Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Mol Cell ; 81(21): 4413-4424.e5, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34480849

ABSTRACT

Based on in vitro studies, it has been demonstrated that the DSIF complex, composed of SPT4 and SPT5, regulates the elongation stage of transcription catalyzed by RNA polymerase II (RNA Pol II). The precise cellular function of SPT5 is not clear, because conventional gene depletion strategies for SPT5 result in loss of cellular viability. Using an acute inducible protein depletion strategy to circumvent this issue, we report that SPT5 loss triggers the ubiquitination and proteasomal degradation of the core RNA Pol II subunit RPB1, a process that we show to be evolutionarily conserved from yeast to human cells. RPB1 degradation requires the E3 ligase Cullin 3, the unfoldase VCP/p97, and a novel form of CDK9 kinase complex. Our study demonstrates that SPT5 stabilizes RNA Pol II specifically at promoter-proximal regions, permitting RNA Pol II release from promoters into gene bodies and providing mechanistic insight into the cellular function of SPT5 in safeguarding accurate gene expression.


Subject(s)
Cullin Proteins/metabolism , Nuclear Proteins/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Transcriptional Elongation Factors/metabolism , Animals , Cell Survival , Chromosomal Proteins, Non-Histone/metabolism , Cullin Proteins/chemistry , Fibroblasts/metabolism , Humans , Indoleacetic Acids/chemistry , Mice , Nedd4 Ubiquitin Protein Ligases/chemistry , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/chemistry , Proteome , Proteomics/methods , Ubiquitin-Protein Ligases/chemistry , Valosin Containing Protein/chemistry , Valosin Containing Protein/metabolism
2.
Cancer Discov ; 10(9): 1388-1409, 2020 09.
Article in English | MEDLINE | ID: mdl-32444465

ABSTRACT

Splicing alterations are common in diseases such as cancer, where mutations in splicing factor genes are frequently responsible for aberrant splicing. Here we present an alternative mechanism for splicing regulation in T-cell acute lymphoblastic leukemia (T-ALL) that involves posttranslational stabilization of the splicing machinery via deubiquitination. We demonstrate there are extensive exon skipping changes in disease, affecting proteasomal subunits, cell-cycle regulators, and the RNA machinery. We present that the serine/arginine-rich splicing factors (SRSF), controlling exon skipping, are critical for leukemia cell survival. The ubiquitin-specific peptidase 7 (USP7) regulates SRSF6 protein levels via active deubiquitination, and USP7 inhibition alters the exon skipping pattern and blocks T-ALL growth. The splicing inhibitor H3B-8800 affects splicing of proteasomal transcripts and proteasome activity and acts synergistically with proteasome inhibitors in inhibiting T-ALL growth. Our study provides the proof-of-principle for regulation of splicing factors via deubiquitination and suggests new therapeutic modalities in T-ALL. SIGNIFICANCE: Our study provides a new proof-of-principle for posttranslational regulation of splicing factors independently of mutations in aggressive T-cell leukemia. It further suggests a new drug combination of splicing and proteasomal inhibitors, a concept that might apply to other diseases with or without mutations affecting the splicing machinery.This article is highlighted in the In This Issue feature, p. 1241.


Subject(s)
Alternative Splicing/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Phosphoproteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Serine-Arginine Splicing Factors/metabolism , Ubiquitin-Specific Peptidase 7/metabolism , Alternative Splicing/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Synergism , Exons/genetics , Humans , Jurkat Cells , Male , Mice , Piperazines/pharmacology , Piperazines/therapeutic use , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proof of Concept Study , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Ubiquitination , Xenograft Model Antitumor Assays
3.
Nucleic Acids Res ; 48(1): 421-431, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31724694

ABSTRACT

COMPlex ASsociating with SET1 (COMPASS) is a histone H3 Lys-4 methyltransferase that typically marks the promoter region of actively transcribed genes. COMPASS is a multi-subunit complex in which the catalytic unit, SET1, is required for H3K4 methylation. An important subunit known to regulate SET1 methyltransferase activity is the CxxC zinc finger protein 1 (Cfp1). Cfp1 binds to COMPASS and is critical to maintain high level of H3K4me3 in cells but the mechanisms underlying its stimulatory activity is poorly understood. In this study, we show that Cfp1 only modestly activates COMPASS methyltransferase activity in vitro. Binding of Cfp1 to COMPASS is in part mediated by a new type of monovalent zinc finger (ZnF). This ZnF interacts with the COMPASS's subunits RbBP5 and disruption of this interaction blunts its methyltransferase activity in cells and in vivo. Collectively, our studies reveal that a novel form of ZnF on Cfp1 enables its integration into COMPASS and contributes to epigenetic signaling.


Subject(s)
Fungal Proteins/chemistry , Histone-Lysine N-Methyltransferase/chemistry , Histones/chemistry , Transcription Factors/chemistry , Zinc Fingers , Amino Acid Sequence , Binding Sites , Chaetomium/genetics , Chaetomium/metabolism , Cloning, Molecular , Crystallography, X-Ray , Epigenesis, Genetic , Escherichia coli/genetics , Escherichia coli/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Kinetics , Methylation , Models, Molecular , Promoter Regions, Genetic , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomycetales/genetics , Saccharomycetales/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc/metabolism
4.
Clin Cancer Res ; 25(1): 222-239, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30224337

ABSTRACT

PURPOSE: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disease, affecting children and adults. Chemotherapy treatments show high response rates but have debilitating effects and carry risk of relapse. Previous work implicated NOTCH1 and other oncogenes. However, direct inhibition of these pathways affects healthy tissues and cancer alike. Our goal in this work has been to identify enzymes active in T-ALL whose activity could be targeted for therapeutic purposes. EXPERIMENTAL DESIGN: To identify and characterize new NOTCH1 druggable partners in T-ALL, we coupled studies of the NOTCH1 interactome to expression analysis and a series of functional analyses in cell lines, patient samples, and xenograft models. RESULTS: We demonstrate that ubiquitin-specific protease 7 (USP7) interacts with NOTCH1 and controls leukemia growth by stabilizing the levels of NOTCH1 and JMJD3 histone demethylase. USP7 is highly expressed in T-ALL and is transcriptionally regulated by NOTCH1. In turn, USP7 controls NOTCH1 levels through deubiquitination. USP7 binds oncogenic targets and controls gene expression through stabilization of NOTCH1 and JMJD3 and ultimately H3K27me3 changes. We also show that USP7 and NOTCH1 bind T-ALL superenhancers, and inhibition of USP7 leads to a decrease of the transcriptional levels of NOTCH1 targets and significantly blocks T-ALL cell growth in vitro and in vivo. CONCLUSIONS: These results provide a new model for USP7 deubiquitinase activity through recruitment to oncogenic chromatin loci and regulation of both oncogenic transcription factors and chromatin marks to promote leukemia. Our studies also show that targeting USP7 inhibition could be a therapeutic strategy in aggressive leukemia.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/genetics , Leukemia, T-Cell/genetics , Receptor, Notch1/genetics , Ubiquitin-Specific Peptidase 7/genetics , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Genetic Therapy , Humans , Jurkat Cells , Leukemia, T-Cell/pathology , Leukemia, T-Cell/therapy , Mice , Signal Transduction/genetics , Xenograft Model Antitumor Assays
5.
Structure ; 26(12): 1594-1603.e4, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30270175

ABSTRACT

Dpy-30 is a regulatory subunit controlling the histone methyltransferase activity of the KMT2 enzymes in vivo. Paradoxically, in vitro methyltransferase assays revealed that Dpy-30 only modestly participates in the positive heterotypic allosteric regulation of these methyltransferases. Detailed genome-wide, molecular and structural studies reveal that an extensive network of interactions taking place at the interface between Dpy-30 and Ash2L are critical for the correct placement, genome-wide, of H3K4me2 and H3K4me3 but marginally contribute to the methyltransferase activity of KMT2 enzymes in vitro. Moreover, we show that H3K4me2 peaks persisting following the loss of Dpy-30 are found in regions of highly transcribed genes, highlighting an interplay between Complex of Proteins Associated with SET1 (COMPASS) kinetics and the cycling of RNA polymerase to control H3K4 methylation. Overall, our data suggest that Dpy-30 couples its modest positive heterotypic allosteric regulation of KMT2 methyltransferase activity with its ability to help the positioning of SET1/COMPASS to control epigenetic signaling.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Histones/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Allosteric Regulation , Animals , Binding Sites , Epigenesis, Genetic , HEK293 Cells , Histone-Lysine N-Methyltransferase/metabolism , Humans , Methylation , Models, Molecular , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Yeasts/genetics , Yeasts/metabolism
6.
Cell ; 174(5): 1117-1126.e12, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30100186

ABSTRACT

The methylation of histone 3 lysine 4 (H3K4) is carried out by an evolutionarily conserved family of methyltransferases referred to as complex of proteins associated with Set1 (COMPASS). The activity of the catalytic SET domain (su(var)3-9, enhancer-of-zeste, and trithorax) is endowed through forming a complex with a set of core proteins that are widely shared from yeast to humans. We obtained cryo-electron microscopy (cryo-EM) maps of the yeast Set1/COMPASS core complex at overall 4.0- to 4.4-Å resolution, providing insights into its structural organization and conformational dynamics. The Cps50 C-terminal tail weaves within the complex to provide a central scaffold for assembly. The SET domain, snugly positioned at the junction of the Y-shaped complex, is extensively contacted by Cps60 (Bre2), Cps50 (Swd1), and Cps30 (Swd3). The mobile SET-I motif of the SET domain is engaged by Cps30, explaining its key role in COMPASS catalytic activity toward higher H3K4 methylation states.


Subject(s)
Fungal Proteins/chemistry , Histone Methyltransferases/chemistry , Histones/chemistry , Animals , Catalytic Domain , Chaetomium/chemistry , Chromatin/chemistry , Cryoelectron Microscopy , DNA-Binding Proteins/chemistry , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/chemistry , Humans , Insecta , Intracellular Signaling Peptides and Proteins , Methylation , Protein Subunits , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Software
7.
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
8.
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
9.
Nat Med ; 23(4): 493-500, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28263307

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is a highly aggressive pediatric brainstem tumor characterized by rapid and uniform patient demise. A heterozygous point mutation of histone H3 occurs in more than 80% of these tumors and results in a lysine-to-methionine substitution (H3K27M). Expression of this histone mutant is accompanied by a reduction in the levels of polycomb repressive complex 2 (PRC2)-mediated H3K27 trimethylation (H3K27me3), and this is hypothesized to be a driving event of DIPG oncogenesis. Despite a major loss of H3K27me3, PRC2 activity is still detected in DIPG cells positive for H3K27M. To investigate the functional roles of H3K27M and PRC2 in DIPG pathogenesis, we profiled the epigenome of H3K27M-mutant DIPG cells and found that H3K27M associates with increased H3K27 acetylation (H3K27ac). In accordance with previous biochemical data, the majority of the heterotypic H3K27M-K27ac nucleosomes colocalize with bromodomain proteins at the loci of actively transcribed genes, whereas PRC2 is excluded from these regions; this suggests that H3K27M does not sequester PRC2 on chromatin. Residual PRC2 activity is required to maintain DIPG proliferative potential, by repressing neuronal differentiation and function. Finally, to examine the therapeutic potential of blocking the recruitment of bromodomain proteins by heterotypic H3K27M-K27ac nucleosomes in DIPG cells, we performed treatments in vivo with BET bromodomain inhibitors and demonstrate that they efficiently inhibit tumor progression, thus identifying this class of compounds as potential therapeutics in DIPG.


Subject(s)
Brain Stem Neoplasms/genetics , Chromatin/metabolism , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , Histone Code/genetics , Histones/genetics , Nucleosomes/metabolism , Polycomb Repressive Complex 2/metabolism , RNA-Binding Proteins/metabolism , Acetylation/drug effects , Animals , Azepines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromatin/drug effects , Epigenomics , Gene Expression Regulation, Neoplastic/drug effects , Histone Code/drug effects , Histones/drug effects , Humans , Methylation/drug effects , Mice , Molecular Targeted Therapy , Mutation , Neurogenesis/drug effects , Neurogenesis/genetics , Nucleosomes/drug effects , Polycomb Repressive Complex 2/drug effects , Protein Transport , RNA-Binding Proteins/antagonists & inhibitors , Triazoles/pharmacology , Xenograft Model Antitumor Assays
10.
Elife ; 52016 06 23.
Article in English | MEDLINE | ID: mdl-27336723

ABSTRACT

In yeast and humans, previous experiences can lead to epigenetic transcriptional memory: repressed genes that exhibit mitotically heritable changes in chromatin structure and promoter recruitment of poised RNA polymerase II preinitiation complex (RNAPII PIC), which enhances future reactivation. Here, we show that INO1 memory in yeast is initiated by binding of the Sfl1 transcription factor to the cis-acting Memory Recruitment Sequence, targeting INO1 to the nuclear periphery. Memory requires a remodeled form of the Set1/COMPASS methyltransferase lacking Spp1, which dimethylates histone H3 lysine 4 (H3K4me2). H3K4me2 recruits the SET3C complex, which plays an essential role in maintaining this mark. Finally, while active INO1 is associated with Cdk8(-) Mediator, during memory, Cdk8(+) Mediator recruits poised RNAPII PIC lacking the Kin28 CTD kinase. Aspects of this mechanism are generalizable to yeast and conserved in human cells. Thus, COMPASS and Mediator are repurposed to promote epigenetic transcriptional poising by a highly conserved mechanism.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Fungal , Histone-Lysine N-Methyltransferase/metabolism , Myo-Inositol-1-Phosphate Synthase/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Humans , Mediator Complex/metabolism , Transcription, Genetic
11.
Genes Dev ; 28(2): 115-20, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24402317

ABSTRACT

The stimulation of trimethylation of histone H3 Lys4 (H3K4) by H2B monoubiquitination (H2Bub) has been widely studied, with multiple mechanisms having been proposed for this form of histone cross-talk. Cps35/Swd2 within COMPASS (complex of proteins associated with Set1) is considered to bridge these different processes. However, a truncated form of Set1 (762-Set1) is reported to function in H3K4 trimethylation (H3K4me3) without interacting with Cps35/Swd2, and such cross-talk is attributed to the n-SET domain of Set1 and its interaction with the Cps40/Spp1 subunit of COMPASS. Here, we used biochemical, structural, in vivo, and chromatin immunoprecipitation (ChIP) sequencing (ChIP-seq) approaches to demonstrate that Cps40/Spp1 and the n-SET domain of Set1 are required for the stability of Set1 and not the cross-talk. Furthermore, the apparent wild-type levels of H3K4me3 in the 762-Set1 strain are due to the rogue methylase activity of this mutant, resulting in the mislocalization of H3K4me3 from the promoter-proximal regions to the gene bodies and intergenic regions. We also performed detailed screens and identified yeast strains lacking H2Bub but containing intact H2Bub enzymes that have normal levels of H3K4me3, suggesting that monoubiquitination may not directly stimulate COMPASS but rather works in the context of the PAF and Rad6/Bre1 complexes. Our study demonstrates that the monoubiquitination machinery and Cps35/Swd2 function to focus COMPASS's H3K4me3 activity at promoter-proximal regions in a context-dependent manner.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Saccharomyces cerevisiae/enzymology , Lysine/metabolism , Membrane Proteins/metabolism , Methylation , Phosphoric Monoester Hydrolases/metabolism , Protein Stability , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism
12.
Genes Dev ; 26(23): 2604-20, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23166019

ABSTRACT

Monomethylation of histone H3 on Lys 4 (H3K4me1) and acetylation of histone H3 on Lys 27 (H3K27ac) are histone modifications that are highly enriched over the body of actively transcribed genes and on enhancers. Although in yeast all H3K4 methylation patterns, including H3K4me1, are implemented by Set1/COMPASS (complex of proteins associated with Set1), there are three classes of COMPASS-like complexes in Drosophila that could carry out H3K4me1 on enhancers: dSet1, Trithorax, and Trithorax-related (Trr). Here, we report that Trr, the Drosophila homolog of the mammalian Mll3/4 COMPASS-like complexes, can function as a major H3K4 monomethyltransferase on enhancers in vivo. Loss of Trr results in a global decrease of H3K4me1 and H3K27ac levels in various tissues. Assays with the cut wing margin enhancer implied a functional role for Trr in enhancer-mediated processes. A genome-wide analysis demonstrated that Trr is required to maintain the H3K4me1 and H3K27ac chromatin signature that resembles the histone modification patterns described for enhancers. Furthermore, studies in the mammalian system suggested a role for the Trr homolog Mll3 in similar processes. Since Trr and mammalian Mll3/4 complexes are distinguished by bearing a unique subunit, the H3K27 demethylase UTX, we propose a model in which the H3K4 monomethyltransferases Trr/Mll3/Mll4 and the H3K27 demethylase UTX cooperate to regulate the transition from inactive/poised to active enhancers.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Enhancer Elements, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Animals , Cell Line , Drosophila Proteins/genetics , Genome-Wide Association Study , Histone-Lysine N-Methyltransferase/genetics , Methylation
13.
Genes Dev ; 26(9): 914-9, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22549955

ABSTRACT

Monoubiquitination of histone H2B on Lys 123 (H2BK123ub) is a transient histone modification considered to be essential for establishing H3K4 and H3K79 trimethylation by Set1/COMPASS and Dot1, respectively. Here, we identified Chd1 as a factor that is required for the maintenance of high levels of H2B monoubiquitination, but not for H3K4 and H3K79 trimethylation. Loss of Chd1 results in a substantial loss of H2BK123ub levels with little to no effect on the genome-wide pattern of H3K4 and H3K79 trimethylation. Our data show that nucleosomal occupancy is reduced in gene bodies in both chd1Δ and, as has been shown, K123A mutant backgrounds. We also demonstrated that Chd1's function in maintaining H2BK123ub levels is conserved from yeast to humans. Our study provides evidence that only small levels of H2BK123ub are necessary for full levels of H3K4 and H3K79 trimethylation in vivo and points to a possible role for Chd1 in positively regulating gene expression through promoting nucleosome reassembly coupled with H2B monoubiquitination.


Subject(s)
Gene Expression Regulation, Developmental , Gene Expression Regulation, Fungal , Histones/metabolism , Nucleosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/genetics , Ubiquitination , Cdh1 Proteins , Genome-Wide Association Study , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
14.
Proc Natl Acad Sci U S A ; 108(51): 20526-31, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22158900

ABSTRACT

Histone H3 lysine 4 (H3K4) methylation is catalyzed by the highly evolutionarily conserved multiprotein complex known as Set1/COMPASS or MLL/COMPASS-like complexes from yeast to human, respectively. Here we have reconstituted fully functional yeast Set1/COMPASS and human MLL/COMPASS-like complex in vitro and have identified the minimum subunit composition required for histone H3K4 methylation. These subunits include the methyltransferase C-terminal SET domain of Set1/MLL, Cps60/Ash2L, Cps50/RbBP5, Cps30/WDR5, and Cps25/Dpy30, which are all common components of the COMPASS family from yeast to human. Three-dimensional (3D) cryo-EM reconstructions of the core yeast complex, combined with immunolabeling and two-dimensional (2D) EM analysis of the individual subcomplexes reveal a Y-shaped architecture with Cps50 and Cps30 localizing on the top two adjacent lobes and Cps60-Cps25 forming the base at the bottom. EM analysis of the human complex reveals a striking similarity to its yeast counterpart, suggesting a common subunit organization. The SET domain of Set1 is located at the juncture of Cps50, Cps30, and the Cps60-Cps25 module, lining the walls of a central channel that may act as the platform for catalysis and regulative processing of various degrees of H3K4 methylation. This structural arrangement suggested that COMPASS family members function as exo-methylases, which we have confirmed by in vitro and in vivo studies.


Subject(s)
Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Histones/chemistry , Animals , Conserved Sequence , Cryoelectron Microscopy/methods , DNA Methylation , Histone Methyltransferases , Humans , Imaging, Three-Dimensional , Insecta , Methylation , Microscopy, Electron/methods , Models, Molecular , Molecular Conformation , Myeloid-Lymphoid Leukemia Protein/chemistry , Recombinant Proteins/chemistry , Saccharomyces cerevisiae/genetics
15.
Mol Cell ; 42(1): 118-26, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21474073

ABSTRACT

The expression of genes residing near telomeres is attenuated through telomere position-effect variegation (TPEV). By using a URA3 reporter located at TEL-VII-L of Saccharomyces cerevisiae, it was proposed that the disruptor of telomeric silencing-1 (Dot1) regulates TPEV by catalyzing H3K79 methylation. URA3 reporter assays also indicated that H3K79 methylation is required for HM silencing. Surprisingly, a genome-wide expression analysis of H3K79 methylation-defective mutants identified only a few telomeric genes, such as COS12 at TEL-VII-L, to be subject to H3K79 methylation-dependent natural silencing. Consistently, loss of Dot1 did not globally alter Sir2 or Sir3 occupancy in subtelomeric regions, but only led to some telomere-specific changes. Furthermore, H3K79 methylation by Dot1 did not play a role in the maintenance of natural HML silencing. Therefore, commonly used URA3 reporter assays may not report on natural PEV, and therefore, studies concerning the epigenetic mechanism of silencing in yeast should also employ assays reporting on natural gene expression patterns.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Acetyltransferases/metabolism , Chromosomal Position Effects , Gene Silencing , Genes, Fungal , Genome-Wide Association Study , Histone-Lysine N-Methyltransferase/genetics , Histones/chemistry , Methylation , N-Terminal Acetyltransferase A , Nuclear Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Sirtuin 2/metabolism , Telomere/genetics , Telomere/metabolism
16.
Genes Dev ; 24(6): 574-89, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20203130

ABSTRACT

Epigenetic modifications of chromatin play an important role in the regulation of gene expression. KMT4/Dot1 is a conserved histone methyltransferase capable of methylating chromatin on Lys79 of histone H3 (H3K79). Here we report the identification of a multisubunit Dot1 complex (DotCom), which includes several of the mixed lineage leukemia (MLL) partners in leukemia such as ENL, AF9/MLLT3, AF17/MLLT6, and AF10/MLLT10, as well as the known Wnt pathway modifiers TRRAP, Skp1, and beta-catenin. We demonstrated that the human DotCom is indeed capable of trimethylating H3K79 and, given the association of beta-catenin, Skp1, and TRRAP, we investigated, and found, a role for Dot1 in Wnt/Wingless signaling in an in vivo model system. Knockdown of Dot1 in Drosophila results in decreased expression of a subset of Wingless target genes. Furthermore, the loss of expression for the Drosophila homologs of the Dot1-associated proteins involved in the regulation of H3K79 shows a similar reduction in expression of these Wingless targets. From yeast to human, specific trimethylation of H3K79 by Dot1 requires the monoubiquitination of histone H2B by the Rad6/Bre1 complex. Here, we demonstrate that depletion of Bre1, the E3 ligase required for H2B monoubiquitination, leads specifically to reduced bulk H3K79 trimethylation levels and a reduction in expression of many Wingless targets. Overall, our study describes for the first time the components of DotCom and links the specific regulation of H3K79 trimethylation by Dot1 and its associated factors to the Wnt/Wingless signaling pathway.


Subject(s)
Histones/metabolism , Methyltransferases/metabolism , Signal Transduction , Wnt Proteins/metabolism , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , HeLa Cells , Histone-Lysine N-Methyltransferase , Humans , Methylation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
18.
J Cell Biol ; 186(3): 371-7, 2009 Aug 10.
Article in English | MEDLINE | ID: mdl-19667127

ABSTRACT

Histone H2B monoubiquitination by Rad6/Bre1 is required for the trimethylation of both histone H3K4 and H3K79 by COMPASS and Dot1 methyltransferases, respectively. The dependency of methylation at H3K4 and H3K79 on the monoubiquitination of H2BK123 was recently challenged, and extragenic mutations in the strain background used for previous studies or epitope-tagged proteins were suggested to be the sources of this discrepancy. In this study, we show that H3K4 and H3K79 methylation is solely dependent on H2B monoubiquitination regardless of any additional alteration to the H2B sequence or genome. Furthermore, we report that Y131, one of the yeast histone H2A/H2B shuffle strains widely used for the last decade in the field of chromatin and transcription biology, carries a wild-type copy of each of the HTA2 and HTB2 genes under the GAL1/10 promoter on chromosome II. Therefore, we generated the entire histone H2A and H2B alanine-scanning mutant strains in another background, which does not express wild-type histones.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae , Ubiquitination , Alanine/genetics , Alanine/metabolism , Histones/genetics , Methylation , Mutation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
19.
Mol Cell Biol ; 29(13): 3478-86, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19398585

ABSTRACT

The multiprotein complex Set1/COMPASS is the founding member of the histone H3 lysine 4 (H3K4) methyltransferases, whose human homologs include the MLL and hSet1 complexes. COMPASS can mono-, di-, and trimethylate H3K4, but transitioning to di- and trimethylation requires prior H2B monoubiquitination followed by recruitment of the Cps35 (Swd2) subunit of COMPASS. Another subunit, Cps40 (Spp1), interacts directly with Set1 and is only required for transitioning to trimethylation. To investigate how the Set1 and COMPASS subunits establish the methylation states of H3K4, we generated a homology model of the catalytic domain of Saccharomyces cerevisiae yeast Set1 and identified several key residues within the Set1 catalytic pocket that are capable of regulating COMPASS's activity. We show that Tyr1052, a putative Phe/Tyr switch of Set1, plays an essential role in the regulation of H3K4 trimethylation by COMPASS and that the mutation to phenylalanine (Y1052F) suppresses the loss of Cps40 in H3K4 trimethylation levels, suggesting that Tyr1052 functions together with Cps40. However, the loss of H2B monoubiquitination is not suppressed by this mutation, while Cps40 is stably assembled in COMPASS on chromatin, demonstrating that Tyr1052- and Cps40-mediated H3K4 trimethylation takes place following and independently of H2B monoubiquitination. Our studies provide a molecular basis for the way in which H3K4 trimethylation is regulated by Tyr1052 and the Cps40 subunit of COMPASS.


Subject(s)
DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Phenylalanine/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Tyrosine/metabolism , DNA-Binding Proteins/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Infant , Methylation , Models, Molecular , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Ubiquitination
20.
Antioxid Redox Signal ; 8(5-6): 743-52, 2006.
Article in English | MEDLINE | ID: mdl-16771666

ABSTRACT

DsbB, an Escherichia coli plasma membrane protein, oxidizes DsbA, the protein dithiol oxidant in the periplasm, in conjunction with respiratory quinone molecules. While its two periplasmic regions, in particular the essential Cys41-Cys44 and the Cys104-Cys130 cysteine pairs, have been characterized in considerable detail, little or no information is available about the functional importance of its three cytosolically disposed regions. In this work the authors introduced insertion and substitution mutations into the short ( approximately 6 residue) central cytosolic loop. The purified mutant proteins proved to have two of the essential cysteines reduced and to exhibit the spectroscopic transition of bound ubiquinone constitutively. A thrombin-cleavage site present in a mutant protein called DsbB-T established that the mutant protein had a rearranged Cys41-Cys130 disulfide that would unpair Cys44. Although this covalent structure of DsbB is reminiscent of the DsbB-DsbA intermediate, in which unpaired Cys44 induces the ubiquinone transition, it is inactive because of the premature disulfide rearrangement without involving DsbA. In addition, ubiquione-mediated in vitro oxidation of reduced DsbB-T was aborted at a half-oxidized state, without rapidly producing the fully oxidized enzyme. Thus, the cytosolic loop alterations compromised the catalytic turnover of DsbB in vitro. These observations suggest that the cytosolic loop is important to coordinate the active-site residues of DsbB and ubiquinone to allow their proper reaction cycles.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Ubiquinone/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Binding Sites , Cysteine/chemistry , Disulfides/chemistry , Escherichia coli Proteins/genetics , Membrane Proteins/genetics , Molecular Sequence Data , Mutation , Oxidation-Reduction , Protein Conformation , Sulfhydryl Compounds/chemistry , Ubiquinone/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...