Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell ; 84(7): 1180-1182, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38579674

ABSTRACT

Using cryo-EM and biochemical methods, Su and Vos1 discover an alternative NELF structural state that enables transcription and switches NELF-RNA polymerase II (RNAPII) compatibility with other RNAPII-associated factors that regulate pausing, elongation, termination, and transcription-coupled DNA repair.


Subject(s)
RNA Polymerase II , Transcription Factors , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
2.
Nat Rev Mol Cell Biol ; 23(11): 732-749, 2022 11.
Article in English | MEDLINE | ID: mdl-35725906

ABSTRACT

The Mediator complex, which in humans is 1.4 MDa in size and includes 26 subunits, controls many aspects of RNA polymerase II (Pol II) function. Apart from its size, a defining feature of Mediator is its intrinsic disorder and conformational flexibility, which contributes to its ability to undergo phase separation and to interact with a myriad of regulatory factors. In this Review, we discuss Mediator structure and function, with emphasis on recent cryogenic electron microscopy data of the 4.0-MDa transcription preinitiation complex. We further discuss how Mediator and sequence-specific DNA-binding transcription factors enable enhancer-dependent regulation of Pol II function at distal gene promoters, through the formation of molecular condensates (or transcription hubs) and chromatin loops. Mediator regulation of Pol II reinitiation is also discussed, in the context of transcription bursting. We propose a working model for Mediator function that combines experimental results and theoretical considerations related to enhancer-promoter interactions, which reconciles contradictory data regarding whether enhancer-promoter communication is direct or indirect. We conclude with a discussion of Mediator's potential as a therapeutic target and of future research directions.


Subject(s)
Mediator Complex , RNA Polymerase II , Humans , RNA Polymerase II/metabolism , Mediator Complex/metabolism , Transcription Factors/metabolism , Chromatin/genetics , DNA , Transcription, Genetic
3.
Elife ; 102021 07 15.
Article in English | MEDLINE | ID: mdl-34263728

ABSTRACT

MLL-rearranged leukemia depends on H3K79 methylation. Depletion of this transcriptionally activating mark by DOT1L deletion or high concentrations of the inhibitor pinometostat downregulates HOXA9 and MEIS1, and consequently reduces leukemia survival. Yet, some MLL-rearranged leukemias are inexplicably susceptible to low-dose pinometostat, far below concentrations that downregulate this canonical proliferation pathway. In this context, we define alternative proliferation pathways that more directly derive from H3K79me2 loss. By ICeChIP-seq, H3K79me2 is markedly depleted at pinometostat-downregulated and MLL-fusion targets, with paradoxical increases of H3K4me3 and loss of H3K27me3. Although downregulation of polycomb components accounts for some of the proliferation defect, transcriptional downregulation of FLT3 is the major pathway. Loss-of-FLT3-function recapitulates the cytotoxicity and gene expression consequences of low-dose pinometostat, whereas overexpression of constitutively active STAT5A, a target of FLT3-ITD-signaling, largely rescues these defects. This pathway also depends on MLL1, indicating combinations of DOT1L, MLL1 and FLT3 inhibitors should be explored for treating FLT3-mutant leukemia.


Subject(s)
Gene Rearrangement , Histones/metabolism , Leukemia/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Leukemic , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia/genetics , Methylation , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , STAT5 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , fms-Like Tyrosine Kinase 3/genetics
4.
Nat Protoc ; 14(12): 3275-3302, 2019 12.
Article in English | MEDLINE | ID: mdl-31723301

ABSTRACT

Chromatin immunoprecipitation coupled to next-generation sequencing (ChIP-seq) has served as the central method for the study of histone modifications for the past decade. In ChIP-seq analyses, antibodies selectively capture nucleosomes bearing a modification of interest and the associated DNA is then mapped to the genome to determine the distribution of the mark. This approach has several important drawbacks: (i) ChIP interpretation necessitates the assumption of perfect antibody specificity, despite growing evidence that this is often not the case. (ii) Common methods for evaluating antibody specificity in other formats have little or no bearing on specificity within a ChIP experiment. (iii) Uncalibrated ChIP is reported as relative enrichment, which is biologically meaningless outside the experimental reference frame defined by a discrete immunoprecipitation (IP), thus preventing facile comparison across experimental conditions or modifications. (iv) Differential library amplification and loading onto next-generation sequencers, as well as computational normalization, can further compromise quantitative relationships that may exist between samples. Consequently, the researcher is presented with a series of potential pitfalls and is blind to nearly all of them. Here we provide a detailed protocol for internally calibrated ChIP (ICeChIP), a method we recently developed to resolve these problems by spike-in of defined nucleosomal standards within a ChIP procedure. This protocol is optimized for specificity and quantitative power, allowing for measurement of antibody specificity and absolute measurement of histone modification density (HMD) at genomic loci on a biologically meaningful scale enabling unambiguous comparisons. We provide guidance on optimal conditions for next-generation sequencing (NGS) and instructions for data analysis. This protocol takes between 17 and 18 h, excluding time for sequencing or bioinformatic analysis. The ICeChIP procedure enables accurate measurement of histone post-translational modifications (PTMs) genome-wide in mammalian cells as well as Drosophila melanogaster and Caenorhabditis elegans, indicating suitability for use in eukaryotic cells more broadly.


Subject(s)
Chromatin Immunoprecipitation Sequencing/methods , Sequence Analysis, DNA/methods , Animals , Antibody Specificity/immunology , Caenorhabditis elegans/genetics , Calibration , Chromatin Immunoprecipitation/methods , Computational Biology , DNA , Drosophila melanogaster/genetics , Gene Library , Genome , High-Throughput Nucleotide Sequencing/methods , Histones/genetics , Histones/immunology , Humans , Nucleosomes/genetics , Nucleosomes/immunology , Protein Processing, Post-Translational , Reproducibility of Results
5.
Proc Natl Acad Sci U S A ; 109(45): 18499-504, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23093672

ABSTRACT

Epigenetic regulation underlies the robust changes in gene expression that occur during development. How precisely epigenetic enzymes contribute to development and differentiation processes is largely unclear. Here we show that one of the enzymes that removes the activating epigenetic mark of trimethylated lysine 4 on histone H3, lysine (K)-specific demethylase 5A (KDM5A), reinforces the effects of the retinoblastoma (RB) family of transcriptional repressors on differentiation. Global location analysis showed that KDM5A cooccupies a substantial portion of target genes with the E2F4 transcription factor. During ES cell differentiation, knockout of KDM5A resulted in derepression of multiple genomic loci that are targets of KDM5A, denoting a direct regulatory function. In terminally differentiated cells, common KDM5A and E2F4 gene targets were bound by the pRB-related protein p130, a DREAM complex component. KDM5A was recruited to the transcription start site regions independently of E2F4; however, it cooperated with E2F4 to promote a state of deepened repression at cell cycle genes during differentiation. These findings reveal a critical role of H3K4 demethylation by KDM5A in the transcriptional silencing of genes that are suppressed by RB family members in differentiated cells.


Subject(s)
Cell Differentiation/genetics , E2F4 Transcription Factor/metabolism , Genes, cdc/genetics , Repressor Proteins/metabolism , Retinoblastoma-Binding Protein 2/metabolism , Animals , Chromatin Immunoprecipitation , Embryonic Stem Cells/metabolism , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Mice , Mice, Inbred C57BL , Multiprotein Complexes/metabolism , Protein Binding , U937 Cells
6.
PLoS One ; 6(8): e24023, 2011.
Article in English | MEDLINE | ID: mdl-21886846

ABSTRACT

Cancer is characterized by aberrant patterns of expression of multiple genes. These major shifts in gene expression are believed to be due to not only genetic but also epigenetic changes. The epigenetic changes are communicated through chemical modifications, including histone modifications. However, it is unclear whether the binding of histone-modifying proteins to genomic regions and the placing of histone modifications efficiently discriminates corresponding genes from the rest of the genes in the human genome. We performed gene expression analysis of histone demethylases (HDMs) and histone methyltransferases (HMTs), their target genes and genes with relevant histone modifications in normal and tumor tissues. Surprisingly, this analysis revealed the existence of correlations in the expression levels of different HDMs and HMTs. The observed HDM/HMT gene expression signature was specific to particular normal and cancer cell types and highly correlated with target gene expression and the expression of genes with histone modifications. Notably, we observed that trimethylation at lysine 4 and lysine 27 separated preferentially expressed and underexpressed genes, which was strikingly different in cancer cells compared to normal cells. We conclude that changes in coordinated regulation of enzymes executing histone modifications may underlie global epigenetic changes occurring in cancer.


Subject(s)
Epigenesis, Genetic , Histone Demethylases/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histone Methyltransferases , Humans , Neoplasms/enzymology
7.
Cell Cycle ; 10(3): 413-24, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21270517

ABSTRACT

Histones are post-translationally modified by multiple histone-modifying enzymes, which in turn influences gene expression. Much of the work in the field to date has focused on genetic, biochemical and structural characterization of these enzymes. The most recent genome-wide methods provide insights into specific recruitment of histone-modifying enzymes in vivo and, therefore, onto mechanisms of establishing a differential expression pattern. Here we focus on the recruitment mechanisms of the enzymes involved in the placement of two contrasting histone marks, histone H3 lysine 4 (H3K4) methylation and histone H3 lysine 27 (H3K27) methylation. We describe distribution of their binding sites and show that recruitment of different histone-modifying proteins can be coordinated, opposed, or alternating. Specifically, genomic sites of the H3K4 histone demethylase KDM5A become accessible to its homolog KDM5B in cells with a lowered KDM5A level. The currently available data on recruitment of H3K4/H3K27 modifying enzymes suggests that the formed protein complexes are targeted in a sequential and temporal manner, but that additional, still unknown, interactions contribute to targeting specificity.


Subject(s)
Histones/metabolism , Protein Processing, Post-Translational , Animals , Binding Sites , Cell Line , Chromosomal Proteins, Non-Histone/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Gene Expression Regulation , Histone Demethylases , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/physiology , Histones/chemistry , Humans , Jumonji Domain-Containing Histone Demethylases/physiology , Lysine/chemistry , Lysine/metabolism , Methylation , Mice , Models, Genetic , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Repressor Proteins/physiology , Retinoblastoma-Binding Protein 2/physiology , Transcription, Genetic
8.
J Vis Exp ; (41)2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20644511

ABSTRACT

Recruitment of transcriptional and epigenetic factors to their targets is a key step in their regulation. Prominently featured in recruitment are the protein domains that bind to specific histone modifications. One such domain is the plant homeodomain (PHD), found in several chromatin-binding proteins. The epigenetic factor RBP2 has multiple PHD domains, however, they have different functions (Figure 4). In particular, the C-terminal PHD domain, found in a RBP2 oncogenic fusion in human leukemia, binds to trimethylated lysine 4 in histone H3 (H3K4me3). The transcript corresponding to the RBP2 isoform containing the C-terminal PHD accumulates during differentiation of promonocytic, lymphoma-derived, U937 cells into monocytes. Consistent with both sets of data, genome-wide analysis showed that in differentiated U937 cells, the RBP2 protein gets localized to genomic regions highly enriched for H3K4me3. Localization of RBP2 to its targets correlates with a decrease in H3K4me3 due to RBP2 histone demethylase activity and a decrease in transcriptional activity. In contrast, two other PHDs of RBP2 are unable to bind H3K4me3. Notably, the C-terminal domain PHD of RBP2 is absent in the smaller RBP2 isoform. It is conceivable that the small isoform of RBP2, which lacks interaction with H3K4me3, differs from the larger isoform in genomic location. The difference in genomic location of RBP2 isoforms may account for the observed diversity in RBP2 function. Specifically, RBP2 is a critical player in cellular differentiation mediated by the retinoblastoma protein (pRB). Consistent with these data, previous genome-wide analysis, without distinction between isoforms, identified two distinct groups of RBP2 target genes: 1) genes bound by RBP2 in a manner that is independent of differentiation; 2) genes bound by RBP2 in a differentiation-dependent manner. To identify differences in localization between the isoforms we performed genome-wide location analysis by ChIP-Seq. Using antibodies that detect both RBP2 isoforms we have located all RBP2 targets. Additionally we have antibodies that only bind large, and not small RBP2 isoform (Figure 4). After identifying the large isoform targets, one can then subtract them from all RBP2 targets to reveal the targets of small isoform. These data show the contribution of chromatin-interacting domain in protein recruitment to its binding sites in the genome.


Subject(s)
Chromatin Immunoprecipitation/methods , Genome, Human , Retinol-Binding Proteins, Cellular/genetics , Humans , Protein Structure, Tertiary , U937 Cells
9.
J Steroid Biochem Mol Biol ; 91(4-5): 197-210, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15336697

ABSTRACT

Recent reports describe the ability of factors to modulate the position of the dose-response curve of receptor-agonist complexes, and the amount of partial agonist activity of receptor-antagonist complexes, of androgen, glucocorticoid (GRs), and progesterone receptors (PRs). We now ask whether this modulation extends to the two remaining steroid receptors: mineralocorticoid (MRs) and estrogen receptors (ERs). These studies of MR were facilitated by our discovery that the antiglucocorticoid dexamethasone 21-mesylate (Dex-Mes) is a new antimineralocorticoid with significant amounts of partial agonist activity. Elevated levels of MR, the co-activators TIF2 and SRC-1, and the co-repressor SMRT do modulate the dose-response curve and partial agonist activity of MR complexes. Interestingly, the precise responses are indistinguishable from those seen with GRs in the same cells. Thus, the unequal transactivation of common genes by MRs versus GRs probably cannot be explained by differential responses to changing cellular concentrations of homologous receptor, co-activators, or co-repressors. We also find that the dose-response curve of ER-estradiol complexes is left-shifted to lower steroid concentrations by higher amounts of exogenous ER. Therefore, the modulation of either the dose-response curve of agonists or the partial agonist activity of antisteroid, and in many cases the modulation of both properties, is a common phenomenon for all of the classical steroid receptors.


Subject(s)
Affinity Labels/pharmacology , Dexamethasone/analogs & derivatives , Hormone Antagonists/pharmacology , Mineralocorticoids/physiology , Receptors, Estrogen/physiology , Transcription Factors/physiology , Transcription, Genetic/drug effects , Animals , CHO Cells , Cricetinae , DNA-Binding Proteins/physiology , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Estradiol/metabolism , Haplorhini , Histone Acetyltransferases , Hormone Antagonists/chemistry , Humans , Kidney , Luciferases/metabolism , Nuclear Proteins/physiology , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivator 2 , Repressor Proteins/physiology , Trans-Activators/physiology , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...