Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cell ; 154(3): 530-40, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23911320

ABSTRACT

To mechanistically characterize the microevolutionary processes active in altering transcription factor (TF) binding among closely related mammals, we compared the genome-wide binding of three tissue-specific TFs that control liver gene expression in six rodents. Despite an overall fast turnover of TF binding locations between species, we identified thousands of TF regions of highly constrained TF binding intensity. Although individual mutations in bound sequence motifs can influence TF binding, most binding differences occur in the absence of nearby sequence variations. Instead, combinatorial binding was found to be significant for genetic and evolutionary stability; cobound TFs tend to disappear in concert and were sensitive to genetic knockout of partner TFs. The large, qualitative differences in genomic regions bound between closely related mammals, when contrasted with the smaller, quantitative TF binding differences among Drosophila species, illustrate how genome structure and population genetics together shape regulatory evolution.


Subject(s)
Evolution, Molecular , Mice/classification , Mice/genetics , Transcription Factors/genetics , Animals , Drosophila/genetics , Liver/metabolism , Mice/metabolism , Mice, Inbred Strains , Mice, Knockout , Rats/genetics , Transcription Factors/metabolism
2.
EMBO J ; 40(1): e102236, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33034061

ABSTRACT

The generation of induced pluripotent stem cells (iPSCs) from somatic cells provides an excellent model to study mechanisms of transcription factor-induced global alterations of the epigenome and genome function. Here, we have investigated the early transcriptional events of cellular reprogramming triggered by the co-expression of Oct4, Sox2, Klf4, and c-Myc (OSKM) in mouse embryonic fibroblasts (MEFs) and mouse hepatocytes (mHeps). In this analysis, we identified a gene regulatory network composed of nine transcriptional regulators (9TR; Cbfa2t3, Gli2, Irf6, Nanog, Ovol1, Rcan1, Taf1c, Tead4, and Tfap4), which are directly targeted by OSKM, in vivo. Functional studies using single and double shRNA knockdowns of any of these factors caused disruption of the network and dramatic reductions in reprogramming efficiency, indicating that this network is essential for the induction and establishment of pluripotency. We demonstrate that the stochastic co-expression of 9TR network components occurs in a remarkably small number of cells, approximating the percentage of terminally reprogrammed cells as a result of dynamic molecular events. Thus, the early DNA-binding patterns of OSKM and the subsequent probabilistic co-expression of essential 9TR components in subpopulations of cells undergoing reprogramming steer the reconstruction of a gene regulatory network marking the transition to pluripotency.


Subject(s)
Cellular Reprogramming/genetics , Fibroblasts/physiology , Gene Regulatory Networks/genetics , Hepatocytes/physiology , Induced Pluripotent Stem Cells/physiology , Animals , Embryonic Stem Cells/physiology , Female , HEK293 Cells , Humans , Kruppel-Like Factor 4 , Mice , Mice, Inbred C57BL , Pregnancy , Transcription Factors/genetics , Transcription, Genetic/genetics
3.
EMBO J ; 34(4): 430-47, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25515659

ABSTRACT

PR-SET7-mediated histone 4 lysine 20 methylation has been implicated in mitotic condensation, DNA damage response and replication licensing. Here, we show that PR-SET7 function in the liver is pivotal for maintaining genome integrity. Hepatocyte-specific deletion of PR-SET7 in mouse embryos resulted in G2 phase arrest followed by massive cell death and defect in liver organogenesis. Inactivation at postnatal stages caused cell duplication-dependent hepatocyte necrosis, accompanied by inflammation, fibrosis and compensatory growth induction of neighboring hepatocytes and resident ductal progenitor cells. Prolonged necrotic regenerative cycles coupled with oncogenic STAT3 activation led to the spontaneous development of hepatic tumors composed of cells with cancer stem cell characteristics. These include a capacity to self-renew in culture or in xenografts and the ability to differentiate to phenotypically distinct hepatic cells. Hepatocellular carcinoma in PR-SET7-deficient mice displays a cancer stem cell gene signature specified by the co-expression of ductal progenitor markers and oncofetal genes.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Neoplastic Stem Cells/enzymology , Animals , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/metabolism , Male , Methylation , Mice , Tumor Cells, Cultured
4.
Semin Cancer Biol ; 42: 70-80, 2017 02.
Article in English | MEDLINE | ID: mdl-27554136

ABSTRACT

SMYD3 is a member of the SET and MYND-domain family of methyl-transferases, the increased expression of which correlates with poor prognosis in various types of cancer. In liver and colon tumors, SMYD3 is localized in the nucleus, where it interacts with RNA Pol II and H3K4me3 and functions as a selective transcriptional amplifier of oncogenes and genes that control cell proliferation and metastatic spread. Smyd3 expression has a high discriminative power for the characterization of liver tumors and positively correlates with poor prognosis. In lung and pancreatic cancer, SMYD3 acts in the cytoplasm, potentiating oncogenic Ras/ERK signaling through the methylation of the MAP3K2 kinase and the subsequent release from its inhibitor. A clinico-pathological analysis of lung cancer patients uncovers prognostic significance of SMYD3 only for first progression survival. However, stratification of patients according to their smoking history significantly expands the prognostic value of SMYD3 to overall survival and other features, suggesting that smoking-related effects saturate the clinical analysis and mask the function of SMYD3 as an oncogenic potentiator.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Neoplasms/genetics , Prognosis , Carcinogenesis/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/biosynthesis , Humans , Neoplasms/pathology , RNA Polymerase II/genetics
5.
EMBO Rep ; 17(5): 769-79, 2016 05.
Article in English | MEDLINE | ID: mdl-26993089

ABSTRACT

Pioneering studies within the last few years have allowed the in vitro expansion of tissue-specific adult stem cells from a variety of endoderm-derived organs, including the stomach, small intestine, and colon. Expansion of these cells requires activation of the receptor Lgr5 by its ligand R-spondin 1 and is likely facilitated by the fact that in healthy adults the stem cells in these organs are highly proliferative. In many other adult organs, such as the liver, proliferating cells are normally not abundant in adulthood. However, upon injury, the liver has a strong regenerative potential that is accompanied by the emergence of Lgr5-positive stem cells; these cells can be isolated and expanded in vitro as organoids. In an effort to isolate stem cells from non-regenerating mouse livers, we discovered that healthy gallbladders are a rich source of stem/progenitor cells that can be propagated in culture as organoids for more than a year. Growth of these organoids was stimulated by R-spondin 1 and noggin, whereas in the absence of these growth factors, the organoids differentiated partially toward the hepatocyte fate. When transplanted under the liver capsule, gallbladder-derived organoids maintained their architecture for 2 weeks. Furthermore, single cells prepared from dissociated organoids and injected into the mesenteric vein populated the liver parenchyma of carbon tetrachloride-treated mice. Human gallbladders were also a source of organoid-forming stem cells. Thus, under specific growth conditions, stem cells can be isolated from healthy gallbladders, expanded almost indefinitely in vitro, and induced to differentiate toward the hepatocyte lineage.


Subject(s)
Carrier Proteins/metabolism , Gallbladder/cytology , Stem Cells/metabolism , Thrombospondins/metabolism , Animals , Biomarkers , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cell Differentiation/genetics , Cells, Cultured , Gene Expression Profiling , Humans , Liver/cytology , Mice , Mice, Transgenic , Organoids , Protein Kinase Inhibitors/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Stem Cells/drug effects , Thrombospondins/genetics , Thrombospondins/pharmacology , Transcriptome
6.
Mol Cell ; 39(1): 152-60, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20603083

ABSTRACT

Histone-modifying enzymes can regulate DNA damage-induced apoptosis through modulation of p53 function. Here, we show that, in p53-deficient tumor cells, Set9 and LSD1 regulate DNA damage-induced cell death in a manner opposite to that observed in p53(+/+) cells, via modulation of E2F1 stabilization. Set9 methylates E2F1 at lysine-185, which prevents E2F1 accumulation during DNA damage and activation of its proapoptotic target gene p73. This methyl mark is removed by LSD1, which is required for E2F1 stabilization and apoptotic function. The molecular mechanism involves crosstalks between lysine methylation and other covalent modifications that affect E2F1 stability. Methylation at lysine-185 inhibits acetylation and phosphorylation at distant positions and, in parallel, stimulates ubiquitination and degradation of the protein. The findings illustrate that the function of methyltransferases can have opposing biological outcomes depending on the specificity of transcription factor targets.


Subject(s)
E2F1 Transcription Factor/metabolism , Lysine/metabolism , Acetylation , Cell Death , Cell Line, Tumor , DNA Damage , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Humans , Methylation , Phosphorylation , Protein Stability , Tumor Suppressor Protein p53/metabolism , Ubiquitination
7.
Genome Res ; 24(11): 1797-807, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25122613

ABSTRACT

The genetic code is an abstraction of how mRNA codons and tRNA anticodons molecularly interact during protein synthesis; the stability and regulation of this interaction remains largely unexplored. Here, we characterized the expression of mRNA and tRNA genes quantitatively at multiple time points in two developing mouse tissues. We discovered that mRNA codon pools are highly stable over development and simply reflect the genomic background; in contrast, precise regulation of tRNA gene families is required to create the corresponding tRNA transcriptomes. The dynamic regulation of tRNA genes during development is controlled in order to generate an anticodon pool that closely corresponds to messenger RNAs. Thus, across development, the pools of mRNA codons and tRNA anticodons are invariant and highly correlated, revealing a stable molecular interaction interlocking transcription and translation.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Developmental , Liver/metabolism , RNA, Messenger/genetics , RNA, Transfer/genetics , Transcriptome , Animals , Anticodon/genetics , Base Sequence , Brain/embryology , Chromatin Immunoprecipitation/methods , Codon/genetics , Computer Simulation , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , High-Throughput Nucleotide Sequencing/methods , Liver/embryology , Male , Mice, Inbred C57BL , Models, Genetic , Open Reading Frames/genetics , Principal Component Analysis , RNA, Messenger/metabolism , RNA, Transfer/metabolism , Time Factors
8.
Biochim Biophys Acta ; 1851(1): 2-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24814048

ABSTRACT

Tight regulation of lipid levels is critical for cellular and organismal homeostasis, not only in terms of energy utilization and storage, but also to prevent potential toxicity. The liver utilizes a set of hepatic transcription factors to regulate the expression of genes implicated in all aspects of lipid metabolism including catabolism, transport, and synthesis. In this article, we will review the main transcriptional mechanisms regulating the expression of genes involved in hepatic lipid metabolism. The principal regulatory pathways are composed of simple modules of transcription factor crosstalks, which correspond to building blocks of more complex regulatory networks. These transcriptional networks contribute to the regulation of proper lipid homeostasis in parallel to posttranslational mechanisms and end product-mediated modulation of lipid metabolizing enzymes. This article is part of a Special Issue entitled Linking transcription to physiology in lipodomics.


Subject(s)
Fatty Acids/genetics , Fatty Acids/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , Animals
9.
Mol Cell ; 31(4): 531-543, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18722179

ABSTRACT

To study the in vivo role of TFIID in the transcriptional regulation of hepatic genes, we generated mice with liver-specific disruption of the TAF10 gene. Inactivation of TAF10 in hepatocytes resulted in the dissociation of TFIID into individual components. This correlated with the downregulation of most hepatocyte-specific genes during embryonic life and a defect in liver organogenesis. Unexpectedly, however, the transcription of less than 5% of active genes was affected by TAF10 inactivation and TFIID disassembly in adult liver. The extent of changes in transcription of the affected genes was dependent on the timing of their activation during liver development, relative to that of TAF10 inactivation. Furthermore, TFIID dissociation from promoters leads to the re-expression of several postnatally silenced hepatic genes. Promoter occupancy analyses, combined with expression profiling, demonstrate that TFIID is required for the initial activation or postnatal repression of genes, while it is dispensable for maintaining ongoing transcription.


Subject(s)
Gene Expression Regulation, Developmental , Liver/metabolism , Transcription Factor TFIID/metabolism , Animals , Gene Expression Profiling , Gene Targeting , Hepatocytes/cytology , Hepatocytes/metabolism , Liver/embryology , Mice , Mice, Knockout , Models, Genetic , Organ Specificity , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding , Protein Subunits/metabolism , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism , Transcription, Genetic
10.
Proc Natl Acad Sci U S A ; 109(8): 2995-3000, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22323595

ABSTRACT

Nucleotide excision repair (NER) defects are associated with cancer, developmental disorders and neurodegeneration. However, with the exception of cancer, the links between defects in NER and developmental abnormalities are not well understood. Here, we show that the ERCC1-XPF NER endonuclease assembles on active promoters in vivo and facilitates chromatin modifications for transcription during mammalian development. We find that Ercc1(-/-) mice demonstrate striking physiological, metabolic and gene expression parallels with Taf10(-/-) animals carrying a liver-specific transcription factor II D (TFIID) defect in transcription initiation. Promoter occupancy studies combined with expression profiling in the liver and in vitro differentiation cell assays reveal that ERCC1-XPF interacts with TFIID and assembles with POL II and the basal transcription machinery on promoters in vivo. Whereas ERCC1-XPF is required for the initial activation of genes associated with growth, it is dispensable for ongoing transcription. Recruitment of ERCC1-XPF on promoters is accompanied by promoter-proximal DNA demethylation and histone marks associated with active hepatic transcription. Collectively, the data unveil a role of ERCC1/XPF endonuclease in transcription initiation establishing its causal contribution to NER developmental disorders.


Subject(s)
DNA Repair/genetics , Growth and Development/genetics , Progeria/genetics , Transcription, Genetic , Adipogenesis/genetics , Animals , Animals, Newborn , DNA Methylation/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Disease Models, Animal , Endonucleases/deficiency , Gene Expression Regulation, Developmental , Genome/genetics , Histones/metabolism , Liver/growth & development , Liver/metabolism , Liver/pathology , Mice , Organ Specificity , Progeria/enzymology , Progeria/pathology , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Protein Processing, Post-Translational/genetics , Transcription Factor TFIID/metabolism , Transcriptome/genetics
11.
Nat Commun ; 15(1): 5843, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992049

ABSTRACT

Setd8 regulates transcription elongation, mitotic DNA condensation, DNA damage response and replication licensing. Here we show that, in mitogen-stimulated liver-specific Setd8-KO mice, most of the hepatocytes are eliminated by necrosis but a significant number of them survive via entering a stage exhibiting several senescence-related features. Setd8-deficient hepatocytes had enlarged nuclei, chromosomal hyperploidy and nuclear engulfments progressing to the formation of intranuclear vesicles surrounded by nuclear lamina. These vesicles contain glycogen, cytoplasmic proteins and even entire organelles. We term this process "endonucleosis". Intranuclear vesicles are absent in hepatocytes of Setd8/Atg5 knockout mice, suggesting that the process requires the function of the canonical autophagy machinery. Endonucleosis and hyperploidization are temporary, early events in the surviving Setd8-deficient cells. Larger vesicles break down into microvesicles over time and are eventually eliminated. The results reveal sequential events in cells with extensive DNA damage, which function as part of survival mechanisms to prevent necrotic death.


Subject(s)
Cell Nucleus , Cytoplasm , Hepatocytes , Mice, Knockout , Animals , Cytoplasm/metabolism , Cell Nucleus/metabolism , Mice , Hepatocytes/metabolism , Necrosis , DNA Damage , Autophagy/physiology , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 5/genetics , Mice, Inbred C57BL , Male
12.
Cells ; 10(8)2021 08 08.
Article in English | MEDLINE | ID: mdl-34440795

ABSTRACT

During liver organogenesis, cellular transcriptional profiles are constantly reshaped by the action of hepatic transcriptional regulators, including FoxA1-3, GATA4/6, HNF1α/ß, HNF4α, HNF6, OC-2, C/EBPα/ß, Hex, and Prox1. These factors are crucial for the activation of hepatic genes that, in the context of compact chromatin, cannot access their targets. The initial opening of highly condensed chromatin is executed by a special class of transcription factors known as pioneer factors. They bind and destabilize highly condensed chromatin and facilitate access to other "non-pioneer" factors. The association of target genes with pioneer and non-pioneer transcription factors takes place long before gene activation. In this way, the underlying gene regulatory regions are marked for future activation. The process is called "bookmarking", which confers transcriptional competence on target genes. Developmental bookmarking is accompanied by a dynamic maturation process, which prepares the genomic loci for stable and efficient transcription. Stable hepatic expression profiles are maintained during development and adulthood by the constant availability of the main regulators. This is achieved by a self-sustaining regulatory network that is established by complex cross-regulatory interactions between the major regulators. This network gradually grows during liver development and provides an epigenetic memory mechanism for safeguarding the optimal expression of the regulators.


Subject(s)
Gene Expression Regulation, Developmental , Liver/metabolism , Organogenesis/genetics , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Endoderm/cytology , Endoderm/embryology , Endoderm/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Liver/cytology , Liver/embryology , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism
13.
iScience ; 24(5): 102473, 2021 May 21.
Article in English | MEDLINE | ID: mdl-34113819

ABSTRACT

The oncogenic function of suppressor of variegation, enhancer of zeste and MYeloid-Nervy-DEAF1-domain family methyltransferase Smyd3 has been implicated in various malignancies, including hepatocellular carcinoma (HCC). Here, we show that targeting Smyd3 by next-generation antisense oligonucleotides (Smyd3-ASO) is an efficient approach to modulate its mRNA levels in vivo and to halt the growth of already initiated liver tumors. Smyd3-ASO treatment dramatically decreased tumor burden in a mouse model of chemically induced HCC and negatively affected the growth rates, migration, oncosphere formation, and xenograft growth capacity of a panel of human hepatic cancer cell lines. Smyd3-ASOs prevented the activation of oncofetal genes and the development of cancer-specific gene expression program. The results point to a mechanism by which Smyd3-ASO treatment blocks cellular de-differentiation, a hallmark feature of HCC development, and, as a result, it inhibits the expansion of hepatic cancer stem cells, a population that has been presumed to resist chemotherapy.

14.
Cell Rep ; 30(5): 1319-1328.e6, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32023452

ABSTRACT

Transcription factor binding to enhancer and promoter regions critical for homeostatic adult gene activation is established during development. To understand how cell-specific gene expression patterns are generated, we study the developmental timing of association of two prominent hepatic transcription factors with gene regulatory regions. Most individual binding events display extraordinarily high temporal variations during liver development. Early and persistent binding is necessary, but not sufficient, for gene activation. Stable gene expression patterns are the result of combinatorial activity of multiple transcription factors, which mark regulatory regions long before activation and promote progressive broadening of active chromatin domains. Both temporally stable and dynamic, short-lived binding events contribute to the developmental maturation of active promoter configurations. The results reveal a developmental bookmarking function of master regulators and illuminate remarkable parallels between the principles employed for gene activation during development, during evolution, and upon mitotic exit.


Subject(s)
Liver/embryology , Liver/metabolism , Transcription Factors/metabolism , Transcriptional Activation/genetics , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Chromatin/metabolism , Gene Expression Regulation, Developmental , Gene Silencing , Hepatocyte Nuclear Factor 4/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Regulatory Sequences, Nucleic Acid
15.
Mol Cell Biol ; 26(19): 7017-29, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16980607

ABSTRACT

Hepatocyte nuclear factor 4 (HNF-4) is a key member of the transcription factor network regulating hepatocyte differentiation and function. Activation of the HNF-4 gene involves physical interaction between a distant enhancer and the proximal promoter region, bound by distinct sets of transcription factors. Here we report that, upon mitogen-activated protein (MAP) kinase activation, HNF-4 expression is downregulated in human hepatoma cells. This effect is mediated by the loss of CEBPalpha expression. During MAP kinase signaling, the recruitment of HNF-3beta and HNF-1alpha to the HNF-4 enhancer and RNA polymerase II to the proximal HNF-4 promoter was compromised. CBP, Brg1, and TFIIB were also dissociated from the HNF-4 regulatory regions, and the enhancer-promoter complex was disrupted. Interestingly, the extent of nucleosome acetylation did not decrease at either regulatory region, and HNF-6 and HNF-1alpha, as well as components of the TFIID, remained associated with the proximal promoter during the repressed state. The results point to an absolute requirement of enhancer-promoter communication for maintaining the active state of the HNF-4 gene and provide evidence for a molecular bookmarking mechanism, which may contribute to the prevention of permanent silencing of the locus during the repressed state.


Subject(s)
Enhancer Elements, Genetic/genetics , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Promoter Regions, Genetic/genetics , Acetylation/drug effects , CCAAT-Enhancer-Binding Protein-alpha/deficiency , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cells, Cultured , DNA Helicases , Down-Regulation/drug effects , Enzyme Activation/drug effects , Hepatocyte Nuclear Factor 4/deficiency , Histones/metabolism , Humans , Models, Genetic , Nuclear Proteins/metabolism , RNA Interference , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/metabolism
16.
Mol Cell Biol ; 25(12): 5095-105, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15923626

ABSTRACT

Posttranslational modification by SUMO elicits a repressive effect on many transcription factors. In principle, sumoylation may either influence transcription factor activity on promoters, or it may act indirectly by targeting the modified factors to specific cellular compartments. To provide direct experimental evidence for the above, not necessarily mutually exclusive models, we analyzed the role of SUMO modification on the localization and the activity of the orphan nuclear receptor LRH-1. We demonstrate, by using fluorescence resonance energy transfer (FRET) and fluorescence recovery after photobleaching (FRAP) assays, that sumoylated LRH-1 is exclusively localized in promyelocytic leukemia protein (PML) nuclear bodies and that this association is a dynamic process. Release of LRH-1 from nuclear bodies correlated with its desumoylation, pointing to the pivotal role of SUMO conjugation in keeping LRH-1 in these locations. SUMO-dependent shuttling of LRH-1 into PML bodies defines two spatially separated pools of the protein, of which only the soluble, unmodified one is associated with actively transcribed target genes. The results suggest that SUMO-PML nuclear bodies may primarily function as dynamic molecular reservoirs, controlling the availability of certain transcription factors to active chromatin domains.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , Intranuclear Inclusion Bodies/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , SUMO-1 Protein/metabolism , Cell Line , DNA-Binding Proteins/genetics , Fluorescence Recovery After Photobleaching , Fluorescence Resonance Energy Transfer , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SUMO-1 Protein/genetics , Transcription Factors , Transcription, Genetic
17.
Cell Rep ; 20(4): 909-922, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28746875

ABSTRACT

H4K20 monomethylation maintains genome integrity by regulating proper mitotic condensation, DNA damage response, and replication licensing. Here, we show that, in non-dividing hepatic cells, H4K20Me1 is specifically enriched in active gene bodies and dynamically regulated by the antagonistic action of Kmt5a methylase and Kdm7b demethylase. In liver-specific Kmt5a-deficient mice, reduced levels of H4K20Me1 correlated with reduced RNA Pol II release from promoter-proximal regions. Genes regulating glucose and fatty acid metabolism were most sensitive to impairment of RNA Pol II release. Downregulation of glycolytic genes resulted in an energy starvation condition partially compensated by AMP-activated protein kinase (AMPK) activation and increased mitochondrial activity. This metabolic reprogramming generated a highly sensitized state that, upon different metabolic stress conditions, quickly aggravated into a senescent phenotype due to ROS overproduction-mediated oxidative DNA damage. The results illustrate how defects in the general process of RNA Pol II transition into a productive elongation phase can trigger specific metabolic changes and genome instability.


Subject(s)
Histones/metabolism , Liver/metabolism , Protein Methyltransferases/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic/genetics , Animals , Gene Expression Regulation , Histones/genetics , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Methyltransferases/genetics , RNA Polymerase II/genetics
18.
ACS Chem Biol ; 12(3): 654-663, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28059499

ABSTRACT

Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII/NR2F2) is an orphan member of the nuclear receptor family of transcription factors whose activities are modulated upon binding of small molecules into an hydrophobic ligand-binding pocket (LBP). Although the LBP of COUP-TFII is filled with aromatic amino-acid side chains, alternative modes of ligand binding could potentially lead to regulation of the orphan receptor. Here, we screened a synthetic and natural compound library in a yeast one-hybrid assay and identified 4-methoxynaphthol as an inhibitor of COUP-TFII. This synthetic inhibitor was able to counteract processes either positively or negatively regulated by COUP-TFII in different mammalian cell systems. Hence, we demonstrate that the true orphan receptor COUP-TFII can be targeted by small chemicals which could be used to study the physiological functions of COUP-TFII or to counteract detrimental COUP-TFII activities in various pathological conditions.


Subject(s)
COUP Transcription Factor II/antagonists & inhibitors , Small Molecule Libraries , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Animals , Binding Sites , COUP Transcription Factor II/metabolism , Cell Differentiation/drug effects , Hep G2 Cells , Humans , Mice
19.
Nucleic Acids Res ; 32(20): 6096-103, 2004.
Article in English | MEDLINE | ID: mdl-15550569

ABSTRACT

Site-specific modification of nucleosomal histones plays a central role in the formation of transcriptionally active and inactive chromatin structures. These modifications may serve as specific recognition motifs for chromatin proteins, which act as a signal for the adoption of the appropriate regulatory responses. Here, we show that the orphan nuclear receptor SHP (small heterodimer partner), a coregulator that inhibits the activity of several nuclear receptors, can associate with unmodified and lysine 9-methylated histone-3, but not with the acetylated protein. The naturally occurring SHP mutant (R213C), which exhibits decreased transrepression potential, interacts less avidly with K9-methylated histone 3. We demonstrate that SHP can functionally interact with histone deacetylase-1 and the G9a methyltransferase and that it is localized exclusively in nuclease-sensitive euchromatin. The results point to the involvement of a multistep mechanism in SHP-dependent transcriptional repression, which includes histone deacetylation, followed by H3-K9 methylation and stable association of SHP itself with chromatin.


Subject(s)
Gene Silencing , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Acetylation , Animals , Cell Line , Euchromatin/chemistry , Histone Deacetylases/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/physiology , Histones/chemistry , Humans , Methylation , Protein Methyltransferases , Receptors, Cytoplasmic and Nuclear/analysis , Repressor Proteins/analysis , Transcription, Genetic
20.
Mol Cell Oncol ; 3(1): e1021946, 2016.
Article in English | MEDLINE | ID: mdl-27308536

ABSTRACT

Cancer stem cells (CSCs) are defined as cells within tumors that can self-renew and differentiate into heterogeneous lineages of cancerous cells. The origin of CSCs is not well understood. Recent evidence suggests that CSCs in hepatocellular carcinoma could be generated via oncogenic transformation and partial differentiation of adult hepatic ductal progenitor cells.

SELECTION OF CITATIONS
SEARCH DETAIL