Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(11): eadk7160, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38489358

ABSTRACT

During development, cells make switch-like decisions to activate new gene programs specifying cell lineage. The mechanisms underlying these decisive choices remain unclear. Here, we show that the cardiovascular transcriptional coactivator myocardin (MYOCD) activates cell identity genes by concentration-dependent and switch-like formation of transcriptional condensates. MYOCD forms such condensates and activates cell identity genes at critical concentration thresholds achieved during smooth muscle cell and cardiomyocyte differentiation. The carboxyl-terminal disordered region of MYOCD is necessary and sufficient for condensate formation. Disrupting this region's ability to form condensates disrupts gene activation and smooth muscle cell reprogramming. Rescuing condensate formation by replacing this region with disordered regions from functionally unrelated proteins rescues gene activation and smooth muscle cell reprogramming. Our findings demonstrate that MYOCD condensate formation is required for gene activation during cardiovascular differentiation. We propose that the formation of transcriptional condensates at critical concentrations of cell type-specific regulators provides a molecular switch underlying the activation of key cell identity genes during development.


Subject(s)
Myocytes, Smooth Muscle , Transcription Factors , Cell Lineage/genetics , Cell Differentiation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Myocytes, Smooth Muscle/metabolism , Transcriptional Activation
3.
Cell ; 186(2): 327-345.e28, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36603581

ABSTRACT

Components of transcriptional machinery are selectively partitioned into specific condensates, often mediated by protein disorder, yet we know little about how this specificity is achieved. Here, we show that condensates composed of the intrinsically disordered region (IDR) of MED1 selectively partition RNA polymerase II together with its positive allosteric regulators while excluding negative regulators. This selective compartmentalization is sufficient to activate transcription and is required for gene activation during a cell-state transition. The IDRs of partitioned proteins are necessary and sufficient for selective compartmentalization and require alternating blocks of charged amino acids. Disrupting this charge pattern prevents partitioning, whereas adding the pattern to proteins promotes partitioning with functional consequences for gene activation. IDRs with similar patterned charge blocks show similar partitioning and function. These findings demonstrate that disorder-mediated interactions can selectively compartmentalize specific functionally related proteins from a complex mixture of biomolecules, leading to regulation of a biochemical pathway.


Subject(s)
Intrinsically Disordered Proteins , RNA Polymerase II , Transcription, Genetic , Intrinsically Disordered Proteins/metabolism , RNA Polymerase II/metabolism , Transcriptional Activation , Animals , Mice
4.
J Mol Biol ; 435(5): 167971, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36690068

ABSTRACT

In the past almost 15 years, we witnessed the birth of a new scientific field focused on the existence, formation, biological functions, and disease associations of membraneless bodies in cells, now referred to as biomolecular condensates. Pioneering studies from several laboratories [reviewed in1-3] supported a model wherein biomolecular condensates associated with diverse biological processes form through the process of phase separation. These and other findings that followed have revolutionized our understanding of how biomolecules are organized in space and time within cells to perform myriad biological functions, including cell fate determination, signal transduction, endocytosis, regulation of gene expression and protein translation, and regulation of RNA metabolism. Further, condensates formed through aberrant phase transitions have been associated with numerous human diseases, prominently including neurodegeneration and cancer. While in some cases, rigorous evidence supports links between formation of biomolecular condensates through phase separation and biological functions, in many others such links are less robustly supported, which has led to rightful scrutiny of the generality of the roles of phase separation in biology and disease.4-7 During a week-long workshop in March 2022 at the Telluride Science Research Center (TSRC) in Telluride, Colorado, ∼25 scientists addressed key questions surrounding the biomolecular condensates field. Herein, we present insights gained through these discussions, addressing topics including, roles of condensates in diverse biological processes and systems, and normal and disease cell states, their applications to synthetic biology, and the potential for therapeutically targeting biomolecular condensates.


Subject(s)
Biomolecular Condensates , Disease , Phase Transition , Humans
5.
Mol Cell ; 82(21): 3965-3967, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36332603

ABSTRACT

In this issue of Molecular Cell, Song et al. demonstrate that mutations to the YEATS domain of ENL aberrantly activate gene expression by forming condensates on specific genomic loci. By using diverse experimental approaches, the authors dissect the molecular underpinnings of ENL mutant condensate formation.


Subject(s)
Transcription Factors , Transcriptional Elongation Factors , Transcriptional Elongation Factors/genetics , Transcription Factors/metabolism , Protein Domains
6.
Mol Cell ; 81(22): 4577-4578, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34798044

ABSTRACT

Shi et al. (2021) demonstrate that tumor-suppressive and developmental functions of UTX require an intrinsically disordered region (IDR) capable of condensate formation. These results provide further evidence for the functional role of IDR-mediated spatial organization in regulating gene expression in development and disease.


Subject(s)
Intrinsically Disordered Proteins , Gene Expression , Intrinsically Disordered Proteins/genetics
7.
Sci Transl Med ; 13(614): eabc0497, 2021 Oct 06.
Article in English | MEDLINE | ID: mdl-34613815

ABSTRACT

Childhood posterior fossa group A ependymomas (PFAs) have limited treatment options and bear dismal prognoses compared to group B ependymomas (PFBs). PFAs overexpress the oncohistone-like protein EZHIP (enhancer of Zeste homologs inhibitory protein), causing global reduction of repressive histone H3 lysine 27 trimethylation (H3K27me3), similar to the oncohistone H3K27M. Integrated metabolic analyses in patient-derived cells and tumors, single-cell RNA sequencing of tumors, and noninvasive metabolic imaging in patients demonstrated enhanced glycolysis and tricarboxylic acid (TCA) cycle metabolism in PFAs. Furthermore, high glycolytic gene expression in PFAs was associated with a poor outcome. PFAs demonstrated high EZHIP expression associated with poor prognosis and elevated activating mark histone H3 lysine 27 acetylation (H3K27ac). Genomic H3K27ac was enriched in PFAs at key glycolytic and TCA cycle­related genes including hexokinase-2 and pyruvate dehydrogenase. Similarly, mouse neuronal stem cells (NSCs) expressing wild-type EZHIP (EZHIP-WT) versus catalytically attenuated EZHIP-M406K demonstrated H3K27ac enrichment at hexokinase-2 and pyruvate dehydrogenase, accompanied by enhanced glycolysis and TCA cycle metabolism. AMPKα-2, a key component of the metabolic regulator AMP-activated protein kinase (AMPK), also showed H3K27ac enrichment in PFAs and EZHIP-WT NSCs. The AMPK activator metformin lowered EZHIP protein concentrations, increased H3K27me3, suppressed TCA cycle metabolism, and showed therapeutic efficacy in vitro and in vivo in patient-derived PFA xenografts in mice. Our data indicate that PFAs and EZHIP-WT­expressing NSCs are characterized by enhanced glycolysis and TCA cycle metabolism. Repurposing the antidiabetic drug metformin lowered pathogenic EZHIP, increased H3K27me3, and suppressed tumor growth, suggesting that targeting integrated metabolic/epigenetic pathways is a potential therapeutic strategy for treating childhood ependymomas.


Subject(s)
Ependymoma , Histones , Animals , Child , Ependymoma/genetics , Epigenesis, Genetic , Epigenomics , Histones/genetics , Humans , Metabolic Networks and Pathways , Mice
8.
Dev Cell ; 55(1): 84-96, 2020 10 12.
Article in English | MEDLINE | ID: mdl-33049213

ABSTRACT

Activating the right gene at the right time and place is essential for development. Emerging evidence suggests that this process is regulated by the mesoscale compartmentalization of the gene-control machinery, RNA polymerase II and its cofactors, within biomolecular condensates. Coupling gene activity to the reversible and dynamic process of condensate formation is proposed to enable the robust and precise changes in gene-regulatory programs during signaling and development. The macromolecular features that enable condensates and the regulatory pathways that control them are dysregulated in disease, highlighting their importance for normal physiology. In this review, we will discuss the role of condensates in gene activation; the multivalent features of protein, RNA, and DNA that enable reversible condensate formation; and how these processes are utilized in normal and disease biology. Understanding the regulation of condensates promises to provide novel insights into how organization of the gene-control machinery regulates development and disease.


Subject(s)
Gene Expression/physiology , Organelles/metabolism , RNA/metabolism , Transcriptional Activation/physiology , Animals , DNA/metabolism , Humans , Proteins/metabolism
9.
Trends Biochem Sci ; 45(11): 961-977, 2020 11.
Article in English | MEDLINE | ID: mdl-32684431

ABSTRACT

Nuclear processes such as DNA replication, transcription, and RNA processing each depend on the concerted action of many different protein and RNA molecules. How biomolecules with shared functions find their way to specific locations has been assumed to occur largely by diffusion-mediated collisions. Recent studies have shown that many nuclear processes occur within condensates that compartmentalize and concentrate the protein and RNA molecules required for each process, typically at specific genomic loci. These condensates have common features and emergent properties that provide the cell with regulatory capabilities beyond canonical molecular regulatory mechanisms. We describe here the shared features of nuclear condensates, the components that produce locus-specific condensates, elements of specificity, and the emerging understanding of mechanisms regulating these compartments.


Subject(s)
Cell Nucleus/metabolism , DNA/metabolism , Proteins/metabolism , RNA/metabolism , Cell Nucleus/chemistry , DNA/chemistry , Humans , Proteins/chemistry , RNA/chemistry
10.
Science ; 368(6497): 1386-1392, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32554597

ABSTRACT

The nucleus contains diverse phase-separated condensates that compartmentalize and concentrate biomolecules with distinct physicochemical properties. Here, we investigated whether condensates concentrate small-molecule cancer therapeutics such that their pharmacodynamic properties are altered. We found that antineoplastic drugs become concentrated in specific protein condensates in vitro and that this occurs through physicochemical properties independent of the drug target. This behavior was also observed in tumor cells, where drug partitioning influenced drug activity. Altering the properties of the condensate was found to affect the concentration and activity of drugs. These results suggest that selective partitioning and concentration of small molecules within condensates contributes to drug pharmacodynamics and that further understanding of this phenomenon may facilitate advances in disease therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Nucleus/metabolism , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Neoplasms/metabolism , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Mol Cell ; 76(5): 753-766.e6, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31563432

ABSTRACT

The gene expression programs that define the identity of each cell are controlled by master transcription factors (TFs) that bind cell-type-specific enhancers, as well as signaling factors, which bring extracellular stimuli to these enhancers. Recent studies have revealed that master TFs form phase-separated condensates with the Mediator coactivator at super-enhancers. Here, we present evidence that signaling factors for the WNT, TGF-ß, and JAK/STAT pathways use their intrinsically disordered regions (IDRs) to enter and concentrate in Mediator condensates at super-enhancers. We show that the WNT coactivator ß-catenin interacts both with components of condensates and DNA-binding factors to selectively occupy super-enhancer-associated genes. We propose that the cell-type specificity of the response to signaling is mediated in part by the IDRs of the signaling factors, which cause these factors to partition into condensates established by the master TFs and Mediator at genes with prominent roles in cell identity.


Subject(s)
Enhancer Elements, Genetic/genetics , Mediator Complex/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Gene Expression Regulation/physiology , Humans , Intrinsically Disordered Proteins/metabolism , Mediator Complex/physiology , STAT Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Smad3 Protein/metabolism , TGF-beta Superfamily Proteins/metabolism , Transcription, Genetic , Wnt Signaling Pathway , beta Catenin/metabolism
12.
Mol Cell ; 75(3): 549-561.e7, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398323

ABSTRACT

Enhancers are DNA elements that are bound by transcription factors (TFs), which recruit coactivators and the transcriptional machinery to genes. Phase-separated condensates of TFs and coactivators have been implicated in assembling the transcription machinery at particular enhancers, yet the role of DNA sequence in this process has not been explored. We show that DNA sequences encoding TF binding site number, density, and affinity above sharply defined thresholds drive condensation of TFs and coactivators. A combination of specific structured (TF-DNA) and weak multivalent (TF-coactivator) interactions allows for condensates to form at particular genomic loci determined by the DNA sequence and the complement of expressed TFs. DNA features found to drive condensation promote enhancer activity and transcription in cells. Our study provides a framework to understand how the genome can scaffold transcriptional condensates at specific loci and how the universal phenomenon of phase separation might regulate this process.


Subject(s)
Chromatin/genetics , Enhancer Elements, Genetic , Transcription Factors/genetics , Transcription, Genetic , Animals , Base Sequence/genetics , Binding Sites/genetics , DNA/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genomics , Mice , Mouse Embryonic Stem Cells
13.
Nature ; 572(7770): 543-548, 2019 08.
Article in English | MEDLINE | ID: mdl-31391587

ABSTRACT

The synthesis of pre-mRNA by RNA polymerase II (Pol II) involves the formation of a transcription initiation complex, and a transition to an elongation complex1-4. The large subunit of Pol II contains an intrinsically disordered C-terminal domain that is phosphorylated by cyclin-dependent kinases during the transition from initiation to elongation, thus influencing the interaction of the C-terminal domain with different components of the initiation or the RNA-splicing apparatus5,6. Recent observations suggest that this model provides only a partial picture of the effects of phosphorylation of the C-terminal domain7-12. Both the transcription-initiation machinery and the splicing machinery can form phase-separated condensates that contain large numbers of component molecules: hundreds of molecules of Pol II and mediator are concentrated in condensates at super-enhancers7,8, and large numbers of splicing factors are concentrated in nuclear speckles, some of which occur at highly active transcription sites9-12. Here we investigate whether the phosphorylation of the Pol II C-terminal domain regulates the incorporation of Pol II into phase-separated condensates that are associated with transcription initiation and splicing. We find that the hypophosphorylated C-terminal domain of Pol II is incorporated into mediator condensates and that phosphorylation by regulatory cyclin-dependent kinases reduces this incorporation. We also find that the hyperphosphorylated C-terminal domain is preferentially incorporated into condensates that are formed by splicing factors. These results suggest that phosphorylation of the Pol II C-terminal domain drives an exchange from condensates that are involved in transcription initiation to those that are involved in RNA processing, and implicates phosphorylation as a mechanism that regulates condensate preference.


Subject(s)
Mediator Complex/chemistry , Mediator Complex/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , RNA Splicing , Transcription, Genetic , Animals , Cell Line , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Humans , Mediator Complex/genetics , Mice , Phosphorylation , Protein Domains , RNA Polymerase II/genetics , RNA Splicing Factors/chemistry , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism
14.
Cell ; 175(7): 1842-1855.e16, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30449618

ABSTRACT

Gene expression is controlled by transcription factors (TFs) that consist of DNA-binding domains (DBDs) and activation domains (ADs). The DBDs have been well characterized, but little is known about the mechanisms by which ADs effect gene activation. Here, we report that diverse ADs form phase-separated condensates with the Mediator coactivator. For the OCT4 and GCN4 TFs, we show that the ability to form phase-separated droplets with Mediator in vitro and the ability to activate genes in vivo are dependent on the same amino acid residues. For the estrogen receptor (ER), a ligand-dependent activator, we show that estrogen enhances phase separation with Mediator, again linking phase separation with gene activation. These results suggest that diverse TFs can interact with Mediator through the phase-separating capacity of their ADs and that formation of condensates with Mediator is involved in gene activation.


Subject(s)
Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Receptors, Estrogen/metabolism , Transcriptional Activation/physiology , Animals , HEK293 Cells , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Octamer Transcription Factor-3/genetics , Protein Domains , Receptors, Estrogen/genetics
15.
Science ; 361(6400)2018 07 27.
Article in English | MEDLINE | ID: mdl-29930091

ABSTRACT

Super-enhancers (SEs) are clusters of enhancers that cooperatively assemble a high density of the transcriptional apparatus to drive robust expression of genes with prominent roles in cell identity. Here we demonstrate that the SE-enriched transcriptional coactivators BRD4 and MED1 form nuclear puncta at SEs that exhibit properties of liquid-like condensates and are disrupted by chemicals that perturb condensates. The intrinsically disordered regions (IDRs) of BRD4 and MED1 can form phase-separated droplets, and MED1-IDR droplets can compartmentalize and concentrate the transcription apparatus from nuclear extracts. These results support the idea that coactivators form phase-separated condensates at SEs that compartmentalize and concentrate the transcription apparatus, suggest a role for coactivator IDRs in this process, and offer insights into mechanisms involved in the control of key cell-identity genes.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Intrinsically Disordered Proteins/metabolism , Mediator Complex Subunit 1/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Conserved Sequence , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic/drug effects , Fluorescence Recovery After Photobleaching , Gene Expression Regulation/drug effects , Glycols/pharmacology , HEK293 Cells , Humans , Immunoprecipitation , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Mediator Complex Subunit 1/chemistry , Mediator Complex Subunit 1/genetics , Mice , Molecular Imaging , NIH 3T3 Cells , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Serine/chemistry , Serine/genetics , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
17.
J Allergy Clin Immunol ; 140(2): 474-485, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28108335

ABSTRACT

BACKGROUND: Mast cells are key effector cells in allergic reactions. When activated to degranulate, they release a plethora of bioactive compounds from their secretory granules, including mast cell-restricted proteases such as tryptase. In a previous study, we showed that tryptase, in addition to its intragranular location, can be found within the nuclei of mast cells where it truncates core histones at their N-terminal ends. OBJECTIVE: Considering that the N-terminal portions of the core histones constitute sites for posttranslational modifications of major epigenetic impact, we evaluated whether histone truncation by tryptase could have an impact on epigenetic events in mast cells. METHODS: Mast cells were cultured from wild-type and tryptase null mice, followed by an assessment of their profile of epigenetic histone modifications and their phenotypic characteristics. RESULTS: We show that tryptase truncates nucleosomal histone 3 and histone 2B (H2B) and that its absence results in accumulation of the epigenetic mark, lysine 5-acetylated H2B. Intriguingly, the accumulation of lysine 5-acetylated H2B was cell age-dependent and was associated with a profound upregulation of markers of non-mast cell lineages, loss of proliferative control, chromatin remodeling as well as extensive morphological alterations. CONCLUSIONS: These findings introduce tryptase-catalyzed histone clipping as a novel epigenetic regulatory mechanism, which in the mast cell context may be crucial for maintaining cellular identity.


Subject(s)
Histones/metabolism , Mast Cells/metabolism , Tryptases/metabolism , Acetylation , Anacardic Acids/pharmacology , Animals , Cathepsin G/genetics , Cells, Cultured , Epigenesis, Genetic , Gene Expression Regulation , Histone Deacetylase Inhibitors/pharmacology , Lysine/metabolism , Mast Cells/drug effects , Mice, Inbred C57BL , Mice, Knockout , Proteoglycans/genetics , Tryptases/genetics , Vesicular Transport Proteins/genetics
18.
Nat Rev Mol Cell Biol ; 18(2): 90-101, 2017 02.
Article in English | MEDLINE | ID: mdl-27924077

ABSTRACT

Eight types of short-chain Lys acylations have recently been identified on histones: propionylation, butyrylation, 2-hydroxyisobutyrylation, succinylation, malonylation, glutarylation, crotonylation and ß-hydroxybutyrylation. Emerging evidence suggests that these histone modifications affect gene expression and are structurally and functionally different from the widely studied histone Lys acetylation. In this Review, we discuss the regulation of non-acetyl histone acylation by enzymatic and metabolic mechanisms, the acylation 'reader' proteins that mediate the effects of different acylations and their physiological functions, which include signal-dependent gene activation, spermatogenesis, tissue injury and metabolic stress. We propose a model to explain our present understanding of how differential histone acylation is regulated by the metabolism of the different acyl-CoA forms, which in turn modulates the regulation of gene expression.


Subject(s)
Gene Expression Regulation , Histones/chemistry , Histones/metabolism , Acetyl Coenzyme A/metabolism , Acyl Coenzyme A/metabolism , Acylation , Animals , Fatty Acids, Volatile/metabolism , Histones/genetics , Humans , Lysine/metabolism , Male , Protein Domains , Protein Processing, Post-Translational , Spermatogenesis , Stress, Physiological
19.
Mol Cell ; 62(2): 181-193, 2016 04 21.
Article in English | MEDLINE | ID: mdl-27105114

ABSTRACT

Recognition of histone covalent modifications by chromatin-binding protein modules ("readers") constitutes a major mechanism for epigenetic regulation, typified by bromodomains that bind acetyllysine. Non-acetyl histone lysine acylations (e.g., crotonylation, butyrylation, propionylation) have been recently identified, but readers that prefer these acylations have not been characterized. Here we report that the AF9 YEATS domain displays selectively higher binding affinity for crotonyllysine over acetyllysine. Structural studies revealed an extended aromatic sandwiching cage with crotonyl specificity arising from π-aromatic and hydrophobic interactions between crotonyl and aromatic rings. These features are conserved among the YEATS, but not the bromodomains. Using a cell-based model, we showed that AF9 co-localizes with crotonylated histone H3 and positively regulates gene expression in a YEATS domain-dependent manner. Our studies define the evolutionarily conserved YEATS domain as a family of crotonyllysine readers and specifically demonstrate that the YEATS domain of AF9 directly links histone crotonylation to active transcription.


Subject(s)
Crotonates/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Transcription, Genetic , Transcriptional Activation , Acetylation , Animals , Binding Sites , Chromatin Assembly and Disassembly , Epigenesis, Genetic , HEK293 Cells , Histones/chemistry , Histones/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Lysine , Mice , Models, Molecular , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Domains , RAW 264.7 Cells , RNA-Binding Proteins/metabolism , Transcription Factors , Transfection
20.
Mol Cell ; 58(2): 203-15, 2015 04 16.
Article in English | MEDLINE | ID: mdl-25818647

ABSTRACT

Acetylation of histones at DNA regulatory elements plays a critical role in transcriptional activation. Histones are also modified by other acyl moieties, including crotonyl, yet the mechanisms that govern acetylation versus crotonylation and the functional consequences of this "choice" remain unclear. We show that the coactivator p300 has both crotonyltransferase and acetyltransferase activities, and that p300-catalyzed histone crotonylation directly stimulates transcription to a greater degree than histone acetylation. Levels of histone crotonylation are regulated by the cellular concentration of crotonyl-CoA, which can be altered through genetic and environmental perturbations. In a cell-based model of transcriptional activation, increasing or decreasing the cellular concentration of crotonyl-CoA leads to enhanced or diminished gene expression, respectively, which correlates with the levels of histone crotonylation flanking the regulatory elements of activated genes. Our findings support a general principle wherein differential histone acylation (i.e., acetylation versus crotonylation) couples cellular metabolism to the regulation of gene expression.


Subject(s)
Acyl Coenzyme A/metabolism , E1A-Associated p300 Protein/metabolism , Histones/metabolism , Macrophages/immunology , RNA, Messenger/metabolism , Transcriptional Activation , Acetate-CoA Ligase/genetics , Acetate-CoA Ligase/metabolism , Acetylation , Acyl Coenzyme A/genetics , Cell Line , Cell-Free System , E1A-Associated p300 Protein/genetics , HEK293 Cells , HeLa Cells , Humans , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...