes underlying learning and memory.


Subject(s)
Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Memory, Long-Term/physiology , Transcription, Genetic/physiology , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Female , Gene Deletion , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
10.
Cell ; 160(3): 367-80, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25619691

ABSTRACT

The discovery that enhancers are regulated transcription units, encoding eRNAs, has raised new questions about the mechanisms of their activation. Here, we report an unexpected molecular mechanism that underlies ligand-dependent enhancer activation, based on DNA nicking to relieve torsional stress from eRNA synthesis. Using dihydrotestosterone (DHT)-induced binding of androgen receptor (AR) to prostate cancer cell enhancers as a model, we show rapid recruitment, within minutes, of DNA topoisomerase I (TOP1) to a large cohort of AR-regulated enhancers. Furthermore, we show that the DNA nicking activity of TOP1 is a prerequisite for robust eRNA synthesis and enhancer activation and is kinetically accompanied by the recruitment of ATR and the MRN complex, followed by additional components of DNA damage repair machinery to the AR-regulated enhancers. Together, our studies reveal a linkage between eRNA synthesis and ligand-dependent TOP1-mediated nicking-a strategy exerting quantitative effects on eRNA expression in regulating AR-bound enhancer-dependent transcriptional programs.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , Receptors, Androgen/metabolism , Cell Line, Tumor , DNA Breaks, Single-Stranded , DNA Repair , DNA Topoisomerases, Type I/genetics , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Homeodomain Proteins/metabolism , Humans , MRE11 Homologue Protein , Transcription Factors/metabolism , Transcription, Genetic
11.
Proc Natl Acad Sci U S A ; 112(5): 1380-5, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25605944

ABSTRACT

Substantial evidence supports the hypothesis that enhancers are critical regulators of cell-type determination, orchestrating both positive and negative transcriptional programs; however, the basic mechanisms by which enhancers orchestrate interactions with cognate promoters during activation and repression events remain incompletely understood. Here we report the required actions of LIM domain-binding protein 1 (LDB1)/cofactor of LIM homeodomain protein 2/nuclear LIM interactor, interacting with the enhancer-binding protein achaete-scute complex homolog 1, to mediate looping to target gene promoters and target gene regulation in corticotrope cells. LDB1-mediated enhancer:promoter looping appears to be required for both activation and repression of these target genes. Although LDB1-dependent activated genes are regulated at the level of transcriptional initiation, the LDB1-dependent repressed transcription units appear to be regulated primarily at the level of promoter pausing, with LDB1 regulating recruitment of metastasis-associated 1 family, member 2, a component of the nucleosome remodeling deacetylase complex, on these negative enhancers, required for the repressive enhancer function. These results indicate that LDB1-dependent looping events can deliver repressive cargo to cognate promoters to mediate promoter pausing events in a pituitary cell type.


Subject(s)
Corticotrophs/physiology , DNA-Binding Proteins/physiology , Enhancer Elements, Genetic , LIM Domain Proteins/physiology , Promoter Regions, Genetic , Animals , Cell Line , DNA-Binding Proteins/metabolism , LIM Domain Proteins/metabolism , Mice , Mice, Knockout
12.
Nature ; 516(7530): 267-71, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25252977

ABSTRACT

Post-translational histone modifications have a critical role in regulating transcription, the cell cycle, DNA replication and DNA damage repair. The identification of new histone modifications critical for transcriptional regulation at initiation, elongation or termination is of particular interest. Here we report a new layer of regulation in transcriptional elongation that is conserved from yeast to mammals. This regulation is based on the phosphorylation of a highly conserved tyrosine residue, Tyr 57, in histone H2A and is mediated by the unsuspected tyrosine kinase activity of casein kinase 2 (CK2). Mutation of Tyr 57 in H2A in yeast or inhibition of CK2 activity impairs transcriptional elongation in yeast as well as in mammalian cells. Genome-wide binding analysis reveals that CK2α, the catalytic subunit of CK2, binds across RNA-polymerase-II-transcribed coding genes and active enhancers. Mutation of Tyr 57 causes a loss of H2B mono-ubiquitination as well as H3K4me3 and H3K79me3, histone marks associated with active transcription. Mechanistically, both CK2 inhibition and the H2A(Y57F) mutation enhance H2B deubiquitination activity of the Spt-Ada-Gcn5 acetyltransferase (SAGA) complex, suggesting a critical role of this phosphorylation in coordinating the activity of the SAGA complex during transcription. Together, these results identify a new component of regulation in transcriptional elongation based on CK2-dependent tyrosine phosphorylation of the globular domain of H2A.


Subject(s)
Casein Kinase II/metabolism , Histones/chemistry , Histones/metabolism , Transcription Elongation, Genetic , Tyrosine/metabolism , Amino Acid Sequence , Cell Line , Conserved Sequence , Histones/genetics , Humans , Molecular Sequence Data , Phosphorylation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Tyrosine/chemistry , Ubiquitination/genetics
13.
PLoS One ; 9(2): e90496, 2014.
Article in English | MEDLINE | ID: mdl-24587380

ABSTRACT

Cathepsin L, a lysosomal protein in mouse embryonic stem cells has been shown to clip the histone H3 N- terminus, an activity associated with gene activity during mouse cell development. Glutamate dehydrogenase (GDH) was also identified as histone H3 specific protease in chicken liver, which has been connected to gene expression during aging. In baker's yeast, Saccharomyces cerevisiae, clipping the histone H3 N-terminus has been associated with gene activation in stationary phase but the protease responsible for the yeast histone H3 endopeptidase activity had not been identified. In searching for a yeast histone H3 endopeptidase, we found that yeast vacuolar protein Prb1 is present in the cellular fraction enriched for the H3 N-terminus endopeptidase activity and this endopeptidase activity is lost in the PRB1 deletion mutant (prb1Δ). In addition, like Cathepsin L and GDH, purified Prb1 from yeast cleaves H3 between Lys23 and Ala24 in the N-terminus in vitro as shown by Edman degradation. In conclusion, our data argue that PRB1 is required for clipping of the histone H3 N-terminal tail in Saccharomyces cerevisiae.


Subject(s)
Endopeptidases/metabolism , Histones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Alanine/metabolism , Blotting, Western , Endopeptidases/genetics , Lysine/metabolism , Mass Spectrometry/methods , Mutation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
14.
Proc Natl Acad Sci U S A ; 110(28): 11493-8, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23798425

ABSTRACT

The presence of acetylated histone H3K56 (H3K56ac) in human ES cells (ESCs) correlates positively with the binding of Nanog, Sox2, and Oct4 (NSO) transcription factors at their target gene promoters. However, the function of H3K56ac there has been unclear. We now report that Oct4 interacts with H3K56ac in mouse ESC nuclear extracts and that perturbing H3K56 acetylation decreases Oct4-H3 binding. This interaction is likely to be direct because it can be recapitulated in vitro in an H3K56ac-dependent manner and is functionally important because H3K56ac combines with NSO factors in chromatin immunoprecipitation sequencing to mark the regions associated with pluripotency better than NSO alone. Moreover, reducing H3K56ac by short hairpin Asf1a decreases expression of pluripotency-related markers and increases expression of differentiation-related ones. Therefore, our data suggest that H3K56ac plays a central role in binding to Oct4 to promote the pluripotency of ESCs.


Subject(s)
Embryonic Stem Cells/cytology , Histones/metabolism , Octamer Transcription Factor-3/metabolism , Acetylation , Animals , Cells, Cultured , Embryonic Stem Cells/metabolism , Mice , Protein Binding
15.
PLoS One ; 6(1): e15336, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21283513

ABSTRACT

Somatic cells can be reprogrammed to a pluripotent state by over-expression of defined factors, and pluripotency has been confirmed by the tetraploid complementation assay. However, especially in human cells, estimating the quality of Induced Pluripotent Stem Cell(iPSC) is still difficult. Here, we present a novel supervised method for the assessment of the quality of iPSCs by estimating the gene expression profile using a 2-D "Differentiation-index coordinate", which consists of two "developing lines" that reflects the directions of ES cell differentiation and the changes of cell states during differentiation. By applying a novel liner model to describe the differentiation trajectory, we transformed the ES cell differentiation time-course expression profiles to linear "developing lines"; and use these lines to construct the 2-D "Differentiation-index coordinate" of mouse and human. We compared the published gene expression profiles of iPSCs, ESCs and fibroblasts in mouse and human "Differentiation-index coordinate". Moreover, we defined the Distance index to indicate the qualities of iPS cells, which based on the projection distance of iPSCs-ESCs and iPSCs-fibroblasts. The results indicated that the "Differentiation-index coordinate" can distinguish differentiation states of the different cells types. Furthermore, by applying this method to the analysis of expression profiles in the tetraploid complementation assay, we showed that the Distance index which reflected spatial distributions correlated the pluripotency of iPSCs. We also analyzed the significantly changed gene sets of "developing lines". The results suggest that the method presented here is not only suitable for the estimation of the quality of iPS cells based on expression profiles, but also is a new approach to analyze time-resolved experimental data.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Gene Expression Profiling/methods , Induced Pluripotent Stem Cells/cytology , Animals , Cell Lineage , Evaluation Studies as Topic , Fibroblasts/cytology , Humans , Mice , Time Factors
16.
BMC Mol Biol ; 10: 12, 2009 Feb 22.
Article in English | MEDLINE | ID: mdl-19232136

ABSTRACT

BACKGROUND: Small noncoding RNAs (ncRNAs), including short interfering RNAs (siRNAs) and microRNAs (miRNAs), can silence genes at the transcriptional, post-transcriptional or translational level 12. RESULTS: Here, we show that microRNA-10a (miR-10a) targets a homologous DNA region in the promoter region of the hoxd4 gene and represses its expression at the transcriptional level. Mutational analysis of the miR-10a sequence revealed that the 3' end of the miRNA sequence is the most critical element for the silencing effect. MicroRNA-10a-induced transcriptional gene inhibition requires the presence of Dicer and Argonautes 1 and 3, and it is related to promoter associated noncoding RNAs. Bisulfite sequencing analysis showed that the reduced hoxd4 expression was accompanied by de novo DNA methylation at the hoxd4 promoter. We further demonstrated that trimethylation of histone 3 lysine 27 (H3K27me3) is involved in the miR-10a-induced hoxd4 transcriptional gene silence. CONCLUSION: In conclusion, our results demonstrate that miR-10a can regulate human gene expression in a transcriptional manner, and indicate that endogenous small noncoding RNA-induced control of transcription may be a potential system for expressional regulation in human breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Argonaute Proteins , Cell Line, Tumor , DNA Methylation , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factors/metabolism , HeLa Cells , Humans , Promoter Regions, Genetic , Ribonuclease III/metabolism
17.
Yi Chuan Xue Bao ; 32(4): 434-41, 2005 Apr.
Article in Chinese | MEDLINE | ID: mdl-16011036

ABSTRACT

RNA interference (RNAi), a process of sequence-specific gene silence,can effectively and specifically suppress the activity of corresponding mRNAs in a gene-dependent manner induced by double-stranded RNA (dsRNA). This powerful technology has been widely employed to manipulate gene expression in mammalian and human cells, elucidate signal transduction pathways and identify gene functions in a whole-genome scale. Simultaneously, it also displays a bright and fascinating future in the research and development of RNAi-based drugs for various diseases such as viral infections, cancers, metabolic disorders and genetic diseases. In present review, we attempt to recapitulate the application of this breakthrough technology in establishing gene-deficient models and show the alluring foreground of RNAi-based gene therapy in these diseases.


Subject(s)
Genetic Therapy/methods , RNA Interference , RNA, Small Interfering/genetics , Animals , Fragile X Syndrome/therapy , Humans , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...