Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
1.
Nat Commun ; 15(1): 5335, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38914563

ABSTRACT

The NuA3 complex is a major regulator of gene transcription and the cell cycle in yeast. Five core subunits are required for complex assembly and function, but it remains unclear how these subunits interact to form the complex. Here, we report that the Taf14 subunit of the NuA3 complex binds to two other subunits of the complex, Yng1 and Sas3, and describe the molecular mechanism by which the extra-terminal domain of Taf14 recognizes the conserved motif present in Yng1 and Sas3. Structural, biochemical, and mutational analyses show that two motifs are sandwiched between the two extra-terminal domains of Taf14. The head-to-toe dimeric complex enhances the DNA binding activity of Taf14, and the formation of the hetero-dimer involving the motifs of Yng1 and Sas3 is driven by sequence complementarity. In vivo assays in yeast demonstrate that the interactions of Taf14 with both Sas3 and Yng1 are required for proper function of the NuA3 complex in gene transcription and DNA repair. Our findings suggest a potential basis for the assembly of three core subunits of the NuA3 complex, Taf14, Yng1 and Sas3.


Subject(s)
Protein Binding , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcription Factor TFIID/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/chemistry , Protein Subunits/metabolism , Protein Subunits/genetics , TATA-Binding Protein Associated Factors/metabolism , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/chemistry , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Protein Multimerization , Models, Molecular , Transcription, Genetic , Amino Acid Sequence
2.
Nat Commun ; 15(1): 4128, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750015

ABSTRACT

Mechanisms of functional cross-talk between global transcriptional repression and efficient DNA damage repair during genotoxic stress are poorly known. In this study, using human AF9 as representative of Super Elongation Complex (SEC) components, we delineate detailed mechanisms of these processes. Mechanistically, we describe that Poly-Serine domain-mediated oligomerization is pre-requisite for AF9 YEATS domain-mediated TFIID interaction-dependent SEC recruitment at the promoter-proximal region for release of paused RNA polymerase II. Interestingly, during genotoxic stress, CaMKII-mediated phosphorylation-dependent nuclear export of AF9-specific deacetylase HDAC5 enhances concomitant PCAF-mediated acetylation of K339 residue. This causes monomerization of AF9 and reduces TFIID interaction for transcriptional downregulation. Furthermore, the K339 acetylation-dependent enhanced AF9-DNA-PKc interaction leads to phosphorylation at S395 residue which reduces AF9-SEC interaction resulting in transcriptional downregulation and efficient repair of DNA damage. After repair, nuclear re-entry of HDAC5 reduces AF9 acetylation and restores its TFIID and SEC interaction to restart transcription.


Subject(s)
DNA Damage , DNA Repair , Histone Deacetylases , Protein Processing, Post-Translational , Transcription, Genetic , Humans , Acetylation , Phosphorylation , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , RNA Polymerase II/metabolism , Transcription Factor TFIID/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/chemistry , Protein Multimerization , HEK293 Cells , HeLa Cells , Transcriptional Elongation Factors/metabolism , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/chemistry
3.
Angew Chem Int Ed Engl ; 63(32): e202404645, 2024 08 05.
Article in English | MEDLINE | ID: mdl-38801173

ABSTRACT

Phenotypic assays detect small-molecule bioactivity at functionally relevant cellular sites, and inherently cover a variety of targets and mechanisms of action. They can uncover new small molecule-target pairs and may give rise to novel biological insights. By means of an osteoblast differentiation assay which employs a Hedgehog (Hh) signaling agonist as stimulus and which monitors an endogenous marker for osteoblasts, we identified a pyrrolo[3,4-g]quinoline (PQ) pseudo-natural product (PNP) class of osteogenesis inhibitors. The most potent PQ, termed Tafbromin, impairs canonical Hh signaling and modulates osteoblast differentiation through binding to the bromodomain 2 of the TATA-box binding protein-associated factor 1 (TAF1). Tafbromin is the most selective TAF1 bromodomain 2 ligand and promises to be an invaluable tool for the study of biological processes mediated by TAF1(2) bromodomains.


Subject(s)
TATA-Binding Protein Associated Factors , Transcription Factor TFIID , TATA-Binding Protein Associated Factors/metabolism , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/antagonists & inhibitors , Humans , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/antagonists & inhibitors , Biological Products/chemistry , Biological Products/pharmacology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/cytology , Cell Differentiation/drug effects , Quinolines/chemistry , Quinolines/pharmacology , Molecular Structure
4.
Curr Opin Struct Biol ; 75: 102404, 2022 08.
Article in English | MEDLINE | ID: mdl-35700575

ABSTRACT

RNA polymerase II (Pol II)-mediated transcription in eukaryotic cells starts with assembly of preinitiation complex (PIC) on core promoter, a DNA sequence of ∼100 base pairs. The transcription PIC consists of Pol II and general transcription factors TFIID, TFIIA, TFIIB, TFIIF, TFIIE, and TFIIH. Previous structural studies focused on PIC assembled on TATA box promoters with TFIID replaced by its subunit, TATA box-binding protein (TBP). However, the megadalton TFIID complex is essential for promoter recognition, TBP loading onto promoter, and PIC assembly for almost all Pol II-mediated transcription, especially on the TATA-less promoters, which account for ∼85% of core promoters of human coding genes. The functions of TFIID could not be replaced by TBP. The recent breakthrough in structure determination of TFIID-based PIC complexes in different assembly stages revealed mechanistic insights into PIC assembly on TATA box and TATA-less promotes and provided a framework for further investigation of transcription initiation.


Subject(s)
RNA Polymerase II , Transcription Factor TFIID , Transcription Initiation, Genetic , Humans , RNA Polymerase II/chemistry , TATA Box , TATA-Box Binding Protein/chemistry , Transcription Factor TFIIA/chemistry , Transcription Factor TFIID/chemistry
5.
J Biol Chem ; 298(6): 101963, 2022 06.
Article in English | MEDLINE | ID: mdl-35452682

ABSTRACT

Formation of transcription factor (TF)-coregulator complexes is a key step in transcriptional regulation, with coregulators having essential functions as hub nodes in molecular networks. How specificity and selectivity are maintained in these nodes remain open questions. In this work, we addressed specificity in transcriptional networks using complexes formed between TFs and αα-hubs, which are defined by a common αα-hairpin secondary structure motif, as a model. Using NMR spectroscopy and binding thermodynamics, we analyzed the structure, dynamics, stability, and ligand-binding properties of the Arabidopsis thaliana RST domains from TAF4 and known binding partner RCD1, and the TAFH domain from human TAF4, allowing comparison across species, functions, and architectural contexts. While these αα-hubs shared the αα-hairpin motif, they differed in length and orientation of accessory helices as well as in their thermodynamic profiles of ligand binding. Whereas biologically relevant RCD1-ligand pairs displayed high affinity driven by enthalpy, TAF4-ligand interactions were entropy driven and exhibited less binding-induced structuring. We in addition identified a thermal unfolding state with a structured core for all three domains, although the temperature sensitivity differed. Thermal stability studies suggested that initial unfolding of the RCD1-RST domain localized around helix 1, lending this region structural malleability, while effects in TAF4-RST were more stochastic, suggesting variability in structural adaptability upon binding. Collectively, our results support a model in which hub structure, flexibility, and binding thermodynamics contribute to αα-hub-TF binding specificity, a finding of general relevance to the understanding of coregulator-ligand interactions and interactome sizes.


Subject(s)
Arabidopsis Proteins/chemistry , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/chemistry , Transcription Factors, TFII/chemistry , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Humans , Ligands , Nuclear Proteins/metabolism , Protein Binding , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Transcription Factors/metabolism , Transcription Factors, TFII/metabolism
6.
J Biol Chem ; 297(5): 101326, 2021 11.
Article in English | MEDLINE | ID: mdl-34688663

ABSTRACT

Bromodomain protein 4 (BRD4) is a transcriptional and epigenetic regulator that is a therapeutic target in many cancers and inflammatory diseases. BRD4 plays important roles in transcription as an active kinase, which phosphorylates the carboxy-terminal domain (CTD) of RNA polymerase II (Pol II), the proto-oncogene c-MYC, and transcription factors TAF7 and CDK9. BRD4 is also a passive scaffold that recruits transcription factors. Despite these well-established functions, there has been little characterization of BRD4's biophysical properties or its kinase activity. We report here that the 156 kD mouse BRD4 exists in an extended dimeric conformation with a sedimentation coefficient of ∼6.7 S and a high frictional ratio. Deletion of the conserved B motif (aa 503-548) disrupts BRD4's dimerization. BRD4 kinase activity maps to amino acids 351 to 598, which span bromodomain-2, the B motif, and the BID domain (BD2-B-BID) and contributes to the in vivo phosphorylation of its substrates. As further assessed by analytical ultracentrifugation, BRD4 directly binds purified Pol II CTD. Importantly, the conserved A motif of BRD4 is essential for phosphorylation of Pol II CTD, but not for phosphorylation of TAF7, mapping its binding site to the A motif. Peptides of the viral MLV integrase (MLVIN) protein and cellular histone lysine methyltransferase, NSD3, which have been shown by NMR to bind to the extra-terminal (ET) domain, also are phosphorylated by BRD4. Thus, BRD4 has multiple distinct substrate-binding sites and a common kinase domain. These results provide new insights into the structure and kinase function of BRD4.


Subject(s)
Nuclear Proteins/chemistry , Protein Kinases/chemistry , Protein Multimerization , Transcription Factors/chemistry , Amino Acid Motifs , Animals , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Structure, Quaternary , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
J Biol Chem ; 297(5): 101288, 2021 11.
Article in English | MEDLINE | ID: mdl-34634302

ABSTRACT

The human general transcription factor TFIID is composed of the TATA-binding protein (TBP) and 13 TBP-associated factors (TAFs). In eukaryotic cells, TFIID is thought to nucleate RNA polymerase II (Pol II) preinitiation complex formation on all protein coding gene promoters and thus, be crucial for Pol II transcription. TFIID is composed of three lobes, named A, B, and C. A 5TAF core complex can be assembled in vitro constituting a building block for the further assembly of either lobe A or B in TFIID. Structural studies showed that TAF8 forms a histone fold pair with TAF10 in lobe B and participates in connecting lobe B to lobe C. To better understand the role of TAF8 in TFIID, we have investigated the requirement of the different regions of TAF8 for the in vitro assembly of lobe B and C and the importance of certain TAF8 regions for mouse embryonic stem cell (ESC) viability. We have identified a region of TAF8 distinct from the histone fold domain important for assembling with the 5TAF core complex in lobe B. We also delineated four more regions of TAF8 each individually required for interacting with TAF2 in lobe C. Moreover, CRISPR/Cas9-mediated gene editing indicated that the 5TAF core-interacting TAF8 domain and the proline-rich domain of TAF8 that interacts with TAF2 are both required for mouse embryonic stem cell survival. Thus, our study defines distinct TAF8 regions involved in connecting TFIID lobe B to lobe C that appear crucial for TFIID function and consequent ESC survival.


Subject(s)
Mouse Embryonic Stem Cells/metabolism , Protein Folding , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Cell Survival , Humans , Mice , Protein Domains , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
8.
Science ; 372(6546)2021 06 04.
Article in English | MEDLINE | ID: mdl-33958484

ABSTRACT

The 1.3-megadalton transcription factor IID (TFIID) is required for preinitiation complex (PIC) assembly and RNA polymerase II (Pol II)-mediated transcription initiation on almost all genes. The 26-subunit Mediator stimulates transcription and cyclin-dependent kinase 7 (CDK7)-mediated phosphorylation of the Pol II C-terminal domain (CTD). We determined the structures of human Mediator in the Tail module-extended (at near-atomic resolution) and Tail-bent conformations and structures of TFIID-based PIC-Mediator (76 polypeptides, ~4.1 megadaltons) in four distinct conformations. PIC-Mediator assembly induces concerted reorganization (Head-tilting and Middle-down) of Mediator and creates a Head-Middle sandwich, which stabilizes two CTD segments and brings CTD to CDK7 for phosphorylation; this suggests a CTD-gating mechanism favorable for phosphorylation. The TFIID-based PIC architecture modulates Mediator organization and TFIIH stabilization, underscoring the importance of TFIID in orchestrating PIC-Mediator assembly.


Subject(s)
Mediator Complex/chemistry , RNA Polymerase II/chemistry , Transcription Factor TFIID/chemistry , Transcription Initiation, Genetic , Cryoelectron Microscopy , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , Humans , Mediator Complex/metabolism , Mediator Complex Subunit 1/chemistry , Models, Molecular , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Conformation, alpha-Helical , Protein Domains , Protein Folding , Protein Structure, Quaternary , Protein Subunits/chemistry , RNA Polymerase II/metabolism , Transcription Factor TFIID/metabolism , Transcription Factor TFIIH/chemistry , Transcription Factor TFIIH/metabolism , Cyclin-Dependent Kinase-Activating Kinase
9.
Science ; 372(6541)2021 04 30.
Article in English | MEDLINE | ID: mdl-33795473

ABSTRACT

Transcription factor IID (TFIID) recognizes core promoters and supports preinitiation complex (PIC) assembly for RNA polymerase II (Pol II)-mediated eukaryotic transcription. We determined the structures of human TFIID-based PIC in three stepwise assembly states and revealed two-track PIC assembly: stepwise promoter deposition to Pol II and extensive modular reorganization on track I (on TATA-TFIID-binding element promoters) versus direct promoter deposition on track II (on TATA-only and TATA-less promoters). The two tracks converge at an ~50-subunit holo PIC in identical conformation, whereby TFIID stabilizes PIC organization and supports loading of cyclin-dependent kinase (CDK)-activating kinase (CAK) onto Pol II and CAK-mediated phosphorylation of the Pol II carboxyl-terminal domain. Unexpectedly, TBP of TFIID similarly bends TATA box and TATA-less promoters in PIC. Our study provides structural visualization of stepwise PIC assembly on highly diversified promoters.


Subject(s)
Multiprotein Complexes/chemistry , Promoter Regions, Genetic , Transcription Factor TFIID/chemistry , Transcription Initiation, Genetic , Animals , Apoptosis Regulatory Proteins/genetics , Corticotropin-Releasing Hormone/genetics , Cryoelectron Microscopy , Cyclin-Dependent Kinases/chemistry , HEK293 Cells , Humans , Phosphorylation , Protein Binding , Protein Domains , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA Polymerase II/chemistry , Swine , Urocortins/genetics
10.
Protein Expr Purif ; 184: 105887, 2021 08.
Article in English | MEDLINE | ID: mdl-33836240

ABSTRACT

The general transcription factor TFIID is a multiprotein complex that is essential for specific transcription initiation by RNA polymerase II. It is composed of the TATA box-binding protein (TBP) and ~13 different TBP-associated factors (TAFs). Purification of TFIID free of other general transcription factors and coactivators is essential to analyze the transcription regulatory mechanisms in reconstituted systems in vitro. A breakthrough in TFIID purification was the generation of HeLa cell lines that express a FLAG epitope-tagged TBP subunit and immunopurification protocols with monoclonal anti-FLAG antibodies. Purification of TFIID from HeLa nuclear extracts generally required a two-step purification procedure involving phosphocellulose P11 chromatography followed by anti-flag M2 affinity purification (Chiang et al., 1993; Ge et al., 1996) [1,2]. Here we show first that the MED26 (CRSP70) coactivator subunit of Mediator co-purifies with TFIID in the above two-step protocol and interacts strongly with TFIID under high salt conditions. We further show that a MED26-free TFIID complex can be obtained by including a simple additional DE52 chromatography step following P11 fractionation. Thus, we demonstrate that MED26 strongly interacts with TFIID and recommend the use of a P11-DE52-M2 resin affinity three-step purification procedure to obtain MED26-free TFIID for analyzing Mediator-dependent transcription regulatory mechanisms in purified transcription systems in vitro.


Subject(s)
TATA-Binding Protein Associated Factors , Transcription Factor TFIID , HeLa Cells , Humans , Mediator Complex/chemistry , Mediator Complex/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , TATA-Binding Protein Associated Factors/biosynthesis , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/isolation & purification , Transcription Factor TFIID/biosynthesis , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics , Transcription Factor TFIID/isolation & purification
11.
J Biol Chem ; 296: 100226, 2021.
Article in English | MEDLINE | ID: mdl-33361159

ABSTRACT

Hub proteins are central nodes in protein-protein interaction networks with critical importance to all living organisms. Recently, a new group of folded hub domains, the αα-hubs, was defined based on a shared αα-hairpin supersecondary structural foundation. The members PAH, RST, TAFH, NCBD, and HHD are found in large proteins such as Sin3, RCD1, TAF4, CBP, and harmonin, which organize disordered transcriptional regulators and membrane scaffolds in interactomes of importance to human diseases and plant quality. In this review, studies of structures, functions, and complexes across the αα-hubs are described and compared to provide a unified description of the group. This analysis expands the associated molecular concepts of "one domain-one binding site", motif-based ligand binding, and coupled folding and binding of intrinsically disordered ligands to additional concepts of importance to signal fidelity. These include context, motif reversibility, multivalency, complex heterogeneity, synergistic αα-hub:ligand folding, accessory binding sites, and supramodules. We propose that these multifaceted protein-protein interaction properties are made possible by the characteristics of the αα-hub fold, including supersite properties, dynamics, variable topologies, accessory helices, and malleability and abetted by adaptability of the disordered ligands. Critically, these features provide additional filters for specificity. With the presentations of new concepts, this review opens for new research questions addressing properties across the group, which are driven from concepts discovered in studies of the individual members. Combined, the members of the αα-hubs are ideal models for deconvoluting signal fidelity maintained by folded hubs and their interactions with intrinsically disordered ligands.


Subject(s)
Arabidopsis Proteins/chemistry , Cell Cycle Proteins/chemistry , Cytoskeletal Proteins/chemistry , Intrinsically Disordered Proteins/chemistry , Sin3 Histone Deacetylase and Corepressor Complex/chemistry , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/chemistry , Transcription Factors, TFII/chemistry , Transcription Factors/chemistry , p300-CBP Transcription Factors/chemistry , Animals , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Humans , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Folding , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/genetics , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
12.
Nat Commun ; 11(1): 4206, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32826896

ABSTRACT

Saccharomyces cerevisiae TBP associated factor 14 (Taf14) is a well-studied transcriptional regulator that controls diverse physiological processes and that physically interacts with at least seven nuclear complexes in yeast. Despite multiple previous Taf14 structural studies, the nature of its disparate transcriptional regulatory functions remains opaque. Here, we demonstrate that the extra-terminal (ET) domain of Taf14 (Taf14ET) recognizes a common motif in multiple transcriptional coactivator proteins from several nuclear complexes, including RSC, SWI/SNF, INO80, NuA3, TFIID, and TFIIF. Moreover, we show that such partner binding promotes liquid-liquid phase separation (LLPS) of Taf14ET, in a mechanism common to YEATS-associated ET domains (e.g., AF9ET) but not Bromo-associated ET domains from BET-family proteins. Thus, beyond identifying the molecular mechanism by which Taf14ET associates with many transcriptional regulators, our study suggests that Taf14 may function as a versatile nuclear hub that orchestrates transcriptional machineries to spatiotemporally regulate diverse cellular pathways.


Subject(s)
Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factor TFIID/metabolism , Carrier Proteins , Cell Cycle Proteins/metabolism , Cluster Analysis , DNA-Binding Proteins , Epigenomics , Gene Expression Regulation, Fungal , Models, Molecular , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation , Protein Domains , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics , Transcription Factors/metabolism
13.
Epigenetics Chromatin ; 13(1): 24, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32460824

ABSTRACT

BACKGROUND: The YEATS domain is a highly conserved protein structure that interacts with acetylated and crotonylated lysine residues in N-terminal tails of histones. The budding yeast genome encodes three YEATS domain proteins (Taf14, Yaf9, and Sas5) that are all the subunits of different complexes involved in histone acetylation, gene transcription, and chromatin remodeling. As the strains deficient in all these three genes are inviable, it has been proposed that the YEATS domain is essential in yeast. In this study we investigate in more detail the requirement of YEATS domain proteins for yeast survival and the possible roles of Taf14 YEATS domain in the regulation of gene transcription. RESULTS: We found that YEATS domains are not essential for the survival of Saccharomyces cerevisiae cells. Although the full deletion of all YEATS proteins is lethal in yeast, we show that the viability of cells can be restored by the expression of the YEATS-less version of Taf14 protein. We also explore the in vivo functions of Taf14 protein and show that the primary role of its YEATS domain is to stabilize the transcription pre-initiation complex (PIC). Our results indicate that Taf14-mediated interactions become crucial for PIC formation in rpb9Δ cells, where the recruitment of TFIIF to the PIC is hampered. Although H3 K9 residue has been identified as the interaction site of the Taf14 YEATS domain in vitro, we found that it is not the only interaction target in vivo. CONCLUSIONS: Lethality of YEATS-deficient cells can be rescued by the expression of truncated Taf14 protein lacking the entire YEATS domain, indicating that the YEATS domains are not required for cell survival. The YEATS domain of Taf14 participates in PIC stabilization and acetylated/crotonylated H3K9 is not the critical target of the Taf14 YEATS domain in vivo.


Subject(s)
Saccharomyces cerevisiae Proteins/metabolism , Transcription Factor TFIID/metabolism , Transcription Initiation, Genetic , Binding Sites , Histones/metabolism , Protein Binding , Protein Domains , Protein Stability , RNA Polymerase II/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics
14.
Nature ; 577(7792): 711-716, 2020 01.
Article in English | MEDLINE | ID: mdl-31969704

ABSTRACT

SAGA (Spt-Ada-Gcn5-acetyltransferase) is a 19-subunit complex that stimulates transcription via two chromatin-modifying enzymatic modules and by delivering the TATA box binding protein (TBP) to nucleate the pre-initiation complex on DNA, a pivotal event in the expression of protein-encoding genes1. Here we present the structure of yeast SAGA with bound TBP. The core of the complex is resolved at 3.5 Å resolution (0.143 Fourier shell correlation). The structure reveals the intricate network of interactions that coordinate the different functional domains of SAGA and resolves an octamer of histone-fold domains at the core of SAGA. This deformed octamer deviates considerably from the symmetrical analogue in the nucleosome and is precisely tuned to establish a peripheral site for TBP, where steric hindrance represses binding of spurious DNA. Complementary biochemical analysis points to a mechanism for TBP delivery and release from SAGA that requires transcription factor IIA and whose efficiency correlates with the affinity of DNA to TBP. We provide the foundations for understanding the specific delivery of TBP to gene promoters and the multiple roles of SAGA in regulating gene expression.


Subject(s)
Fungal Proteins/chemistry , Fungal Proteins/metabolism , Pichia , Promoter Regions, Genetic/genetics , TATA-Box Binding Protein/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , Binding Sites , DNA, Fungal/chemistry , DNA, Fungal/metabolism , Gene Expression Regulation, Fungal , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/metabolism , Histones/chemistry , Histones/metabolism , Models, Molecular , Pichia/chemistry , Pichia/genetics , Protein Binding , Protein Conformation , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , TATA-Box Binding Protein/chemistry , Transcription Factor TFIIA/chemistry , Transcription Factor TFIIA/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/metabolism
15.
Curr Opin Struct Biol ; 61: 17-24, 2020 04.
Article in English | MEDLINE | ID: mdl-31751889

ABSTRACT

TFIID is a large multiprotein assembly that serves as a general transcription factor for transcription initiation by eukaryotic RNA polymerase II (Pol II). TFIID is involved in the recognition of the core promoter sequences and neighboring chromatin marks, and can interact with gene-specific activators and repressors. In order to obtain a better molecular and mechanistic understanding of the function of TFIID, its structure has been pursued for many years. However, the scarcity of TFIID and its highly flexible nature have made this pursuit very challenging. Recent breakthroughs, largely due to methodological advances in cryo-electron microscopy, have finally described the structure of this complex, both alone and engaged with core promoter DNA, revealing the functional significance of its conformational complexity in the process of core promoter recognition and initiation of Pol II transcription. Here, we review these recent structural insights and discuss their implications for our understanding of eukaryotic transcription initiation.


Subject(s)
Molecular Docking Simulation , Molecular Dynamics Simulation , Promoter Regions, Genetic , Protein Conformation , Transcription Factor TFIID/chemistry , Amino Acid Sequence , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Humans , Molecular Conformation , Protein Binding , Structure-Activity Relationship , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , Transcription Factor TFIID/metabolism
16.
Biochemistry ; 59(2): 183-196, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31478652

ABSTRACT

The metabolic serine hydrolase family is, arguably, one of the largest functional enzyme classes in mammals, including humans, comprising 1-2% of the total proteome. This enzyme family uses a conserved nucleophilic serine residue in the active site to perform diverse hydrolytic reactions and consists of proteases, lipases, esterases, amidases, and transacylases, which are prototypical members of this family. In humans, this enzyme family consists of >250, of which approximately 40% members remain unannotated, in terms of both their endogenous substrates and the biological pathways that they regulate. The enzyme ABHD14B, an outlying member of this family, is also known as CCG1/TAFII250-interacting factor B, as it was found to be associated with transcription initiation factor TFIID. The crystal structure of human ABHD14B was determined more than a decade ago; however, its endogenous substrates remain elusive. In this paper, we annotate ABHD14B as a lysine deacetylase (KDAC), showing this enzyme's ability to transfer an acetyl group from a post-translationally acetylated lysine to coenzyme A (CoA), to yield acetyl-CoA, while regenerating the free amine of protein lysine residues. We validate these findings by in vitro biochemical assays using recombinantly purified human ABHD14B in conjunction with cellular studies in a mammalian cell line by knocking down ABHD14B and by identification of a putative substrate binding site. Finally, we report the development and characterization of a much-needed, exquisitely selective ABHD14B antibody, and using it, we map the cellular and tissue distribution of ABHD14B and prospective metabolic pathways that this enzyme might biologically regulate.


Subject(s)
Acetyltransferases/metabolism , Histone Acetyltransferases/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Acetylation , Acetyltransferases/chemistry , Acetyltransferases/genetics , Animals , Catalytic Domain , Cell Line, Tumor , Coenzyme A/chemistry , Enzyme Assays , Escherichia coli/genetics , Gene Knockdown Techniques , HEK293 Cells , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Humans , Hydrolases , Mice, Inbred C57BL , Rabbits , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics
17.
Nat Struct Mol Biol ; 26(11): 1035-1043, 2019 11.
Article in English | MEDLINE | ID: mdl-31686052

ABSTRACT

Transcription factor c-MYC is a potent oncoprotein; however, the mechanism of transcriptional regulation via MYC-protein interactions remains poorly understood. The TATA-binding protein (TBP) is an essential component of the transcription initiation complex TFIID and is required for gene expression. We identify two discrete regions mediating MYC-TBP interactions using structural, biochemical and cellular approaches. A 2.4 -Å resolution crystal structure reveals that human MYC amino acids 98-111 interact with TBP in the presence of the amino-terminal domain 1 of TBP-associated factor 1 (TAF1TAND1). Using biochemical approaches, we have shown that MYC amino acids 115-124 also interact with TBP independently of TAF1TAND1. Modeling reveals that this region of MYC resembles a TBP anchor motif found in factors that regulate TBP promoter loading. Site-specific MYC mutants that abrogate MYC-TBP interaction compromise MYC activity. We propose that MYC-TBP interactions propagate transcription by modulating the energetic landscape of transcription initiation complex assembly.


Subject(s)
Protein Interaction Maps , Proto-Oncogene Proteins c-myc/metabolism , TATA-Box Binding Protein/metabolism , Cell Line, Tumor , Crystallography, X-Ray , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/metabolism , Humans , Models, Molecular , Protein Conformation , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-myc/chemistry , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , TATA-Box Binding Protein/chemistry , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/metabolism
18.
Nat Commun ; 10(1): 4925, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664040

ABSTRACT

AML1-ETO (AE) is a fusion transcription factor, generated by the t(8;21) translocation, that functions as a leukemia promoting oncogene. Here, we demonstrate that TATA-Box Binding Protein Associated Factor 1 (TAF1) associates with K43 acetylated AE and this association plays a pivotal role in the proliferation of AE-expressing acute myeloid leukemia (AML) cells. ChIP-sequencing indicates significant overlap of the TAF1 and AE binding sites. Knockdown of TAF1 alters the association of AE with chromatin, affecting of the expression of genes that are activated or repressed by AE. Furthermore, TAF1 is required for leukemic cell self-renewal and its reduction promotes the differentiation and apoptosis of AE+ AML cells, thereby impairing AE driven leukemogenesis. Together, our findings reveal a role of TAF1 in leukemogenesis and identify TAF1 as a potential therapeutic target for AE-expressing leukemia.


Subject(s)
Carcinogenesis/pathology , Core Binding Factor Alpha 2 Subunit/metabolism , Histone Acetyltransferases/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Oncogene Proteins, Fusion/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Acetylation , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Self Renewal , Chromatin/metabolism , Gene Expression Regulation, Leukemic , Histone Acetyltransferases/chemistry , Humans , Lysine/metabolism , Mice, Inbred C57BL , Myeloid Cells/pathology , Protein Binding , Protein Domains , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/chemistry
19.
Trends Cell Biol ; 29(9): 752-763, 2019 09.
Article in English | MEDLINE | ID: mdl-31300188

ABSTRACT

Basal transcription factor TFIID connects transcription activation to the assembly of the RNA polymerase II preinitiation complex at the core promoter of genes. The mechanistic understanding of TFIID function and dynamics has been limited by the lack of high-resolution structures of the holo-TFIID complex. Recent cryo-electron microscopy studies of yeast and human TFIID complexes provide insight into the molecular organization and structural dynamics of this highly conserved transcription factor. Here, we discuss how these TFIID structures provide new paradigms for: (i) the dynamic recruitment of TFIID; (ii) the binding of TATA-binding protein (TBP) to promoter DNA; (iii) the multivalency of TFIID interactions with (co)activators, nucleosomes, or promoter DNA; and (iv) the opportunities for regulation of TBP turnover and promoter dynamics.


Subject(s)
DNA/chemistry , Promoter Regions, Genetic , Protein Conformation , Transcription Factor TFIID/chemistry , Cryoelectron Microscopy , DNA/ultrastructure , Humans , Models, Molecular , Schizosaccharomyces/chemistry , TATA-Box Binding Protein/metabolism , Transcription Factor TFIID/ultrastructure , Transcriptional Activation
20.
Nat Commun ; 10(1): 1740, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988355

ABSTRACT

Cells dedicate significant energy to build proteins often organized in multiprotein assemblies with tightly regulated stoichiometries. As genes encoding subunits assembling in a multisubunit complex are dispersed in the genome of eukaryotes, it is unclear how these protein complexes assemble. Here, we show that mammalian nuclear transcription complexes (TFIID, TREX-2 and SAGA) composed of a large number of subunits, but lacking precise architectural details are built co-translationally. We demonstrate that dimerization domains and their positions in the interacting subunits determine the co-translational assembly pathway (simultaneous or sequential). The lack of co-translational interaction can lead to degradation of the partner protein. Thus, protein synthesis and complex assembly are linked in building mammalian multisubunit complexes, suggesting that co-translational assembly is a general principle in mammalian cells to avoid non-specific interactions and protein aggregation. These findings will also advance structural biology by defining endogenous co-translational building blocks in the architecture of multisubunit complexes.


Subject(s)
Protein Multimerization , Protein Subunits/metabolism , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , HeLa Cells , Humans , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Domains , Protein Folding , Protein Subunits/chemistry , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL