Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 8128, 2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38065958

ABSTRACT

Acetylation of histones is a key post-translational modification that guides gene expression regulation. In yeast, the class I histone deacetylase containing Rpd3S complex plays a critical role in the suppression of spurious transcription by removing histone acetylation from actively transcribed genes. The S. cerevisiae Rpd3S complex has five subunits (Rpd3, Sin3, Rco1, Eaf3, and Ume1) but its subunit stoichiometry and how the complex engages nucleosomes to achieve substrate specificity remains elusive. Here we report the cryo-EM structure of the complete Rpd3S complex bound to a nucleosome. Sin3 and two copies of subunits Rco1 and Eaf3 encircle the deacetylase subunit Rpd3 and coordinate the positioning of Ume1. The Rpd3S complex binds both trimethylated H3 tails at position lysine 36 and makes multiple additional contacts with the nucleosomal DNA and the H2A-H2B acidic patch. Direct regulation via the Sin3 subunit coordinates binding of the acetylated histone substrate to achieve substrate specificity.


Subject(s)
Nucleosomes , Saccharomyces cerevisiae Proteins , Nucleosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Histones/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Methylation , Acetylation , Acetyltransferases/metabolism
2.
Curr Opin Struct Biol ; 82: 102690, 2023 10.
Article in English | MEDLINE | ID: mdl-37633188

ABSTRACT

Transcription of most protein-coding genes requires the passage of RNA polymerase II through chromatin. Chromatin with its fundamental unit, the nucleosome, represents a barrier to transcription. How RNA polymerase II and associated factors traverse through nucleosomes and how chromatin architecture is maintained have remained largely enigmatic. Only recently, cryo-EM structures have visualized the transcription process through chromatin. These structures have elucidated how transcription initiation and transcription elongation influence and are influenced by a chromatinized DNA substrate. This review provides a summary of our current structural understanding of transcription through chromatin, highlighting common mechanisms during nucleosomal traversal and novel regulatory mechanisms that have emerged in the last five years.


Subject(s)
Chromatin , Nucleosomes , RNA Polymerase II
3.
bioRxiv ; 2023 Aug 03.
Article in English | MEDLINE | ID: mdl-37577459

ABSTRACT

Acetylation of histones is a key post-translational modification that guides gene expression regulation. In yeast, the class I histone deacetylase containing Rpd3S complex plays a critical role in the suppression of spurious transcription by removing histone acetylation from actively transcribed genes. The Saccharomyces cerevisiae Rpd3S complex has five subunits (Rpd3, Sin3, Rco1, Eaf3, and Ume1) but its subunit stoichiometry and how the complex engages nucleosomes to achieve substrate specificity remains elusive. Here we report the cryo-EM structure of the complete Rpd3S complex bound to a nucleosome. Sin3 and two copies of subunits Rco1 and Eaf3 encircle the deacetylase subunit Rpd3 and coordinate the binding of Ume1. The Rpd3S complex binds both trimethylated H3 tails at position lysine 36 and makes multiple additional contacts with the nucleo-somal DNA, the H2A-H2B acidic patch, and histone H3. Direct regulation via the Sin3 subunit coordinates binding of the acetylated histone substrate to achieve substrate specificity.

4.
Mol Cell ; 83(14): 2464-2477.e5, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37369200

ABSTRACT

Co-transcriptional capping of the nascent pre-mRNA 5' end prevents degradation of RNA polymerase (Pol) II transcripts and suppresses the innate immune response. Here, we provide mechanistic insights into the three major steps of human co-transcriptional pre-mRNA capping based on six different cryoelectron microscopy (cryo-EM) structures. The human mRNA capping enzyme, RNGTT, first docks to the Pol II stalk to position its triphosphatase domain near the RNA exit site. The capping enzyme then moves onto the Pol II surface, and its guanylyltransferase receives the pre-mRNA 5'-diphosphate end. Addition of a GMP moiety can occur when the RNA is ∼22 nt long, sufficient to reach the active site of the guanylyltransferase. For subsequent cap(1) methylation, the methyltransferase CMTR1 binds the Pol II stalk and can receive RNA after it is grown to ∼29 nt in length. The observed rearrangements of capping factors on the Pol II surface may be triggered by the completion of catalytic reaction steps and are accommodated by domain movements in the elongation factor DRB sensitivity-inducing factor (DSIF).


Subject(s)
RNA Processing, Post-Transcriptional , RNA, Messenger , Humans , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Messenger/ultrastructure , Cryoelectron Microscopy , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , RNA Polymerase II/ultrastructure , Transcription, Genetic , Methyltransferases/chemistry , Methyltransferases/metabolism , Methyltransferases/ultrastructure , Models, Chemical
5.
J Am Chem Soc ; 145(12): 6811-6822, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36930461

ABSTRACT

The reversible acetylation of histone lysine residues is controlled by the action of acetyltransferases and deacetylases (HDACs), which regulate chromatin structure and gene expression. The sirtuins are a family of NAD-dependent HDAC enzymes, and one member, sirtuin 6 (Sirt6), influences DNA repair, transcription, and aging. Here, we demonstrate that Sirt6 is efficient at deacetylating several histone H3 acetylation sites, including its canonical site Lys9, in the context of nucleosomes but not free acetylated histone H3 protein substrates. By installing a chemical warhead at the Lys9 position of histone H3, we trap a catalytically poised Sirt6 in complex with a nucleosome and employ this in cryo-EM structural analysis. The structure of Sirt6 bound to a nucleosome reveals extensive interactions between distinct segments of Sirt6 and the H2A/H2B acidic patch and nucleosomal DNA, which accounts for the rapid deacetylation of nucleosomal H3 sites and the disfavoring of histone H2B acetylation sites. These findings provide a new framework for understanding how HDACs target and regulate chromatin.


Subject(s)
Nucleosomes , Sirtuins , Histones/chemistry , Chromatin , Sirtuins/metabolism , Acetylation , Glycosyltransferases/metabolism , Catalysis
6.
Mol Cell ; 82(17): 3126-3134.e7, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35858621

ABSTRACT

During gene transcription, RNA polymerase II (RNA Pol II) passes nucleosomes with the help of various elongation factors. Here, we show that RNA Pol II achieves efficient nucleosome passage when the human elongation factors DSIF, PAF1 complex (PAF), RTF1, SPT6, and TFIIS are present. The cryo-EM structure of an intermediate of the nucleosome passage shows a partially unraveled hexasome that lacks the proximal H2A-H2B dimer and interacts with the RNA Pol II jaw, DSIF, and the CTR9trestle helix. RNA Pol II adopts a backtracked state with the RNA 3' end dislodged from the active site and bound in the RNA Pol II pore. Additional structures and biochemical data show that human TFIIS enters the RNA Pol II pore and stimulates the cleavage of the backtracked RNA and nucleosome passage.


Subject(s)
Nucleosomes , RNA Polymerase II , Cell Nucleus/metabolism , Humans , Nucleosomes/genetics , RNA , RNA Polymerase II/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Elongation Factors/metabolism
7.
Science ; 376(6599): 1313-1316, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35709268

ABSTRACT

In eukaryotes, RNA polymerase (Pol) II transcribes chromatin and must move past nucleosomes, often resulting in nucleosome displacement. How Pol II unwraps the DNA from nucleosomes to allow transcription and how DNA rewraps to retain nucleosomes has been unclear. Here, we report the 3.0-angstrom cryo-electron microscopy structure of a mammalian Pol II-DSIF-SPT6-PAF1c-TFIIS-nucleosome complex stalled 54 base pairs within the nucleosome. The structure provides a mechanistic basis for nucleosome retention during transcription elongation where upstream DNA emerging from the Pol II cleft has rewrapped the proximal side of the nucleosome. The structure uncovers a direct role for Pol II and transcription elongation factors in nucleosome retention and explains how nucleosomes are retained to prevent the disruption of chromatin structure across actively transcribed genes.


Subject(s)
Nucleosomes , RNA Polymerase II , Transcription Elongation, Genetic , Animals , Chromatin/chemistry , Cryoelectron Microscopy , DNA/genetics , Humans , Nucleosomes/chemistry , RNA Polymerase II/chemistry , Sus scrofa
8.
Curr Opin Struct Biol ; 73: 102335, 2022 04.
Article in English | MEDLINE | ID: mdl-35183822

ABSTRACT

The first step of eukaryotic gene expression is the assembly of RNA polymerase II and general transcription factors on promoter DNA. This highly regulated process involves ∼80 different proteins that together form the preinitiation complex (PIC). Decades of work have gone into understanding PIC assembly using biochemical and structural approaches. These efforts have yielded significant but partial descriptions of PIC assembly. Over the past few years, cryo-electron microscopy has provided the first high-resolution structures of the near-complete mammalian PIC assembly. These structures have revealed that PIC assembly is a highly dynamic process. This review will summarize recent structural findings and discuss their implications for understanding cell type-specific gene expression.


Subject(s)
RNA Polymerase II , Transcription, Genetic , Animals , Cryoelectron Microscopy , Mammals/genetics , Mammals/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism
9.
Mol Cell ; 81(15): 3096-3109.e8, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34146481

ABSTRACT

Transcription by RNA polymerase II (RNA Pol II) relies on the elongation factors PAF1 complex (PAF), RTF1, and SPT6. Here, we use rapid factor depletion and multi-omics analysis to investigate how these elongation factors influence RNA Pol II elongation activity in human cells. Whereas depletion of PAF subunits PAF1 and CTR9 has little effect on cellular RNA synthesis, depletion of RTF1 or SPT6 strongly compromises RNA Pol II activity, albeit in fundamentally different ways. RTF1 depletion decreases RNA Pol II velocity, whereas SPT6 depletion impairs RNA Pol II progression through nucleosomes. These results show that distinct elongation factors stimulate either RNA Pol II velocity or RNA Pol II progression through chromatin in vivo. Further analysis provides evidence for two distinct barriers to early elongation: the promoter-proximal pause site and the +1 nucleosome. It emerges that the first barrier enables loading of elongation factors that are required to overcome the second and subsequent barriers to transcription.


Subject(s)
RNA Polymerase II/metabolism , RNA/biosynthesis , Transcription Factors/metabolism , Humans , K562 Cells , Nucleosomes/genetics , Nucleosomes/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA Polymerase II/genetics , Transcription Factors/genetics
10.
Nat Commun ; 12(1): 3232, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34050140

ABSTRACT

Arrays of regularly spaced nucleosomes dominate chromatin and are often phased by alignment to reference sites like active promoters. How the distances between nucleosomes (spacing), and between phasing sites and nucleosomes are determined remains unclear, and specifically, how ATP-dependent chromatin remodelers impact these features. Here, we used genome-wide reconstitution to probe how Saccharomyces cerevisiae ATP-dependent remodelers generate phased arrays of regularly spaced nucleosomes. We find that remodelers bear a functional element named the 'ruler' that determines spacing and phasing in a remodeler-specific way. We use structure-based mutagenesis to identify and tune the ruler element residing in the Nhp10 and Arp8 modules of the INO80 remodeler complex. Generally, we propose that a remodeler ruler regulates nucleosome sliding direction bias in response to (epi)genetic information. This finally conceptualizes how remodeler-mediated nucleosome dynamics determine stable steady-state nucleosome positioning relative to other nucleosomes, DNA bound factors, DNA ends and DNA sequence elements.


Subject(s)
Chromatin Assembly and Disassembly , Nucleosomes/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Animals , Drosophila Proteins/genetics , Drosophila Proteins/isolation & purification , Drosophila Proteins/metabolism , Drosophila melanogaster , Epigenesis, Genetic , Genome, Fungal/genetics , High Mobility Group Proteins/genetics , High Mobility Group Proteins/isolation & purification , High Mobility Group Proteins/metabolism , Histones/genetics , Histones/metabolism , Larva/genetics , Larva/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/isolation & purification , Microfilament Proteins/metabolism , Mutagenesis , Nucleosomes/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/isolation & purification , Saccharomyces cerevisiae Proteins/metabolism , Whole Genome Sequencing
11.
Nat Struct Mol Biol ; 28(4): 382-387, 2021 04.
Article in English | MEDLINE | ID: mdl-33846633

ABSTRACT

Efficient transcription of RNA polymerase II (Pol II) through nucleosomes requires the help of various factors. Here we show biochemically that Pol II transcription through a nucleosome is facilitated by the chromatin remodeler Chd1 and the histone chaperone FACT when the elongation factors Spt4/5 and TFIIS are present. We report cryo-EM structures of transcribing Saccharomyces cerevisiae Pol II-Spt4/5-nucleosome complexes with bound Chd1 or FACT. In the first structure, Pol II transcription exposes the proximal histone H2A-H2B dimer that is bound by Spt5. Pol II has also released the inhibitory DNA-binding region of Chd1 that is poised to pump DNA toward Pol II. In the second structure, Pol II has generated a partially unraveled nucleosome that binds FACT, which excludes Chd1 and Spt5. These results suggest that Pol II progression through a nucleosome activates Chd1, enables FACT binding and eventually triggers transfer of FACT together with histones to upstream DNA.


Subject(s)
Chromosomal Proteins, Non-Histone/ultrastructure , DNA-Binding Proteins/ultrastructure , High Mobility Group Proteins/ultrastructure , Saccharomyces cerevisiae Proteins/ultrastructure , Transcription, Genetic , Transcriptional Elongation Factors/ultrastructure , Chromatin/genetics , Chromatin/ultrastructure , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Histones/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , Nucleosomes/genetics , Nucleosomes/ultrastructure , RNA Polymerase II/genetics , RNA Polymerase II/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/genetics , Transcriptional Elongation Factors/genetics
12.
Nat Commun ; 12(1): 279, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436624

ABSTRACT

Remdesivir is the only FDA-approved drug for the treatment of COVID-19 patients. The active form of remdesivir acts as a nucleoside analog and inhibits the RNA-dependent RNA polymerase (RdRp) of coronaviruses including SARS-CoV-2. Remdesivir is incorporated by the RdRp into the growing RNA product and allows for addition of three more nucleotides before RNA synthesis stalls. Here we use synthetic RNA chemistry, biochemistry and cryo-electron microscopy to establish the molecular mechanism of remdesivir-induced RdRp stalling. We show that addition of the fourth nucleotide following remdesivir incorporation into the RNA product is impaired by a barrier to further RNA translocation. This translocation barrier causes retention of the RNA 3'-nucleotide in the substrate-binding site of the RdRp and interferes with entry of the next nucleoside triphosphate, thereby stalling RdRp. In the structure of the remdesivir-stalled state, the 3'-nucleotide of the RNA product is matched and located with the template base in the active center, and this may impair proofreading by the viral 3'-exonuclease. These mechanistic insights should facilitate the quest for improved antivirals that target coronavirus replication.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , RNA-Dependent RNA Polymerase/drug effects , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , Aptamers, Nucleotide , Coronavirus RNA-Dependent RNA Polymerase/drug effects , Nucleotides , RNA, Viral , RNA-Dependent RNA Polymerase/genetics , SARS-CoV-2/enzymology , Virus Replication/drug effects , COVID-19 Drug Treatment
13.
Nat Struct Mol Biol ; 27(7): 668-677, 2020 07.
Article in English | MEDLINE | ID: mdl-32541898

ABSTRACT

Transcription by RNA polymerase II (Pol II) is carried out by an elongation complex. We previously reported an activated porcine Pol II elongation complex, EC*, encompassing the human elongation factors DSIF, PAF1 complex (PAF) and SPT6. Here we report the cryo-EM structure of the complete EC* that contains RTF1, a dissociable PAF subunit critical for chromatin transcription. The RTF1 Plus3 domain associates with Pol II subunit RPB12 and the phosphorylated C-terminal region of DSIF subunit SPT5. RTF1 also forms four α-helices that extend from the Plus3 domain along the Pol II protrusion and RPB10 to the polymerase funnel. The C-terminal 'fastener' helix retains PAF and is followed by a 'latch' that reaches the end of the bridge helix, a flexible element of the Pol II active site. RTF1 strongly stimulates Pol II elongation, and this requires the latch, possibly suggesting that RTF1 activates transcription allosterically by influencing Pol II translocation.


Subject(s)
Multiprotein Complexes/chemistry , RNA Polymerase II/metabolism , Transcription Factors/chemistry , Allosteric Regulation , Cryoelectron Microscopy , Humans , Models, Molecular , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation , RNA Polymerase II/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/metabolism
14.
Elife ; 92020 06 16.
Article in English | MEDLINE | ID: mdl-32543371

ABSTRACT

Chromatin remodeling plays important roles in gene regulation during development, differentiation and in disease. The chromatin remodeling enzyme CHD4 is a component of the NuRD and ChAHP complexes that are involved in gene repression. Here, we report the cryo-electron microscopy (cryo-EM) structure of Homo sapiens CHD4 engaged with a nucleosome core particle in the presence of the non-hydrolysable ATP analogue AMP-PNP at an overall resolution of 3.1 Å. The ATPase motor of CHD4 binds and distorts nucleosomal DNA at superhelical location (SHL) +2, supporting the 'twist defect' model of chromatin remodeling. CHD4 does not induce unwrapping of terminal DNA, in contrast to its homologue Chd1, which functions in gene activation. Our structure also maps CHD4 mutations that are associated with human cancer or the intellectual disability disorder Sifrim-Hitz-Weiss syndrome.


Subject(s)
Adenylyl Imidodiphosphate/metabolism , Chromatin Assembly and Disassembly/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mutation , Nucleosomes/metabolism , Animals , Cryoelectron Microscopy , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Xenopus laevis
15.
Nature ; 584(7819): 154-156, 2020 08.
Article in English | MEDLINE | ID: mdl-32438371

ABSTRACT

The new coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses an RNA-dependent RNA polymerase (RdRp) for the replication of its genome and the transcription of its genes1-3. Here we present a cryo-electron microscopy structure of the SARS-CoV-2 RdRp in an active form that mimics the replicating enzyme. The structure comprises the viral proteins non-structural protein 12 (nsp12), nsp8 and nsp7, and more than two turns of RNA template-product duplex. The active-site cleft of nsp12 binds to the first turn of RNA and mediates RdRp activity with conserved residues. Two copies of nsp8 bind to opposite sides of the cleft and position the second turn of RNA. Long helical extensions in nsp8 protrude along exiting RNA, forming positively charged 'sliding poles'. These sliding poles can account for the known processivity of RdRp that is required for replicating the long genome of coronaviruses3. Our results enable a detailed analysis of the inhibitory mechanisms that underlie the antiviral activity of substances such as remdesivir, a drug for the treatment of coronavirus disease 2019 (COVID-19)4.


Subject(s)
Betacoronavirus/enzymology , Cryoelectron Microscopy , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Betacoronavirus/drug effects , Betacoronavirus/genetics , Betacoronavirus/ultrastructure , Coronavirus RNA-Dependent RNA Polymerase , Models, Molecular , Protein Conformation , RNA, Viral/chemistry , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/ultrastructure , SARS-CoV-2 , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/ultrastructure
16.
Nat Struct Mol Biol ; 27(1): 8-13, 2020 01.
Article in English | MEDLINE | ID: mdl-31819277

ABSTRACT

Recognition of histone-modified nucleosomes by specific reader domains underlies the regulation of chromatin-associated processes. Whereas structural studies revealed how reader domains bind modified histone peptides, it is unclear how reader domains interact with modified nucleosomes. Here, we report the cryo-electron microscopy structure of the PWWP reader domain of human transcriptional coactivator LEDGF in complex with an H3K36-methylated nucleosome at 3.2-Å resolution. The structure reveals multivalent binding of the reader domain to the methylated histone tail and to both gyres of nucleosomal DNA, explaining the known cooperative interactions. The observed cross-gyre binding may contribute to nucleosome integrity during transcription. The structure also explains how human PWWP domain-containing proteins are recruited to H3K36-methylated regions of the genome for transcription, histone acetylation and methylation, and for DNA methylation and repair.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Histones/metabolism , Nucleosomes/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Sequence , Cryoelectron Microscopy , DNA/chemistry , DNA/metabolism , Histones/chemistry , Humans , Methylation , Models, Molecular , Nucleosomes/chemistry , Protein Binding , Protein Conformation , Protein Domains , Sequence Alignment , Transcription Factors/chemistry
17.
Nat Commun ; 9(1): 5432, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30575770

ABSTRACT

Transcription of eukaryotic protein-coding genes requires passage of RNA polymerase II (Pol II) through nucleosomes, but it is unclear how this is achieved. Here we report the cryo-EM structure of transcribing Saccharomyces cerevisiae Pol II engaged with a downstream nucleosome core particle at an overall resolution of 4.4 Å. Pol II and the nucleosome are observed in a defined relative orientation that is not predicted. Pol II contacts both sides of the nucleosome dyad using its clamp head and lobe domains. Structural comparisons reveal that the elongation factors TFIIS, DSIF, NELF, SPT6, and PAF1 complex can be accommodated on the Pol II surface in the presence of the oriented nucleosome. Our results provide a starting point for analysing the mechanisms of chromatin transcription.


Subject(s)
Nucleosomes/metabolism , Nucleosomes/ultrastructure , RNA Polymerase II/metabolism , RNA Polymerase II/ultrastructure , Animals , Cryoelectron Microscopy , Molecular Structure , Nucleosomes/chemistry , Protein Conformation , RNA Polymerase II/chemistry , Saccharomyces cerevisiae , Xenopus laevis
18.
Nature ; 562(7725): 76-81, 2018 10.
Article in English | MEDLINE | ID: mdl-30250250

ABSTRACT

Nucleosomes cover most of the genome and are thought to be displaced by transcription factors in regions that direct gene expression. However, the modes of interaction between transcription factors and nucleosomal DNA remain largely unknown. Here we systematically explore interactions between the nucleosome and 220 transcription factors representing diverse structural families. Consistent with earlier observations, we find that the majority of the studied transcription factors have less access to nucleosomal DNA than to free DNA. The motifs recovered from transcription factors bound to nucleosomal and free DNA are generally similar. However, steric hindrance and scaffolding by the nucleosome result in specific positioning and orientation of the motifs. Many transcription factors preferentially bind close to the end of nucleosomal DNA, or to periodic positions on the solvent-exposed side of the DNA. In addition, several transcription factors usually bind to nucleosomal DNA in a particular orientation. Some transcription factors specifically interact with DNA located at the dyad position at which only one DNA gyre is wound, whereas other transcription factors prefer sites spanning two DNA gyres and bind specifically to each of them. Our work reveals notable differences in the binding of transcription factors to free and nucleosomal DNA, and uncovers a diverse interaction landscape between transcription factors and the nucleosome.


Subject(s)
Nucleosomes/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , DNA/chemistry , DNA/genetics , DNA/metabolism , Humans , Mice , Models, Molecular , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleotide Motifs , Protein Binding , Rotation , SELEX Aptamer Technique , Transcription Factors/chemistry , Transcription Factors/classification
19.
Nature ; 560(7720): 607-612, 2018 08.
Article in English | MEDLINE | ID: mdl-30135578

ABSTRACT

Gene regulation involves activation of RNA polymerase II (Pol II) that is paused and bound by the protein complexes DRB sensitivity-inducing factor (DSIF) and negative elongation factor (NELF). Here we show that formation of an activated Pol II elongation complex in vitro requires the kinase function of the positive transcription elongation factor b (P-TEFb) and the elongation factors PAF1 complex (PAF) and SPT6. The cryo-EM structure of an activated elongation complex of Sus scrofa Pol II and Homo sapiens DSIF, PAF and SPT6 was determined at 3.1 Å resolution and compared to the structure of the paused elongation complex formed by Pol II, DSIF and NELF. PAF displaces NELF from the Pol II funnel for pause release. P-TEFb phosphorylates the Pol II linker to the C-terminal domain. SPT6 binds to the phosphorylated C-terminal-domain linker and opens the RNA clamp formed by DSIF. These results provide the molecular basis for Pol II pause release and elongation activation.


Subject(s)
Cryoelectron Microscopy , Nuclear Proteins/ultrastructure , RNA Polymerase II/metabolism , RNA Polymerase II/ultrastructure , Transcription Factors/ultrastructure , Transcriptional Elongation Factors/ultrastructure , Animals , DNA/chemistry , DNA/ultrastructure , Humans , Models, Molecular , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Phosphoproteins/ultrastructure , Positive Transcriptional Elongation Factor B/metabolism , RNA/chemistry , RNA/ultrastructure , Sus scrofa , Transcription Elongation, Genetic , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism
20.
Nature ; 560(7720): 601-606, 2018 08.
Article in English | MEDLINE | ID: mdl-30135580

ABSTRACT

Metazoan gene regulation often involves the pausing of RNA polymerase II (Pol II) in the promoter-proximal region. Paused Pol II is stabilized by the protein complexes DRB sensitivity-inducing factor (DSIF) and negative elongation factor (NELF). Here we report the cryo-electron microscopy structure of a paused transcription elongation complex containing Sus scrofa Pol II and Homo sapiens DSIF and NELF at 3.2 Å resolution. The structure reveals a tilted DNA-RNA hybrid that impairs binding of the nucleoside triphosphate substrate. NELF binds the polymerase funnel, bridges two mobile polymerase modules, and contacts the trigger loop, thereby restraining Pol II mobility that is required for pause release. NELF prevents binding of the anti-pausing transcription elongation factor IIS (TFIIS). Additionally, NELF possesses two flexible 'tentacles' that can contact DSIF and exiting RNA. These results define the paused state of Pol II and provide the molecular basis for understanding the function of NELF during promoter-proximal gene regulation.


Subject(s)
Cryoelectron Microscopy , Nuclear Proteins/ultrastructure , RNA Polymerase II/metabolism , RNA Polymerase II/ultrastructure , Transcription Elongation, Genetic , Transcription Factors/ultrastructure , Transcriptional Elongation Factors/ultrastructure , Animals , DNA/genetics , DNA/metabolism , HIV-1/genetics , Humans , Models, Molecular , Movement , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Conformation , Proviruses/genetics , RNA/genetics , RNA/metabolism , Sus scrofa , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...