Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
1.
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
2.
Nature ; 577(7792): 717-720, 2020 01.
Article in English | MEDLINE | ID: mdl-31969703

ABSTRACT

Gene transcription by RNA polymerase II is regulated by activator proteins that recruit the coactivator complexes SAGA (Spt-Ada-Gcn5-acetyltransferase)1,2 and transcription factor IID (TFIID)2-4. SAGA is required for all regulated transcription5 and is conserved among eukaryotes6. SAGA contains four modules7-9: the activator-binding Tra1 module, the core module, the histone acetyltransferase (HAT) module and the histone deubiquitination (DUB) module. Previous studies provided partial structures10-14, but the structure of the central core module is unknown. Here we present the cryo-electron microscopy structure of SAGA from the yeast Saccharomyces cerevisiae and resolve the core module at 3.3 Å resolution. The core module consists of subunits Taf5, Sgf73 and Spt20, and a histone octamer-like fold. The octamer-like fold comprises the heterodimers Taf6-Taf9, Taf10-Spt7 and Taf12-Ada1, and two histone-fold domains in Spt3. Spt3 and the adjacent subunit Spt8 interact with the TATA box-binding protein (TBP)2,7,15-17. The octamer-like fold and its TBP-interacting region are similar in TFIID, whereas Taf5 and the Taf6 HEAT domain adopt distinct conformations. Taf12 and Spt20 form flexible connections to the Tra1 module, whereas Sgf73 tethers the DUB module. Binding of a nucleosome to SAGA displaces the HAT and DUB modules from the core-module surface, allowing the DUB module to bind one face of an ubiquitinated nucleosome.


Subject(s)
Cryoelectron Microscopy , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/ultrastructure , Saccharomyces cerevisiae , Trans-Activators/chemistry , Trans-Activators/ultrastructure , Transcription, Genetic , Gene Expression Regulation, Fungal , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/ultrastructure , Histones/metabolism , Models, Molecular , Nucleosomes/chemistry , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Protein Binding , Protein Domains , Protein Subunits/chemistry , Protein Subunits/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/metabolism , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , Trans-Activators/metabolism , Transcription Factor TFIID/metabolism , Ubiquitination
3.
Mol Cell ; 64(4): 815-825, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27840029

ABSTRACT

The five-subunit yeast Paf1 complex (Paf1C) regulates all stages of transcription and is critical for the monoubiquitylation of histone H2B (H2Bub), a modification that broadly influences chromatin structure and eukaryotic transcription. Here, we show that the histone modification domain (HMD) of Paf1C subunit Rtf1 directly interacts with the ubiquitin conjugase Rad6 and stimulates H2Bub independently of transcription. We present the crystal structure of the Rtf1 HMD and use site-specific, in vivo crosslinking to identify a conserved Rad6 interaction surface. Utilizing ChIP-exo analysis, we define the localization patterns of the H2Bub machinery at high resolution and demonstrate the importance of Paf1C in targeting the Rtf1 HMD, and thereby H2Bub, to its appropriate genomic locations. Finally, we observe HMD-dependent stimulation of H2Bub in a transcription-free, reconstituted in vitro system. Taken together, our results argue for an active role for Paf1C in promoting H2Bub and ensuring its proper localization in vivo.


Subject(s)
Gene Expression Regulation, Fungal , Histones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , TATA-Box Binding Protein/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Amino Acid Motifs , Binding Sites , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cross-Linking Reagents/chemistry , Crystallography, X-Ray , Formaldehyde/chemistry , Histones/chemistry , Histones/genetics , Models, Molecular , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitination
4.
Mol Cell ; 59(5): 755-67, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26257282

ABSTRACT

Genome/chromosome organization is highly ordered and controls various nuclear events, although the molecular mechanisms underlying the functional organization remain largely unknown. Here, we show that the TATA box-binding protein (TBP) interacts with the Cnd2 kleisin subunit of condensin to mediate interphase and mitotic chromosomal organization in fission yeast. TBP recruits condensin onto RNA polymerase III-transcribed (Pol III) genes and highly transcribed Pol II genes; condensin in turn associates these genes with centromeres. Inhibition of the Cnd2-TBP interaction disrupts condensin localization across the genome and the proper assembly of mitotic chromosomes, leading to severe defects in chromosome segregation and eventually causing cellular lethality. We propose that the Cnd2-TBP interaction coordinates transcription with chromosomal architecture by linking dispersed gene loci with centromeres. This chromosome arrangement can contribute to the efficient transmission of physical force at the kinetochore to chromosomal arms, thereby supporting the fidelity of chromosome segregation.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/chemistry , Centromere/genetics , Centromere/metabolism , Chromosome Segregation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, Fungal , Mitosis , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Point Mutation , Protein Interaction Domains and Motifs , Protein Subunits , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , Schizosaccharomyces/cytology , Schizosaccharomyces pombe Proteins/chemistry , TATA-Box Binding Protein/chemistry
5.
Nature ; 531(7596): 604-9, 2016 03 31.
Article in English | MEDLINE | ID: mdl-27007846

ABSTRACT

The general transcription factor IID (TFIID) plays a central role in the initiation of RNA polymerase II (Pol II)-dependent transcription by nucleating pre-initiation complex (PIC) assembly at the core promoter. TFIID comprises the TATA-binding protein (TBP) and 13 TBP-associated factors (TAF1-13), which specifically interact with a variety of core promoter DNA sequences. Here we present the structure of human TFIID in complex with TFIIA and core promoter DNA, determined by single-particle cryo-electron microscopy at sub-nanometre resolution. All core promoter elements are contacted by subunits of TFIID, with TAF1 and TAF2 mediating major interactions with the downstream promoter. TFIIA bridges the TBP-TATA complex with lobe B of TFIID. We also present the cryo-electron microscopy reconstruction of a fully assembled human TAF-less PIC. Superposition of common elements between the two structures provides novel insights into the general role of TFIID in promoter recognition, PIC assembly, and transcription initiation.


Subject(s)
Promoter Regions, Genetic/genetics , Transcription Factor TFIID/metabolism , Transcription Factor TFIID/ultrastructure , Transcription Initiation, Genetic , Cryoelectron Microscopy , DNA/chemistry , DNA/metabolism , DNA/ultrastructure , Humans , Models, Molecular , Protein Binding , Substrate Specificity , TATA Box/genetics , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , TATA-Binding Protein Associated Factors/ultrastructure , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , TATA-Box Binding Protein/ultrastructure , Transcription Factor TFIIA/chemistry , Transcription Factor TFIIA/metabolism , Transcription Factor TFIIA/ultrastructure , Transcription Factor TFIID/chemistry
6.
Nature ; 533(7603): 353-8, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27193681

ABSTRACT

Transcription of eukaryotic protein-coding genes begins with assembly of the RNA polymerase (Pol) II initiation complex and promoter DNA opening. Here we report cryo-electron microscopy (cryo-EM) structures of yeast initiation complexes containing closed and open DNA at resolutions of 8.8 Å and 3.6 Å, respectively. DNA is positioned and retained over the Pol II cleft by a network of interactions between the TATA-box-binding protein TBP and transcription factors TFIIA, TFIIB, TFIIE, and TFIIF. DNA opening occurs around the tip of the Pol II clamp and the TFIIE 'extended winged helix' domain, and can occur in the absence of TFIIH. Loading of the DNA template strand into the active centre may be facilitated by movements of obstructing protein elements triggered by allosteric binding of the TFIIE 'E-ribbon' domain. The results suggest a unified model for transcription initiation with a key event, the trapping of open promoter DNA by extended protein-protein and protein-DNA contacts.


Subject(s)
DNA/metabolism , DNA/ultrastructure , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Nucleic Acid Conformation , Promoter Regions, Genetic , Transcription Initiation, Genetic , Allosteric Site , Base Sequence , Cryoelectron Microscopy , DNA/chemistry , Models, Biological , Molecular Sequence Data , Movement , Multiprotein Complexes/metabolism , Protein Binding , Protein Structure, Tertiary , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , RNA Polymerase II/ultrastructure , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , TATA-Box Binding Protein/ultrastructure , Templates, Genetic , Transcription Factors, TFII/chemistry , Transcription Factors, TFII/metabolism , Transcription Factors, TFII/ultrastructure
7.
J Biol Chem ; 295(49): 16470-16486, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33051202

ABSTRACT

RNA-protein interfaces control key replication events during the HIV-1 life cycle. The viral trans-activator of transcription (Tat) protein uses an archetypal arginine-rich motif (ARM) to recruit the host positive transcription elongation factor b (pTEFb) complex onto the viral trans-activation response (TAR) RNA, leading to activation of HIV transcription. Efforts to block this interaction have stimulated production of biologics designed to disrupt this essential RNA-protein interface. Here, we present four co-crystal structures of lab-evolved TAR-binding proteins (TBPs) in complex with HIV-1 TAR. Our results reveal that high-affinity binding requires a distinct sequence and spacing of arginines within a specific ß2-ß3 hairpin loop that arose during selection. Although loops with as many as five arginines were analyzed, only three arginines could bind simultaneously with major-groove guanines. Amino acids that promote backbone interactions within the ß2-ß3 loop were also observed to be important for high-affinity interactions. Based on structural and affinity analyses, we designed two cyclic peptide mimics of the TAR-binding ß2-ß3 loop sequences present in two high-affinity TBPs (KD values of 4.2 ± 0.3 and 3.0 ± 0.3 nm). Our efforts yielded low-molecular weight compounds that bind TAR with low micromolar affinity (KD values ranging from 3.6 to 22 µm). Significantly, one cyclic compound within this series blocked binding of the Tat-ARM peptide to TAR in solution assays, whereas its linear counterpart did not. Overall, this work provides insight into protein-mediated TAR recognition and lays the ground for the development of cyclic peptide inhibitors of a vital HIV-1 RNA-protein interaction.


Subject(s)
Arginine/chemistry , HIV Long Terminal Repeat/genetics , HIV-1/metabolism , Peptides, Cyclic/chemistry , RNA, Viral/metabolism , TATA-Box Binding Protein/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Drug Design , Humans , Kinetics , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Nucleic Acid Conformation , Peptides, Cyclic/metabolism , Protein Binding , RNA, Viral/chemistry , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Thermodynamics
8.
Nucleic Acids Res ; 47(6): 2793-2806, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30649478

ABSTRACT

The TATA-box Binding Protein (TBP) plays a central role in regulating gene expression and is the first step in the process of pre-initiation complex (PIC) formation on promoter DNA. The lifetime of TBP at the promoter site is controlled by several cofactors including the Modifier of transcription 1 (Mot1), an essential TBP-associated ATPase. Based on ensemble measurements, Mot1 can use adenosine triphosphate (ATP) hydrolysis to displace TBP from DNA and various models for how this activity is coupled to transcriptional regulation have been proposed. However, the underlying molecular mechanism of Mot1 action is not well understood. In this work, the interaction of Mot1 with the DNA/TBP complex was investigated by single-pair Förster resonance energy transfer (spFRET). Upon Mot1 binding to the DNA/TBP complex, a transition in the DNA/TBP conformation was observed. Hydrolysis of ATP by Mot1 led to a conformational change but was not sufficient to efficiently disrupt the complex. SpFRET measurements of dual-labeled DNA suggest that Mot1's ATPase activity primes incorrectly oriented TBP for dissociation from DNA and additional Mot1 in solution is necessary for TBP unbinding. These findings provide a framework for understanding how the efficiency of Mot1's catalytic activity is tuned to establish a dynamic pool of TBP without interfering with stable and functional TBP-containing complexes.


Subject(s)
Adenosine Triphosphatases/physiology , DNA, Fungal/metabolism , Saccharomyces cerevisiae Proteins/physiology , TATA-Binding Protein Associated Factors/physiology , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Catalysis , DNA, Fungal/chemistry , Escherichia coli , Gene Expression Regulation, Fungal , Models, Molecular , Nucleic Acid Conformation , Promoter Regions, Genetic , Protein Binding , Protein Conformation , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism
9.
Nucleic Acids Res ; 47(16): 8410-8423, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31226204

ABSTRACT

The nucleosome core regulates DNA-templated processes through the highly conserved nucleosome acidic patch. While structural and biochemical studies have shown that the acidic patch controls chromatin factor binding and activity, few studies have elucidated its functions in vivo. We employed site-specific crosslinking to identify proteins that directly bind the acidic patch in Saccharomyces cerevisiae and demonstrated crosslinking of histone H2A to Paf1 complex subunit Rtf1 and FACT subunit Spt16. Rtf1 bound to nucleosomes through its histone modification domain, supporting its role as a cofactor in H2B K123 ubiquitylation. An acidic patch mutant showed defects in nucleosome positioning and occupancy genome-wide. Our results provide new information on the chromatin engagement of two central players in transcription elongation and emphasize the importance of the nucleosome core as a hub for proteins that regulate chromatin during transcription.


Subject(s)
DNA, Fungal/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Fungal , Genome, Fungal , High Mobility Group Proteins/genetics , Nuclear Proteins/genetics , Nucleosomes/ultrastructure , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Transcriptional Elongation Factors/genetics , Binding Sites , DNA, Fungal/chemistry , DNA, Fungal/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/chemistry , High Mobility Group Proteins/metabolism , Histones/chemistry , Histones/genetics , Histones/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Nucleosomes/metabolism , Protein Binding , Protein Conformation , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Transcription, Genetic , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/metabolism , Ubiquitination
10.
J Chem Inf Model ; 60(2): 866-879, 2020 02 24.
Article in English | MEDLINE | ID: mdl-31917925

ABSTRACT

The TATA-box binding protein (TBP) is an important element of the transcription machinery in archaea and eukaryotic organisms. TBP is expressed in organisms adapted to different temperatures, indicating a robust structure, and experimental studies have shown that the mid-unfolding temperature (Tm) of TBP is directly correlated with the optimal growth temperature (OGT) of the organism. To understand which are the relevant structural requirements for its stability, we present the first structural and dynamic computational study of TBPs, combining molecular dynamics (MD) simulations and a quantitative structure-property relationship (QSPR) over a set of TBPs of organisms adapted to different temperatures. We found that the main structural properties of TBP used to adapt to high temperatures are an increase in the ease of desolvation of charged residues at the surface, an increase in the local resiliency, the presence of Leu clusters in the protein core, and an increase in the loss of hydrophobic packing in the N-terminal subdomain. In view of our results, we consider that TBP is a good model to study thermal adaptation, and our analysis opens the possibility of performing protein engineering on TBPs to study transcription at high or low temperatures.


Subject(s)
Molecular Dynamics Simulation , Quantitative Structure-Activity Relationship , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , Temperature , Adaptation, Physiological , Humans , Hydrophobic and Hydrophilic Interactions , Protein Conformation , Sulfides/chemistry
11.
Mol Cell ; 45(4): 439-46, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22365827

ABSTRACT

Recent studies of the three eukaryotic transcription machineries revealed that all initiation complexes share a conserved core. This core consists of the RNA polymerase (I, II, or III), the TATA box-binding protein (TBP), and transcription factors TFIIB, TFIIE, and TFIIF (for Pol II) or proteins structurally and functionally related to parts of these factors (for Pol I and Pol III). The conserved core initiation complex stabilizes the open DNA promoter complex and directs initial RNA synthesis. The periphery of the core initiation complex is decorated by additional polymerase-specific factors that account for functional differences in promoter recognition and opening, and gene class-specific regulation. This review outlines the similarities and differences between these important molecular machines.


Subject(s)
RNA Polymerase III/metabolism , RNA Polymerase II/metabolism , RNA Polymerase I/metabolism , Transcription, Genetic , Conserved Sequence , Models, Molecular , Protein Structure, Quaternary , Protein Structure, Tertiary , RNA Polymerase I/chemistry , RNA Polymerase I/physiology , RNA Polymerase II/chemistry , RNA Polymerase II/physiology , RNA Polymerase III/chemistry , RNA Polymerase III/physiology , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/physiology , Transcription Factor TFIIB/chemistry , Transcription Factor TFIIB/physiology , Transcription Factors, TFII/chemistry , Transcription Factors, TFII/physiology
12.
Biochem Soc Trans ; 47(1): 411-423, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30710057

ABSTRACT

In all domains of life, the regulation of transcription by DNA-dependent RNA polymerases (RNAPs) is achieved at the level of initiation to a large extent. Whereas bacterial promoters are recognized by a σ-factor bound to the RNAP, a complex set of transcription factors that recognize specific promoter elements is employed by archaeal and eukaryotic RNAPs. These initiation factors are of particular interest since the regulation of transcription critically relies on initiation rates and thus formation of pre-initiation complexes. The most conserved initiation factor is the TATA-binding protein (TBP), which is of crucial importance for all archaeal-eukaryotic transcription initiation complexes and the only factor required to achieve full rates of initiation in all three eukaryotic and the archaeal transcription systems. Recent structural, biochemical and genome-wide mapping data that focused on the archaeal and specialized RNAP I and III transcription system showed that the involvement and functional importance of TBP is divergent from the canonical role TBP plays in RNAP II transcription. Here, we review the role of TBP in the different transcription systems including a TBP-centric discussion of archaeal and eukaryotic initiation complexes. We furthermore highlight questions concerning the function of TBP that arise from these findings.


Subject(s)
TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , Transcription Initiation, Genetic , Archaeal Proteins , DNA-Directed RNA Polymerases/metabolism , Evolution, Molecular , Protein Conformation
13.
Nature ; 495(7442): 481-6, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23446344

ABSTRACT

Eukaryotic transcription initiation requires the assembly of general transcription factors into a pre-initiation complex that ensures the accurate loading of RNA polymerase II (Pol II) at the transcription start site. The molecular mechanism and function of this assembly have remained elusive due to lack of structural information. Here we have used an in vitro reconstituted system to study the stepwise assembly of human TBP, TFIIA, TFIIB, Pol II, TFIIF, TFIIE and TFIIH onto promoter DNA using cryo-electron microscopy. Our structural analyses provide pseudo-atomic models at various stages of transcription initiation that illuminate critical molecular interactions, including how TFIIF engages Pol II and promoter DNA to stabilize both the closed pre-initiation complex and the open-promoter complex, and to regulate start--initiation complexes, combined with the localization of the TFIIH helicases XPD and XPB, support a DNA translocation model of XPB and explain its essential role in promoter opening.


Subject(s)
Promoter Regions, Genetic/genetics , RNA Polymerase II/chemistry , RNA Polymerase II/ultrastructure , Transcription Factors, TFII/chemistry , Transcription Factors, TFII/ultrastructure , Transcription Initiation, Genetic/physiology , Base Sequence , Cryoelectron Microscopy , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Helicases/chemistry , DNA Helicases/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , RNA Polymerase II/metabolism , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , Transcription Factor TFIIH/chemistry , Transcription Factor TFIIH/metabolism , Transcription Factors, TFII/metabolism , Transcription Initiation Site
14.
Parasitol Res ; 118(10): 3019-3031, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31473857

ABSTRACT

The protozoan parasite Trichomonas vaginalis is a common human pathogen from one of the earliest-diverging eukaryotic lineages. At the transcriptional level, the highly conserved Inr element of RNA pol II-transcribed genes surrounds the transcription start site and is recognised by IBP39, a protein exclusive of T. vaginalis. Typical TATA boxes have not been identified in this organism but, in contrast, BLAST analyses of the T. vaginalis genome identified two genes encoding putative TATA-binding proteins (herein referred to as TvTBP1 and TvTBP2). The goal of this work was to characterise these two proteins at the molecular level. Our results show that both TvTBPs theoretically adopt the saddle-shaped structure distinctive to TBPs and both Tvtbp genes are expressed in T. vaginalis. TvTBP1 did not complement a Saccharomyces cerevisiae mutant lacking TBP; however, TvTBP1 and TvTBP2 proteins bound T. vaginalis DNA promoter sequences in EMSA assays. We propose that TvTBP1 may be part of the preinitiation transcription complex in T. vaginalis since TvTBP1 recombinant protein was able to bind IBP39 in vitro. This work represents the first approach towards the characterisation of general transcription factors in this early divergent organism.


Subject(s)
DNA-Binding Proteins/metabolism , Protozoan Proteins/metabolism , TATA-Box Binding Protein/metabolism , Trichomonas vaginalis/metabolism , Models, Molecular , Promoter Regions, Genetic , Protein Binding , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/genetics , Transcription, Genetic , Trichomonas vaginalis/genetics
15.
Angew Chem Int Ed Engl ; 58(35): 12010-12013, 2019 08 26.
Article in English | MEDLINE | ID: mdl-31268220

ABSTRACT

Hoogsteen DNA base pairs (bps) are an alternative base pairing to canonical Watson-Crick bps and are thought to play important biochemical roles. Hoogsteen bps have been reported in a handful of X-ray structures of protein-DNA complexes. However, there are several examples of Hoogsteen bps in crystal structures that form Watson-Crick bps when examined under solution conditions. Furthermore, Hoogsteen bps can sometimes be difficult to resolve in DNA:protein complexes by X-ray crystallography due to ambiguous electron density and by solution-state NMR spectroscopy due to size limitations. Here, using infrared spectroscopy, we report the first direct solution-state observation of a Hoogsteen (G-C+ ) bp in a DNA:protein complex under solution conditions with specific application to DNA-bound TATA-box binding protein. These results support a previous assignment of a G-C+ Hoogsteen bp in the complex, and indicate that Hoogsteen bps do indeed exist under solution conditions in DNA:protein complexes.


Subject(s)
Cytosine/chemistry , DNA/metabolism , Guanine/chemistry , TATA-Box Binding Protein/metabolism , Base Pairing , Crystallography, X-Ray , DNA/chemistry , Nucleic Acid Conformation , Spectrophotometry, Infrared , TATA-Box Binding Protein/chemistry
16.
Biophys J ; 115(12): 2310-2326, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30527334

ABSTRACT

Single-pair Förster resonance energy transfer (spFRET) has become an important tool for investigating conformational dynamics in biological systems. To extract dynamic information from the spFRET traces measured with total internal reflection fluorescence microscopy, we extended the hidden Markov model (HMM) approach. In our extended HMM analysis, we incorporated the photon-shot noise from camera-based systems into the HMM. Thus, the variance in Förster resonance energy transfer (FRET) efficiency of the various states, which is typically a fitted parameter, is explicitly included in the analysis estimated from the number of detected photons. It is also possible to include an additional broadening of the FRET state, which would then only reflect the inherent flexibility of the dynamic biological systems. This approach is useful when comparing the dynamics of individual molecules for which the total intensities vary significantly. We used spFRET with the extended HMM analysis to investigate the dynamics of TATA-box-binding protein (TBP) on promoter DNA in the presence of negative cofactor 2 (NC2). We compared the dynamics of two promoters as well as DNAs of different length and labeling location. For the adenovirus major late promoter, four FRET states were observed; three states correspond to different conformations of the DNA in the TBP-DNA-NC2 complex and a four-state model in which the complex has shifted along the DNA. The HMM analysis revealed that the states are connected via a linear, four-well model. For the H2B promoter, more complex dynamics were observed. By clustering the FRET states detected with the HMM analysis, we could compare the general dynamics observed for the two promoter sequences. We observed that the dynamics from a stretched DNA conformation to a bent conformation for the two promoters were similar, whereas the bent conformation of the TBP-DNA-NC2 complex for the H2B promoter is approximately three times more stable than for the adenovirus major late promoter.


Subject(s)
DNA/metabolism , Fluorescence Resonance Energy Transfer , Markov Chains , TATA-Box Binding Protein/metabolism , Transcription Factors/metabolism , DNA/chemistry , Models, Molecular , Nucleic Acid Conformation , Protein Conformation , TATA-Box Binding Protein/chemistry , Transcription Factors/chemistry
17.
J Biol Chem ; 292(28): 11873-11885, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28539359

ABSTRACT

RNA polymerase II (pol II) is required for the transcription of all protein-coding genes and as such represents a major enzyme whose activity is tightly regulated. Transcriptional initiation therefore requires numerous general transcriptional factors and cofactors that associate with pol II at the core promoter to form a pre-initiation complex. Transcription factor IIA (TFIIA) is a general cofactor that binds TFIID and stabilizes the TFIID-DNA complex during transcription initiation. Previous studies showed that TFIIA can make contact with the DNA sequence upstream or downstream of the TATA box, and that the region bound by TFIIA could overlap with the elements recognized by another factor, TFIIB, at adenovirus major late core promoter. Whether core promoters contain a DNA motif recognized by TFIIA remains unknown. Here we have identified a core promoter element upstream of the TATA box that is recognized by TFIIA. A search of the human promoter database revealed that many natural promoters contain a TFIIA recognition element (IIARE). We show that the IIARE enhances TFIIA-promoter binding and enhances the activity of TATA-containing promoters, but represses or activates promoters that lack a TATA box. Chromatin immunoprecipitation assays revealed that the IIARE activates transcription by increasing the recruitment of pol II, TFIIA, TAF4, and P300 at TATA-dependent promoters. These findings extend our understanding of the role of TFIIA in transcription, and provide new insights into the regulatory mechanism of core promoter elements in gene transcription by pol II.


Subject(s)
Gene Expression Regulation , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Response Elements , TATA Box , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIIA/metabolism , Transcription Factor TFIID/metabolism , Binding Sites , Chromatin Immunoprecipitation , DNA, Recombinant , E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/metabolism , Genes, Reporter , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Mutation , Nucleotide Motifs , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Polymerase II/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , TATA-Binding Protein Associated Factors/chemistry , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Transcription Factor TFIIA/chemistry , Transcription Factor TFIIA/genetics , Transcription Factor TFIID/chemistry , Upstream Stimulatory Factors/chemistry , Upstream Stimulatory Factors/genetics , Upstream Stimulatory Factors/metabolism
18.
Nucleic Acids Res ; 44(20): 9990-10002, 2016 Nov 16.
Article in English | MEDLINE | ID: mdl-27658967

ABSTRACT

We have studied the dynamics of three transcription factor-DNA complexes using all-atom, microsecond-scale MD simulations. In each case, the salt bridges and hydrogen bond interactions formed at the protein-DNA interface are found to be dynamic, with lifetimes typically in the range of tens to hundreds of picoseconds, although some interactions, notably those involving specific binding to DNA bases, can be a hundred times longer lived. Depending on the complex studied, this dynamics may or may not lead to the existence of distinct conformational substates. Using a sequence threading technique, it has been possible to determine whether DNA sequence recognition is sensitive or not to such conformational changes, and, in one case, to show that recognition appears to be locally dependent on protein-mediated cation distributions.


Subject(s)
DNA-Binding Proteins/chemistry , DNA/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Base Sequence , Binding Sites , DNA/metabolism , DNA-Binding Proteins/metabolism , Humans , Hydrogen Bonding , Nucleic Acid Conformation , Nucleotide Motifs , Position-Specific Scoring Matrices , Protein Binding , Protein Conformation , SOXB1 Transcription Factors/chemistry , SOXB1 Transcription Factors/metabolism , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
19.
Nature ; 475(7356): 403-7, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21734658

ABSTRACT

Swi2/Snf2-type ATPases regulate genome-associated processes such as transcription, replication and repair by catalysing the disruption, assembly or remodelling of nucleosomes or other protein-DNA complexes. It has been suggested that ATP-driven motor activity along DNA disrupts target protein-DNA interactions in the remodelling reaction. However, the complex and highly specific remodelling reactions are poorly understood, mostly because of a lack of high-resolution structural information about how remodellers bind to their substrate proteins. Mot1 (modifier of transcription 1 in Saccharomyces cerevisiae, denoted BTAF1 in humans) is a Swi2/Snf2 enzyme that specifically displaces the TATA box binding protein (TBP) from the promoter DNA and regulates transcription globally by generating a highly dynamic TBP pool in the cell. As a Swi2/Snf2 enzyme that functions as a single polypeptide and interacts with a relatively simple substrate, Mot1 offers an ideal system from which to gain a better understanding of this important enzyme family. To reveal how Mot1 specifically disrupts TBP-DNA complexes, we combined crystal and electron microscopy structures of Mot1-TBP from Encephalitozoon cuniculi with biochemical studies. Here we show that Mot1 wraps around TBP and seems to act like a bottle opener: a spring-like array of 16 HEAT (huntingtin, elongation factor 3, protein phosphatase 2A and lipid kinase TOR) repeats grips the DNA-distal side of TBP via loop insertions, and the Swi2/Snf2 domain binds to upstream DNA, positioned to weaken the TBP-DNA interaction by DNA translocation. A 'latch' subsequently blocks the DNA-binding groove of TBP, acting as a chaperone to prevent DNA re-association and ensure efficient promoter clearance. This work shows how a remodelling enzyme can combine both motor and chaperone activities to achieve functional specificity using a conserved Swi2/Snf2 translocase.


Subject(s)
Encephalitozoon cuniculi/chemistry , Fungal Proteins/chemistry , Fungal Proteins/metabolism , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism , Binding Sites , Crystallization , Crystallography, X-Ray , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA/ultrastructure , Fungal Proteins/ultrastructure , Microscopy, Electron , Models, Biological , Models, Molecular , Promoter Regions, Genetic/genetics , Protein Conformation , Protein Structure, Tertiary , Structure-Activity Relationship , Substrate Specificity , TATA-Box Binding Protein/ultrastructure , Transcription Factor TFIIB/chemistry , Transcription Factor TFIIB/metabolism
20.
Hum Mol Genet ; 23(25): 6878-93, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25104854

ABSTRACT

Spinocerebellar ataxia type 17 (SCA17) is an autosomal dominant cerebellar ataxia caused by the expansion of polyglutamine (polyQ) within the TATA box-binding protein (TBP). Previous studies have shown that polyQ-expanded TBP forms neurotoxic aggregates and alters downstream genes. However, how expanded polyQ tracts affect the function of TBP and the link between dysfunctional TBP and SCA17 is not clearly understood. In this study, we generated novel Drosophila models for SCA17 that recapitulate pathological features such as aggregate formation, mobility defects and premature death. In addition to forming neurotoxic aggregates, we determined that polyQ-expanded TBP reduces its own intrinsic DNA-binding and transcription abilities. Dysfunctional TBP also disrupts normal TBP function. Furthermore, heterozygous dTbp amorph mutant flies exhibited SCA17-like phenotypes and flies expressing polyQ-expanded TBP exhibited enhanced retinal degeneration, suggesting that loss of TBP function may contribute to SCA17 pathogenesis. We further determined that the downregulation of TBP activity enhances retinal degeneration in SCA3 and Huntington's disease fly models, indicating that the deactivation of TBP is likely to play a common role in polyQ-induced neurodegeneration.


Subject(s)
Drosophila melanogaster/genetics , Huntington Disease/genetics , Machado-Joseph Disease/genetics , Retinal Degeneration/genetics , Spinocerebellar Ataxias/genetics , TATA-Box Binding Protein/genetics , Animals , Disease Models, Animal , Drosophila melanogaster/metabolism , Gene Expression Regulation , Heterozygote , Humans , Huntington Disease/metabolism , Huntington Disease/pathology , Longevity/genetics , Machado-Joseph Disease/metabolism , Machado-Joseph Disease/pathology , Peptides/chemistry , Phenotype , Protein Aggregates , Protein Binding , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Signal Transduction , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology , TATA-Box Binding Protein/chemistry , TATA-Box Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL