Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Genome Res ; 31(9): 1663-1679, 2021 09.
Article in English | MEDLINE | ID: mdl-34426512

ABSTRACT

Antibodies offer a powerful means to interrogate specific proteins in a complex milieu. However, antibody availability and reliability can be problematic, whereas epitope tagging can be impractical in many cases. To address these limitations, the Protein Capture Reagents Program (PCRP) generated over a thousand renewable monoclonal antibodies (mAbs) against human presumptive chromatin proteins. However, these reagents have not been widely field-tested. We therefore performed a screen to test their ability to enrich genomic regions via chromatin immunoprecipitation (ChIP) and a variety of orthogonal assays. Eight hundred eighty-seven unique antibodies against 681 unique human transcription factors (TFs) were assayed by ultra-high-resolution ChIP-exo/seq, generating approximately 1200 ChIP-exo data sets, primarily in a single pass in one cell type (K562). Subsets of PCRP mAbs were further tested in ChIP-seq, CUT&RUN, STORM super-resolution microscopy, immunoblots, and protein binding microarray (PBM) experiments. About 5% of the tested antibodies displayed high-confidence target (i.e., cognate antigen) enrichment across at least one assay and are strong candidates for additional validation. An additional 34% produced ChIP-exo data that were distinct from background and thus warrant further testing. The remaining 61% were not substantially different from background, and likely require consideration of a much broader survey of cell types and/or assay optimizations. We show and discuss the metrics and challenges to antibody validation in chromatin-based assays.


Subject(s)
Chromatin Immunoprecipitation Sequencing , Transcription Factors , Binding Sites , Chromatin Immunoprecipitation , Humans , Indicators and Reagents , Reproducibility of Results , Transcription Factors/metabolism
2.
Genes Dev ; 35(3-4): 273-285, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33446572

ABSTRACT

The regulation of gene expression catalyzed by RNA polymerase II (Pol II) requires a host of accessory factors to ensure cell growth, differentiation, and survival under environmental stress. Here, using the auxin-inducible degradation (AID) system to study transcriptional activities of the bromodomain and extraterminal domain (BET) and super elongation complex (SEC) families, we found that the CDK9-containing BRD4 complex is required for the release of Pol II from promoter-proximal pausing for most genes, while the CDK9-containing SEC is required for activated transcription in the heat shock response. By using both the proteolysis targeting chimera (PROTAC) dBET6 and the AID system, we found that dBET6 treatment results in two major effects: increased pausing due to BRD4 loss, and reduced enhancer activity attributable to BRD2 loss. In the heat shock response, while auxin-mediated depletion of the AFF4 subunit of the SEC has a more severe defect than AFF1 depletion, simultaneous depletion of AFF1 and AFF4 leads to a stronger attenuation of the heat shock response, similar to treatment with the SEC inhibitor KL-1, suggesting a possible redundancy among SEC family members. This study highlights the usefulness of orthogonal acute depletion/inhibition strategies to identify distinct and redundant biological functions among Pol II elongation factor paralogs.


Subject(s)
Gene Expression/genetics , Peptide Elongation Factors/metabolism , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , HCT116 Cells , Heat-Shock Response , Humans , Peptide Elongation Factors/genetics , Proteins/genetics , Proteins/metabolism , RNA Polymerase II/genetics , Transcription Factors/genetics
3.
Genes Dev ; 34(21-22): 1493-1502, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33033055

ABSTRACT

Catalytic-inactivating mutations within the Drosophila enhancer H3K4 mono-methyltransferase Trr and its mammalian homologs, MLL3/4, cause only minor changes in gene expression compared with whole-gene deletions for these COMPASS members. To identify essential histone methyltransferase-independent functions of Trr, we screened to identify a minimal Trr domain sufficient to rescue Trr-null lethality and demonstrate that this domain binds and stabilizes Utx in vivo. Using the homologous MLL3/MLL4 human sequences, we mapped a short ∼80-amino-acid UTX stabilization domain (USD) that promotes UTX stability in the absence of the rest of MLL3/4. Nuclear UTX stability is enhanced when the USD is fused with the MLL4 HMG-box. Thus, COMPASS-dependent UTX stabilization is an essential noncatalytic function of Trr/MLL3/MLL4, suggesting that stabilizing UTX could be a therapeutic strategy for cancers with MLL3/4 loss-of-function mutations.


Subject(s)
Conserved Sequence/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Genes, Lethal/genetics , Histone-Lysine N-Methyltransferase/genetics , Oxidoreductases, N-Demethylating/genetics , Animals , Gene Deletion , Gene Expression Regulation/genetics , HCT116 Cells , Humans , Protein Domains , Protein Stability
4.
Proc Natl Acad Sci U S A ; 117(44): 27365-27373, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33077595

ABSTRACT

Actively transcribed genes in mammals are decorated by H3K79 methylation, which is correlated with transcription levels and is catalyzed by the histone methyltransferase DOT1L. DOT1L is required for mammalian development, and the inhibition of its catalytic activity has been extensively studied for cancer therapy; however, the mechanisms underlying DOT1L's functions in normal development and cancer pathogenesis remain elusive. To dissect the relationship between H3K79 methylation, cellular differentiation, and transcription regulation, we systematically examined the role of DOT1L and its catalytic activity in embryonic stem cells (ESCs). DOT1L is dispensable for ESC self-renewal but is required for establishing the proper expression signature of neural progenitor cells, while catalytic inactivation of DOT1L has a lesser effect. Furthermore, DOT1L loss, rather than its catalytic inactivation, causes defects in glial cell specification. Although DOT1L loss by itself has no major defect in transcription elongation, transcription elongation defects seen with the super elongation complex inhibitor KL-2 are exacerbated in DOT1L knockout cells, but not in catalytically dead DOT1L cells, revealing a role of DOT1L in promoting productive transcription elongation that is independent of H3K79 methylation. Taken together, our study reveals a catalytic-independent role of DOT1L in modulating cell-fate determination and in transcriptional elongation control.


Subject(s)
Cell Differentiation/genetics , Histone-Lysine N-Methyltransferase/metabolism , Transcription Elongation, Genetic/physiology , Cell Proliferation/drug effects , DNA Methylation/physiology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/genetics , Epigenomics , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Methyltransferases/metabolism , Neural Stem Cells/metabolism , Protein Processing, Post-Translational , Transcriptional Elongation Factors/metabolism
5.
Nat Genet ; 52(6): 615-625, 2020 06.
Article in English | MEDLINE | ID: mdl-32393859

ABSTRACT

The COMPASS protein family catalyzes histone H3 Lys 4 (H3K4) methylation and its members are essential for regulating gene expression. MLL2/COMPASS methylates H3K4 on many developmental genes and bivalent clusters. To understand MLL2-dependent transcriptional regulation, we performed a CRISPR-based screen with an MLL2-dependent gene as a reporter in mouse embryonic stem cells. We found that MLL2 functions in gene expression by protecting developmental genes from repression via repelling PRC2 and DNA methylation machineries. Accordingly, repression in the absence of MLL2 is relieved by inhibition of PRC2 and DNA methyltransferases. Furthermore, DNA demethylation on such loci leads to reactivation of MLL2-dependent genes not only by removing DNA methylation but also by opening up previously CpG methylated regions for PRC2 recruitment, diluting PRC2 at Polycomb-repressed genes. These findings reveal how the context and function of these three epigenetic modifiers of chromatin can orchestrate transcriptional decisions and demonstrate that prevention of active repression by the context of the enzyme and not H3K4 trimethylation underlies transcriptional regulation on MLL2/COMPASS targets.


Subject(s)
DNA Methylation , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Mouse Embryonic Stem Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , Chromosomal Proteins, Non-Histone/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Lysine/metabolism , Methylation , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/physiology , Myeloid-Lymphoid Leukemia Protein/genetics , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Promoter Regions, Genetic , Trans-Activators/genetics
6.
Mol Cell ; 78(2): 261-274.e5, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32155413

ABSTRACT

RNA polymerase II (RNA Pol II) is generally paused at promoter-proximal regions in most metazoans, and based on in vitro studies, this function has been attributed to the negative elongation factor (NELF). Here, we show that upon rapid depletion of NELF, RNA Pol II fails to be released into gene bodies, stopping instead around the +1 nucleosomal dyad-associated region. The transition to the 2nd pause region is independent of positive transcription elongation factor P-TEFb. During the heat shock response, RNA Pol II is rapidly released from pausing at heat shock-induced genes, while most genes are paused and transcriptionally downregulated. Both of these aspects of the heat shock response remain intact upon NELF loss. We find that NELF depletion results in global loss of cap-binding complex from chromatin without global reduction of nascent transcript 5' cap stability. Thus, our studies implicate NELF functioning in early elongation complexes distinct from RNA Pol II pause-release.


Subject(s)
Positive Transcriptional Elongation Factor B/genetics , RNA Polymerase II/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Heat-Shock Response/genetics , Humans , Mice , Nucleosomes/genetics , Promoter Regions, Genetic
7.
Sci Adv ; 5(7): eaax2887, 2019 07.
Article in English | MEDLINE | ID: mdl-31281901

ABSTRACT

Using biochemical characterization of fusion proteins associated with endometrial stromal sarcoma, we identified JAZF1 as a new subunit of the NuA4 acetyltransferase complex and CXORF67 as a subunit of the Polycomb Repressive Complex 2 (PRC2). Since CXORF67's interaction with PRC2 leads to decreased PRC2-dependent H3K27me2/3 deposition, we propose a new name for this gene: CATACOMB (catalytic antagonist of Polycomb; official gene name: EZHIP ). We map CATACOMB's inhibitory function to a short highly conserved region and identify a single methionine residue essential for diminution of H3K27me2/3 levels. Remarkably, the amino acid sequence surrounding this critical methionine resembles the oncogenic histone H3 Lys27-to-methionine (H3K27M) mutation found in high-grade pediatric gliomas. As CATACOMB expression is regulated through DNA methylation/demethylation, we propose CATACOMB as the potential interlocutor between DNA methylation and PRC2 activity. We raise the possibility that similar regulatory mechanisms could exist for other methyltransferase complexes such as Trithorax/COMPASS.


Subject(s)
Glioma/metabolism , Histones/metabolism , Neoplasm Proteins/metabolism , Oncogene Proteins/biosynthesis , Polycomb Repressive Complex 2/metabolism , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , DNA Methylation , DNA, Neoplasm , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , HCT116 Cells , Histones/genetics , Humans , Methylation , Neoplasm Proteins/genetics , Oncogene Proteins/genetics , Polycomb Repressive Complex 2/genetics
8.
Nat Commun ; 10(1): 555, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30696825

ABSTRACT

The original version of this Article contained an error in the author affiliations. The affiliation of Alice Chen-Plotkin with the Department of Neurology, Perelman School of Medicine, Philadelphia, PA, 19104 USA was inadvertently omitted. This has now been corrected in both the PDF and HTML versions of the Article.

9.
Sci Adv ; 4(11): eaau6986, 2018 11.
Article in English | MEDLINE | ID: mdl-30417100

ABSTRACT

The tet methylcytosine dioxygenase 2 (TET2) enzyme catalyzes the conversion of the modified DNA base 5-methylcytosine to 5-hydroxymethylcytosine. TET2 is frequently mutated or dysregulated in multiple human cancers, and loss of TET2 is associated with changes in DNA methylation patterns. Here, using newly developed TET2-specific antibodies and the estrogen response as a model system for studying the regulation of gene expression, we demonstrate that endogenous TET2 occupies active enhancers and facilitates the proper recruitment of estrogen receptor α (ERα). Knockout of TET2 by CRISPR-CAS9 leads to a global increase of DNA methylation at enhancers, resulting in attenuation of the estrogen response. We further identified a positive feedback loop between TET2 and ERα, which further requires MLL3 COMPASS at these enhancers. Together, this study reveals an epigenetic axis coordinating a transcriptional program through enhancer activation via DNA demethylation.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Demethylation , Enhancer Elements, Genetic , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CRISPR-Cas Systems , Cell Differentiation , Cohort Studies , DNA Methylation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Dioxygenases , Epigenesis, Genetic , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Female , Humans , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Survival Rate , Tumor Cells, Cultured
10.
Cell ; 175(3): 766-779.e17, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30340042

ABSTRACT

The super elongation complex (SEC) is required for robust and productive transcription through release of RNA polymerase II (Pol II) with its P-TEFb module and promoting transcriptional processivity with its ELL2 subunit. Malfunction of SEC contributes to multiple human diseases including cancer. Here, we identify peptidomimetic lead compounds, KL-1 and its structural homolog KL-2, which disrupt the interaction between the SEC scaffolding protein AFF4 and P-TEFb, resulting in impaired release of Pol II from promoter-proximal pause sites and a reduced average rate of processive transcription elongation. SEC is required for induction of heat-shock genes and treating cells with KL-1 and KL-2 attenuates the heat-shock response from Drosophila to human. SEC inhibition downregulates MYC and MYC-dependent transcriptional programs in mammalian cells and delays tumor progression in a mouse xenograft model of MYC-driven cancer, indicating that small-molecule disruptors of SEC could be used for targeted therapy of MYC-induced cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms, Experimental/drug therapy , Positive Transcriptional Elongation Factor B/metabolism , Repressor Proteins/metabolism , Transcription Elongation, Genetic/drug effects , Transcriptional Elongation Factors/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Drosophila , Female , HCT116 Cells , HEK293 Cells , Heat-Shock Response , Humans , Male , Mice , Mice, Inbred BALB C , Protein Binding/drug effects , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Polymerase II/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
11.
Nat Commun ; 9(1): 4406, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30353006

ABSTRACT

TDP-43 is the major disease protein associated with amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with ubiquitinated inclusions (FTLD-TDP). Here we identify the transcriptional elongation factor Ell-a shared component of little elongation complex (LEC) and super elongation complex (SEC)-as a strong modifier of TDP-43-mediated neurodegeneration. Our data indicate select targets of LEC and SEC become upregulated in the fly ALS/FTLD-TDP model. Among them, U12 snRNA and a stress-induced long non-coding RNA Hsrω, functionally contribute to TDP-43-mediated degeneration. We extend the findings of Hsrω, which we identify as a chromosomal target of TDP-43, to show that the human orthologue Sat III is elevated in a human cellular disease model and FTLD-TDP patient tissue. We further demonstrate an interaction between TDP-43 and human ELL2 by co-immunoprecipitation from human cells. These findings reveal important roles of Ell-complexes LEC and SEC in TDP-43-associated toxicity, providing potential therapeutic insight for TDP-43-associated neurodegeneration.


Subject(s)
DNA-Binding Proteins/toxicity , RNA, Untranslated/genetics , Transcription Elongation, Genetic , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Female , Genetic Loci , HEK293 Cells , Humans , Male , Models, Biological , Nuclear Proteins/metabolism , Polytene Chromosomes/metabolism , Protein Binding , RNA, Small Nuclear/genetics , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism
12.
Nat Med ; 24(6): 758-769, 2018 06.
Article in English | MEDLINE | ID: mdl-29785026

ABSTRACT

The lysine methyltransferase KMT2C (also known as MLL3), a subunit of the COMPASS complex, implements monomethylation of Lys4 on histone H3 (H3K4) at gene enhancers. KMT2C (hereafter referred to as MLL3) frequently incurs point mutations across a range of human tumor types, but precisely how these lesions alter MLL3 function and contribute to oncogenesis is unclear. Here we report a cancer mutational hotspot in MLL3 within the region encoding its plant homeodomain (PHD) repeats and demonstrate that this domain mediates association of MLL3 with the histone H2A deubiquitinase and tumor suppressor BAP1. Cancer-associated mutations in the sequence encoding the MLL3 PHD repeats disrupt the interaction between MLL3 and BAP1 and correlate with poor patient survival. Cancer cells that had PHD-associated MLL3 mutations or lacked BAP1 showed reduced recruitment of MLL3 and the H3K27 demethylase KDM6A (also known as UTX) to gene enhancers. As a result, inhibition of the H3K27 methyltransferase activity of the Polycomb repressive complex 2 (PRC2) in tumor cells harboring BAP1 or MLL3 mutations restored normal gene expression patterns and impaired cell proliferation in vivo. This study provides mechanistic insight into the oncogenic effects of PHD-associated mutations in MLL3 and suggests that restoration of a balanced state of Polycomb-COMPASS activity may have therapeutic efficacy in tumors that bear mutations in the genes encoding these epigenetic factors.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Epigenesis, Genetic , Polycomb-Group Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Chromatin/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Histone Demethylases/metabolism , Mice, Nude , Mutation/genetics , Nuclear Proteins/metabolism , PHD Zinc Fingers , Protein Binding , Survival Analysis , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
13.
Nat Rev Mol Cell Biol ; 19(7): 464-478, 2018 07.
Article in English | MEDLINE | ID: mdl-29740129

ABSTRACT

The dynamic regulation of transcription elongation by RNA polymerase II (Pol II) is an integral part of the implementation of gene expression programmes during development. In most metazoans, the majority of transcribed genes exhibit transient pausing of Pol II at promoter-proximal regions, and the release of Pol II into gene bodies is controlled by many regulatory factors that respond to environmental and developmental cues. Misregulation of the elongation stage of transcription is implicated in cancer and other human diseases, suggesting that mechanistic understanding of transcription elongation control is therapeutically relevant. In this Review, we discuss the features, establishment and maintenance of Pol II pausing, the transition into productive elongation, the control of transcription elongation by enhancers and by factors of other cellular processes, such as topoisomerases and poly(ADP-ribose) polymerases (PARPs), and the potential of therapeutic targeting of the elongation stage of transcription by Pol II.


Subject(s)
RNA Polymerase II/genetics , Transcription, Genetic/genetics , Animals , Humans , Isomerases/genetics , Promoter Regions, Genetic/genetics
14.
Sci Adv ; 4(1): eaap8747, 2018 01.
Article in English | MEDLINE | ID: mdl-29404406

ABSTRACT

Chromatin regulators control cellular differentiation by orchestrating dynamic developmental gene expression programs, and hence, malfunctions in the regulation of chromatin state contribute to both developmental disorders and disease state. Mll4 (Kmt2d), a member of the COMPASS (COMplex of Proteins ASsociated with Set1) protein family that implements histone H3 lysine 4 monomethylation (H3K4me1) at enhancers, is essential for embryonic development and functions as a pancancer tumor suppressor. We define the roles of Mll4/COMPASS and its catalytic activity in the maintenance and exit of ground-state pluripotency in murine embryonic stem cells (ESCs). Mll4 is required for ESC to exit the naive pluripotent state; however, its intrinsic catalytic activity is dispensable for this process. The depletion of the H3K4 demethylase Lsd1 (Kdm1a) restores the ability of Mll4 null ESCs to transition from naive to primed pluripotency. Thus, we define an opposing regulatory axis, wherein Lsd1 and associated co-repressors directly repress Mll4-activated gene targets. This finding has broad reaching implications for human developmental syndromes and the treatment of tumors carrying Mll4 mutations.


Subject(s)
Cell Differentiation , Enhancer Elements, Genetic , Epigenesis, Genetic , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Mouse Embryonic Stem Cells/metabolism , Multiprotein Complexes/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Self Renewal/genetics , Histone-Lysine N-Methyltransferase/chemistry , Mice , Mice, Knockout , Mouse Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Transcription, Genetic
15.
Nat Genet ; 49(11): 1647-1653, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28967912

ABSTRACT

Histone H3 lysine 4 monomethylation (H3K4me1) is an evolutionarily conserved feature of enhancer chromatin catalyzed by the COMPASS-like methyltransferase family, which includes Trr in Drosophila melanogaster and MLL3 (encoded by KMT2C) and MLL4 (encoded by KMT2D) in mammals. Here we demonstrate that Drosophila embryos expressing catalytically deficient Trr eclose and develop to productive adulthood. Parallel experiments with a trr allele that augments enzyme product specificity show that conversion of H3K4me1 at enhancers to H3K4me2 and H3K4me3 is also compatible with life and results in minimal changes in gene expression. Similarly, loss of the catalytic SET domains of MLL3 and MLL4 in mouse embryonic stem cells (mESCs) does not disrupt self-renewal. Drosophila embryos with trr alleles encoding catalytic mutants manifest subtle developmental abnormalities when subjected to temperature stress or altered cohesin levels. Collectively, our findings suggest that animal development can occur in the context of Trr or mammalian COMPASS-like proteins deficient in H3K4 monomethylation activity and point to a possible role for H3K4me1 on cis-regulatory elements in specific settings to fine-tune transcriptional regulation in response to environmental stress.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Amino Acid Sequence , Animals , Baculoviridae/genetics , CRISPR-Cas Systems , Chromatin/chemistry , Chromatin/metabolism , Drosophila Proteins/deficiency , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Methylation , Mice , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Sf9 Cells , Spodoptera
16.
Science ; 357(6357): 1294-1298, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28860207

ABSTRACT

Gene expression in metazoans is regulated by RNA polymerase II (Pol II) promoter-proximal pausing and its release. Previously, we showed that Pol II-associated factor 1 (PAF1) modulates the release of paused Pol II into productive elongation. Here, we found that PAF1 occupies transcriptional enhancers and restrains hyperactivation of a subset of these enhancers. Enhancer activation as the result of PAF1 loss releases Pol II from paused promoters of nearby PAF1 target genes. Knockout of PAF1-regulated enhancers attenuates the release of paused Pol II on PAF1 target genes without major interference in the establishment of pausing at their cognate promoters. Thus, a subset of enhancers can primarily modulate gene expression by controlling the release of paused Pol II in a PAF1-dependent manner.


Subject(s)
Enhancer Elements, Genetic , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Cell Line, Tumor , Gene Knockout Techniques , Humans , Immediate-Early Proteins/genetics , Nuclear Proteins/genetics , Serpin E2/genetics , Transcription Factors , Up-Regulation
17.
Environ Sci Technol ; 51(15): 8713-8724, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28671818

ABSTRACT

Current environmental monitoring approaches focus primarily on chemical occurrence. However, based on concentration alone, it can be difficult to identify which compounds may be of toxicological concern and should be prioritized for further monitoring, in-depth testing, or management. This can be problematic because toxicological characterization is lacking for many emerging contaminants. New sources of high-throughput screening (HTS) data, such as the ToxCast database, which contains information for over 9000 compounds screened through up to 1100 bioassays, are now available. Integrated analysis of chemical occurrence data with HTS data offers new opportunities to prioritize chemicals, sites, or biological effects for further investigation based on concentrations detected in the environment linked to relative potencies in pathway-based bioassays. As a case study, chemical occurrence data from a 2012 study in the Great Lakes Basin along with the ToxCast effects database were used to calculate exposure-activity ratios (EARs) as a prioritization tool. Technical considerations of data processing and use of the ToxCast database are presented and discussed. EAR prioritization identified multiple sites, biological pathways, and chemicals that warrant further investigation. Prioritized bioactivities from the EAR analysis were linked to discrete adverse outcome pathways to identify potential adverse outcomes and biomarkers for use in subsequent monitoring efforts.


Subject(s)
Biological Assay , Environmental Monitoring , High-Throughput Screening Assays , Toxicity Tests , Biomarkers , Great Lakes Region , Humans , Lakes
18.
Mol Cell ; 65(3): 460-475.e6, 2017 Feb 02.
Article in English | MEDLINE | ID: mdl-28157506

ABSTRACT

The spatiotemporal regulation of gene expression is central for cell-lineage specification during embryonic development and is achieved through the combinatorial action of transcription factors/co-factors and epigenetic states at cis-regulatory elements. Here, we show that in addition to implementing H3K4me3 at promoters of bivalent genes, Mll2 (KMT2B)/COMPASS can also implement H3K4me3 at a subset of non-TSS regulatory elements, a subset of which shares epigenetic signatures of active enhancers. Our mechanistic studies reveal that association of Mll2's CXXC domain with CpG-rich regions plays an instrumental role for chromatin targeting and subsequent implementation of H3K4me3. Although Mll2/COMPASS is required for H3K4me3 implementation on thousands of loci, generation of catalytically mutant MLL2/COMPASS demonstrated that H3K4me3 implemented by this enzyme was essential for expression of a subset of genes, including those functioning in the control of transcriptional programs during embryonic development. Our findings suggest that not all H3K4 trimethylations implemented by MLL2/COMPASS are functionally equivalent.


Subject(s)
DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Germ Cells/cytology , Histones/metabolism , Mouse Embryonic Stem Cells/cytology , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasm Proteins/metabolism , Animals , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Genetic Speciation , Germ Cells/metabolism , HEK293 Cells , Histone-Lysine N-Methyltransferase , Humans , Mice , Mouse Embryonic Stem Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/chemistry , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Protein Domains
19.
Cell ; 168(1-2): 59-72.e13, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-28065413

ABSTRACT

Chromosomal translocations of the mixed-lineage leukemia (MLL) gene with various partner genes result in aggressive leukemia with dismal outcomes. Despite similar expression at the mRNA level from the wild-type and chimeric MLL alleles, the chimeric protein is more stable. We report that UBE2O functions in regulating the stability of wild-type MLL in response to interleukin-1 signaling. Targeting wild-type MLL degradation impedes MLL leukemia cell proliferation, and it downregulates a specific group of target genes of the MLL chimeras and their oncogenic cofactor, the super elongation complex. Pharmacologically inhibiting this pathway substantially delays progression, and it improves survival of murine leukemia through stabilizing wild-type MLL protein, which displaces the MLL chimera from some of its target genes and, therefore, relieves the cellular oncogenic addiction to MLL chimeras. Stabilization of MLL provides us with a paradigm in the development of therapies for aggressive MLL leukemia and perhaps for other cancers caused by translocations.


Subject(s)
Leukemia, Biphenotypic, Acute/drug therapy , Leukemia, Biphenotypic, Acute/metabolism , Proteolysis/drug effects , Animals , Disease Models, Animal , Histone-Lysine N-Methyltransferase/metabolism , Humans , Interleukin-1/metabolism , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/metabolism , Ubiquitin-Conjugating Enzymes
20.
Genes Dev ; 30(1): 92-101, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26728555

ABSTRACT

Genomic imprinting is a critical developmental process characteristic of parent of origin-specific gene expression. It is well accepted that differentially DNA-methylated regions (DMRs) and enhancers are two major classes of cis-elements determining parent of origin-specific gene expression, with each recruiting different sets of transcription factors. Previously, we identified the AF4/FMR2 (AFF) family protein AFF3 within the transcription elongation complex SEC-L3. Here, we report that AFF3 can specifically bind both gametic DMRs (gDMRs) and enhancers within imprinted loci in an allele-specific manner. We identify the molecular regulators involved in the recruitment of AFF3 to gDMRs and provide mechanistic insight into the requirement of AFF3 at an enhancer for the expression of an ∼200-kb polycistronic transcript within the imprinted Dlk1-Dio3 locus. Our data suggest that the heterochromatic environment at the gDMR reinforces silencing of its related enhancer by controlling the binding and activity of AFF3 in an allele-specific manner. In summary, this study provides molecular details about the regulation of dosage-critical imprinted gene expression through the regulated binding of the transcription elongation factor AFF3 between a DMR and an enhancer.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Genomic Imprinting/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Alleles , Animals , Calcium-Binding Proteins , Cell Line , Chromatin Immunoprecipitation , DNA Methylation , Embryonic Stem Cells , Gene Silencing , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...