Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Cell ; 185(18): 3458-3458.e1, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36055203

ABSTRACT

The Mediator complex controls RNA polymerase II transcription genome-wide. In humans, Mediator consists of 26 subunits; furthermore, a four-subunit "Mediator kinase module" can reversibly associate with the complex. Mediator structure is generally conserved from yeast to humans, although the human complex is larger, more structurally disordered, and contains metazoan-specific subunits. To view this SnapShot, open or download the PDF.


Subject(s)
Mediator Complex , Animals , Humans , Mediator Complex/metabolism , RNA Polymerase II/metabolism , Yeasts
2.
Nat Rev Mol Cell Biol ; 23(11): 732-749, 2022 11.
Article in English | MEDLINE | ID: mdl-35725906

ABSTRACT

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


Subject(s)
Mediator Complex , RNA Polymerase II , Humans , RNA Polymerase II/metabolism , Mediator Complex/metabolism , Transcription Factors/metabolism , Chromatin/genetics , DNA , Transcription, Genetic
3.
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
4.
Mol Cell ; 84(7): 1180-1182, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38579674

ABSTRACT

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


Subject(s)
RNA Polymerase II , Transcription Factors , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
5.
Nature ; 613(7945): 759-766, 2023 01.
Article in English | MEDLINE | ID: mdl-36631611

ABSTRACT

Protein phosphorylation is one of the most widespread post-translational modifications in biology1,2. With advances in mass-spectrometry-based phosphoproteomics, 90,000 sites of serine and threonine phosphorylation have so far been identified, and several thousand have been associated with human diseases and biological processes3,4. For the vast majority of phosphorylation events, it is not yet known which of the more than 300 protein serine/threonine (Ser/Thr) kinases encoded in the human genome are responsible3. Here we used synthetic peptide libraries to profile the substrate sequence specificity of 303 Ser/Thr kinases, comprising more than 84% of those predicted to be active in humans. Viewed in its entirety, the substrate specificity of the kinome was substantially more diverse than expected and was driven extensively by negative selectivity. We used our kinome-wide dataset to computationally annotate and identify the kinases capable of phosphorylating every reported phosphorylation site in the human Ser/Thr phosphoproteome. For the small minority of phosphosites for which the putative protein kinases involved have been previously reported, our predictions were in excellent agreement. When this approach was applied to examine the signalling response of tissues and cell lines to hormones, growth factors, targeted inhibitors and environmental or genetic perturbations, it revealed unexpected insights into pathway complexity and compensation. Overall, these studies reveal the intrinsic substrate specificity of the human Ser/Thr kinome, illuminate cellular signalling responses and provide a resource to link phosphorylation events to biological pathways.


Subject(s)
Phosphoproteins , Protein Serine-Threonine Kinases , Proteome , Serine , Threonine , Humans , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Substrate Specificity , Threonine/metabolism , Proteome/chemistry , Proteome/metabolism , Datasets as Topic , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Cell Line , Phosphoserine/metabolism , Phosphothreonine/metabolism
6.
Mol Cell ; 78(4): 785-793.e8, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32229306

ABSTRACT

RNA polymerase II (RNAPII) transcription is governed by the pre-initiation complex (PIC), which contains TFIIA, TFIIB, TFIID, TFIIE, TFIIF, TFIIH, RNAPII, and Mediator. After initiation, RNAPII enzymes pause after transcribing less than 100 bases; precisely how RNAPII pausing is enforced and regulated remains unclear. To address specific mechanistic questions, we reconstituted human RNAPII promoter-proximal pausing in vitro, entirely with purified factors (no extracts). As expected, NELF and DSIF increased pausing, and P-TEFb promoted pause release. Unexpectedly, the PIC alone was sufficient to reconstitute pausing, suggesting RNAPII pausing is an inherent PIC function. In agreement, pausing was lost upon replacement of the TFIID complex with TATA-binding protein (TBP), and PRO-seq experiments revealed widespread disruption of RNAPII pausing upon acute depletion (t = 60 min) of TFIID subunits in human or Drosophila cells. These results establish a TFIID requirement for RNAPII pausing and suggest pause regulatory factors may function directly or indirectly through TFIID.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Promoter Regions, Genetic , RNA Polymerase II/genetics , Transcription Factor TFIID/metabolism , Transcription, Genetic , Animals , Drosophila/genetics , Drosophila Proteins/genetics , HCT116 Cells , Humans , Protein Binding , RNA Polymerase II/metabolism , Transcription Factor TFIID/genetics
7.
Genes Dev ; 34(7-8): 465-488, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32238450

ABSTRACT

RNA polymerase II (Pol II) transcribes all protein-coding genes and many noncoding RNAs in eukaryotic genomes. Although Pol II is a complex, 12-subunit enzyme, it lacks the ability to initiate transcription and cannot consistently transcribe through long DNA sequences. To execute these essential functions, an array of proteins and protein complexes interact with Pol II to regulate its activity. In this review, we detail the structure and mechanism of over a dozen factors that govern Pol II initiation (e.g., TFIID, TFIIH, and Mediator), pausing, and elongation (e.g., DSIF, NELF, PAF, and P-TEFb). The structural basis for Pol II transcription regulation has advanced rapidly in the past decade, largely due to technological innovations in cryoelectron microscopy. Here, we summarize a wealth of structural and functional data that have enabled a deeper understanding of Pol II transcription mechanisms; we also highlight mechanistic questions that remain unanswered or controversial.


Subject(s)
RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Transcription, Genetic/genetics , Animals , Enzyme Activation , Humans , Protein Binding , Protein Structure, Quaternary , Research/trends
8.
Genes Dev ; 34(21-22): 1452-1473, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33060135

ABSTRACT

CDK7 associates with the 10-subunit TFIIH complex and regulates transcription by phosphorylating the C-terminal domain (CTD) of RNA polymerase II (RNAPII). Few additional CDK7 substrates are known. Here, using the covalent inhibitor SY-351 and quantitative phosphoproteomics, we identified CDK7 kinase substrates in human cells. Among hundreds of high-confidence targets, the vast majority are unique to CDK7 (i.e., distinct from other transcription-associated kinases), with a subset that suggest novel cellular functions. Transcription-associated factors were predominant CDK7 substrates, including SF3B1, U2AF2, and other splicing components. Accordingly, widespread and diverse splicing defects, such as alternative exon inclusion and intron retention, were characterized in CDK7-inhibited cells. Combined with biochemical assays, we establish that CDK7 directly activates other transcription-associated kinases CDK9, CDK12, and CDK13, invoking a "master regulator" role in transcription. We further demonstrate that TFIIH restricts CDK7 kinase function to the RNAPII CTD, whereas other substrates (e.g., SPT5 and SF3B1) are phosphorylated by the three-subunit CDK-activating kinase (CAK; CCNH, MAT1, and CDK7). These results suggest new models for CDK7 function in transcription and implicate CAK dissociation from TFIIH as essential for kinase activation. This straightforward regulatory strategy ensures CDK7 activation is spatially and temporally linked to transcription, and may apply toward other transcription-associated kinases.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Models, Biological , Transcription Factor TFIIH/metabolism , Transcription, Genetic/genetics , Alternative Splicing/genetics , Cell Survival/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Enzyme Activation/genetics , HL-60 Cells , Humans , Cyclin-Dependent Kinase-Activating Kinase
9.
Nat Rev Mol Cell Biol ; 16(3): 155-66, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25693131

ABSTRACT

The RNA polymerase II (Pol II) enzyme transcribes all protein-coding and most non-coding RNA genes and is globally regulated by Mediator - a large, conformationally flexible protein complex with a variable subunit composition (for example, a four-subunit cyclin-dependent kinase 8 module can reversibly associate with it). These biochemical characteristics are fundamentally important for Mediator's ability to control various processes that are important for transcription, including the organization of chromatin architecture and the regulation of Pol II pre-initiation, initiation, re-initiation, pausing and elongation. Although Mediator exists in all eukaryotes, a variety of Mediator functions seem to be specific to metazoans, which is indicative of more diverse regulatory requirements.


Subject(s)
Leiomyoma/genetics , Mediator Complex/genetics , RNA Polymerase II/genetics , Transcription Factors/genetics , Transcription, Genetic , Uterine Neoplasms/genetics , Animals , Chromatin/chemistry , Chromatin/metabolism , Female , Gene Expression Regulation , Histones/chemistry , Histones/metabolism , Humans , Leiomyoma/metabolism , Leiomyoma/pathology , Mediator Complex/metabolism , RNA Polymerase II/metabolism , RNA, Untranslated/metabolism , Signal Transduction , Transcription Factors/metabolism , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
10.
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
11.
Mol Cell ; 76(3): 485-499.e8, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31495563

ABSTRACT

Transcriptional responses to external stimuli remain poorly understood. Using global nuclear run-on followed by sequencing (GRO-seq) and precision nuclear run-on sequencing (PRO-seq), we show that CDK8 kinase activity promotes RNA polymerase II pause release in response to interferon-γ (IFN-γ), a universal cytokine involved in immunity and tumor surveillance. The Mediator kinase module contains CDK8 or CDK19, which are presumed to be functionally redundant. We implemented cortistatin A, chemical genetics, transcriptomics, and other methods to decouple their function while assessing enzymatic versus structural roles. Unexpectedly, CDK8 and CDK19 regulated different gene sets via distinct mechanisms. CDK8-dependent regulation required its kinase activity, whereas CDK19 governed IFN-γ responses through its scaffolding function (i.e., it was kinase independent). Accordingly, CDK8, not CDK19, phosphorylates the STAT1 transcription factor (TF) during IFN-γ stimulation, and CDK8 kinase inhibition blocked activation of JAK-STAT pathway TFs. Cytokines such as IFN-γ rapidly mobilize TFs to "reprogram" cellular transcription; our results implicate CDK8 and CDK19 as essential for this transcriptional reprogramming.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/metabolism , Fibroblasts/drug effects , Interferon-gamma/pharmacology , Transcription, Genetic/drug effects , Animals , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinases/genetics , Fibroblasts/enzymology , Fibroblasts/virology , HCT116 Cells , Host-Pathogen Interactions , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , RNA Polymerase II/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Vesiculovirus/pathogenicity
12.
Trends Biochem Sci ; 47(4): 314-327, 2022 04.
Article in English | MEDLINE | ID: mdl-35193797

ABSTRACT

The Mediator complex controls RNA polymerase II (pol II) activity by coordinating the assembly of pol II regulatory factors at transcription start sites and by mediating interactions between enhancer-bound transcription factors (TFs) and the pol II enzyme. Mediator structure and function is completely altered upon binding the Mediator kinase module, a multi-subunit complex that contains CDK8 or its vertebrate-specific paralog CDK19. Here, we review the mechanisms by which the Mediator kinase module controls pol II transcription, emphasizing its impact on TF activity, pol II elongation, enhancer function, and chromatin architecture. We also highlight how the Mediator kinase module integrates signaling pathways with transcription to enable rapid, stimulus-specific responses, as well as its links to human disease.


Subject(s)
Cyclin-Dependent Kinase 8 , Mediator Complex , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/metabolism , Humans , Mediator Complex/genetics , Mediator Complex/metabolism , RNA Polymerase II/metabolism , Signal Transduction , Transcription, Genetic
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.
Mol Cell ; 68(1): 1-2, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28985500

ABSTRACT

Using improved techniques, Baptista et al. (2017) and Warfield et al. (2017) revisit fundamental questions about SAGA and TFIID function in yeast. They conclude that each complex independently contributes to the expression of all genes transcribed by RNA polymerase II.


Subject(s)
RNA Polymerase II/genetics , Transcription Factor TFIID/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , TATA-Binding Protein Associated Factors/genetics , Transcription Factors/genetics , Transcription, Genetic
15.
PLoS Biol ; 19(8): e3001364, 2021 08.
Article in English | MEDLINE | ID: mdl-34351910

ABSTRACT

The naturally occurring Δ40p53 isoform heterotetramerizes with wild-type p53 (WTp53) to regulate development, aging, and stress responses. How Δ40p53 alters WTp53 function remains enigmatic because their co-expression causes tetramer heterogeneity. We circumvented this issue with a well-tested strategy that expressed Δ40p53:WTp53 as a single transcript, ensuring a 2:2 tetramer stoichiometry. Human MCF10A cell lines expressing Δ40p53:WTp53, WTp53, or WTp53:WTp53 (as controls) from the native TP53 locus were examined with transcriptomics (precision nuclear run-on sequencing [PRO-seq] and RNA sequencing [RNA-seq]), metabolomics, and other methods. Δ40p53:WTp53 was transcriptionally active, and, although phenotypically similar to WTp53 under normal conditions, it failed to induce growth arrest upon Nutlin-induced p53 activation. This occurred via Δ40p53:WTp53-dependent inhibition of enhancer RNA (eRNA) transcription and subsequent failure to induce mRNA biogenesis, despite similar genomic occupancy to WTp53. A different stimulus (5-fluorouracil [5FU]) also showed Δ40p53:WTp53-specific changes in mRNA induction; however, other transcription factors (TFs; e.g., E2F2) could then drive the response, yielding similar outcomes vs. WTp53. Our results establish that Δ40p53 tempers WTp53 function to enable compensatory responses by other stimulus-specific TFs. Such modulation of WTp53 activity may be an essential physiological function for Δ40p53. Moreover, Δ40p53:WTp53 functional distinctions uncovered herein suggest an eRNA requirement for mRNA biogenesis and that human p53 evolved as a tetramer to support eRNA transcription.


Subject(s)
Tumor Suppressor Protein p53/metabolism , Cell Line , Fluorouracil , Genes, p53 , Humans , Imidazoles , Piperazines , Protein Isoforms , Protein Structure, Quaternary , Transcription Factors/metabolism , Transcription, Genetic , Transcriptome
16.
J Biol Chem ; 298(10): 102433, 2022 10.
Article in English | MEDLINE | ID: mdl-36041630

ABSTRACT

TFIIH is an evolutionarily conserved complex that plays central roles in both RNA polymerase II (pol II) transcription and DNA repair. As an integral component of the pol II preinitiation complex, TFIIH regulates pol II enzyme activity in numerous ways. The TFIIH subunit XPB/Ssl2 is an ATP-dependent DNA translocase that stimulates promoter opening prior to transcription initiation. Crosslinking-mass spectrometry and cryo-EM results have shown a conserved interaction network involving XPB/Ssl2 and the C-terminal Hub region of the TFIIH p52/Tfb2 subunit, but the functional significance of specific residues is unclear. Here, we systematically mutagenized the HubA region of Tfb2 and screened for growth phenotypes in a TFB6 deletion background in Saccharomyces cerevisiae. We identified six lethal and 12 conditional mutants. Slow growth phenotypes of all but three conditional mutants were relieved in the presence of TFB6, thus identifying a functional interaction between Tfb2 HubA mutants and Tfb6, a protein that dissociates Ssl2 from TFIIH. Our biochemical analysis of Tfb2 mutants with severe growth phenotypes revealed defects in Ssl2 association, with similar results in human cells. Further characterization of these tfb2 mutant cells revealed defects in GAL gene induction, and reduced occupancy of TFIIH and pol II at GAL gene promoters, suggesting that functionally competent TFIIH is required for proper pol II recruitment to preinitiation complexes in vivo. Consistent with recent structural models of TFIIH, our results identify key residues in the p52/Tfb2 HubA domain that are required for stable incorporation of XPB/Ssl2 into TFIIH and for pol II transcription.


Subject(s)
DNA Helicases , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Factor TFIIH , Humans , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair , Mutagenesis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factor TFIIH/genetics , Transcription Factor TFIIH/metabolism , Transcription, Genetic
17.
Mol Cell ; 57(6): 952-954, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25794613

ABSTRACT

In this issue of Molecular Cell, Yi et al. (2015) use biochemical assays and cryo-EM to determine the molecular architecture of an estrogen receptor (ERα) co-activator complex bound to DNA.


Subject(s)
E1A-Associated p300 Protein/chemistry , Estrogen Receptor alpha/chemistry , Nuclear Receptor Coactivator 3/chemistry , Humans
18.
Mol Cell ; 59(5): 794-806, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26340423

ABSTRACT

TFIIH is essential for both RNA polymerase II transcription and DNA repair, and mutations in TFIIH can result in human disease. Here, we determine the molecular architecture of human and yeast TFIIH by an integrative approach using chemical crosslinking/mass spectrometry (CXMS) data, biochemical analyses, and previously published electron microscopy maps. We identified four new conserved "topological regions" that function as hubs for TFIIH assembly and more than 35 conserved topological features within TFIIH, illuminating a network of interactions involved in TFIIH assembly and regulation of its activities. We show that one of these conserved regions, the p62/Tfb1 Anchor region, directly interacts with the DNA helicase subunit XPD/Rad3 in native TFIIH and is required for the integrity and function of TFIIH. We also reveal the structural basis for defects in patients with xeroderma pigmentosum and trichothiodystrophy, with mutations found at the interface between the p62 Anchor region and the XPD subunit.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factor TFIIH/chemistry , Transcription Factor TFIIH/metabolism , Cross-Linking Reagents , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair , Humans , Mass Spectrometry , Models, Molecular , Mutation , Protein Interaction Domains and Motifs , Protein Subunits , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factor TFIIH/genetics , Transcription Factors, TFII/chemistry , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism , Transcription, Genetic , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum Group D Protein/chemistry , Xeroderma Pigmentosum Group D Protein/genetics , Xeroderma Pigmentosum Group D Protein/metabolism
19.
Immunity ; 38(2): 250-62, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23352233

ABSTRACT

Gene regulation by cytokine-activated transcription factors of the signal transducer and activator of transcription (STAT) family requires serine phosphorylation within the transactivation domain (TAD). STAT1 and STAT3 TAD phosphorylation occurs upon promoter binding by an unknown kinase. Here, we show that the cyclin-dependent kinase 8 (CDK8) module of the Mediator complex phosphorylated regulatory sites within the TADs of STAT1, STAT3, and STAT5, including S727 within the STAT1 TAD in the interferon (IFN) signaling pathway. We also observed a CDK8 requirement for IFN-γ-inducible antiviral responses. Microarray analyses revealed that CDK8-mediated STAT1 phosphorylation positively or negatively regulated over 40% of IFN-γ-responsive genes, and RNA polymerase II occupancy correlated with gene expression changes. This divergent regulation occurred despite similar CDK8 occupancy at both S727 phosphorylation-dependent and -independent genes. These data identify CDK8 as a key regulator of STAT1 and antiviral responses and suggest a general role for CDK8 in STAT-mediated transcription. As such, CDK8 represents a promising target for therapeutic manipulation of cytokine responses.


Subject(s)
Cyclin-Dependent Kinase 8/genetics , Gene Expression Regulation/drug effects , Interferon-gamma/pharmacology , STAT1 Transcription Factor/genetics , Animals , Cyclin-Dependent Kinase 8/immunology , Cyclin-Dependent Kinase 8/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/virology , Hep G2 Cells , Humans , Interferon-gamma/immunology , Interleukin-6/immunology , Interleukin-6/pharmacology , Mice , Oligonucleotide Array Sequence Analysis , Phosphorylation , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA Polymerase II/immunology , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Vesiculovirus/physiology
20.
Nature ; 526(7572): 273-276, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26416749

ABSTRACT

Super-enhancers (SEs), which are composed of large clusters of enhancers densely loaded with the Mediator complex, transcription factors and chromatin regulators, drive high expression of genes implicated in cell identity and disease, such as lineage-controlling transcription factors and oncogenes. BRD4 and CDK7 are positive regulators of SE-mediated transcription. By contrast, negative regulators of SE-associated genes have not been well described. Here we show that the Mediator-associated kinases cyclin-dependent kinase 8 (CDK8) and CDK19 restrain increased activation of key SE-associated genes in acute myeloid leukaemia (AML) cells. We report that the natural product cortistatin A (CA) selectively inhibits Mediator kinases, has anti-leukaemic activity in vitro and in vivo, and disproportionately induces upregulation of SE-associated genes in CA-sensitive AML cell lines but not in CA-insensitive cell lines. In AML cells, CA upregulated SE-associated genes with tumour suppressor and lineage-controlling functions, including the transcription factors CEBPA, IRF8, IRF1 and ETV6 (refs 6-8). The BRD4 inhibitor I-BET151 downregulated these SE-associated genes, yet also has anti-leukaemic activity. Individually increasing or decreasing the expression of these transcription factors suppressed AML cell growth, providing evidence that leukaemia cells are sensitive to the dosage of SE-associated genes. Our results demonstrate that Mediator kinases can negatively regulate SE-associated gene expression in specific cell types, and can be pharmacologically targeted as a therapeutic approach to AML.


Subject(s)
Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, Neoplasm/genetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Animals , Cell Cycle Proteins , Cell Division/drug effects , Cell Line, Tumor , Cell Lineage/drug effects , Cell Lineage/genetics , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/metabolism , Disease Progression , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred Strains , Mice, SCID , Nuclear Proteins/antagonists & inhibitors , Polycyclic Compounds/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL