Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Nat Commun ; 15(1): 4189, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760379

ABSTRACT

The viral polymerase complex, comprising the large protein (L) and phosphoprotein (P), is crucial for both genome replication and transcription in non-segmented negative-strand RNA viruses (nsNSVs), while structures corresponding to these activities remain obscure. Here, we resolved two L-P complex conformations from the mumps virus (MuV), a typical member of nsNSVs, via cryogenic-electron microscopy. One conformation presents all five domains of L forming a continuous RNA tunnel to the methyltransferase domain (MTase), preferably as a transcription state. The other conformation has the appendage averaged out, which is inaccessible to MTase. In both conformations, parallel P tetramers are revealed around MuV L, which, together with structures of other nsNSVs, demonstrates the diverse origins of the L-binding X domain of P. Our study links varying structures of nsNSV polymerase complexes with genome replication and transcription and points to a sliding model for polymerase complexes to advance along the RNA templates.


Subject(s)
Cryoelectron Microscopy , Mumps virus , Viral Proteins , Mumps virus/genetics , Mumps virus/ultrastructure , Mumps virus/metabolism , Viral Proteins/metabolism , Viral Proteins/ultrastructure , Viral Proteins/chemistry , Viral Proteins/genetics , Models, Molecular , RNA, Viral/metabolism , RNA, Viral/ultrastructure , RNA, Viral/genetics , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Protein Domains , Phosphoproteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/ultrastructure , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/ultrastructure , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Virus Replication , Transcription, Genetic , Protein Conformation
2.
J Mol Biol ; 436(10): 168568, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38583515

ABSTRACT

Porphyromonas gingivalis, an anaerobic CFB (Cytophaga, Fusobacterium, and Bacteroides) group bacterium, is the keystone pathogen of periodontitis and has been implicated in various systemic diseases. Increased antibiotic resistance and lack of effective antibiotics necessitate a search for new intervention strategies. Here we report a 3.5 Å resolution cryo-EM structure of P. gingivalis RNA polymerase (RNAP). The structure displays new structural features in its ω subunit and multiple domains in ß and ß' subunits, which differ from their counterparts in other bacterial RNAPs. Superimpositions with E. coli RNAP holoenzyme and initiation complex further suggest that its ω subunit may contact the σ4 domain, thereby possibly contributing to the assembly and stabilization of initiation complexes. In addition to revealing the unique features of P. gingivalis RNAP, our work offers a framework for future studies of transcription regulation in this important pathogen, as well as for structure-based drug development.


Subject(s)
Cryoelectron Microscopy , DNA-Directed RNA Polymerases , Models, Molecular , Porphyromonas gingivalis , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/genetics , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , DNA-Directed RNA Polymerases/genetics , Protein Conformation , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/enzymology , Protein Subunits/chemistry , Protein Subunits/metabolism
3.
Nature ; 622(7984): 872-879, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37821701

ABSTRACT

Transcription initiation is a key regulatory step in gene expression during which RNA polymerase (RNAP) initiates RNA synthesis de novo, and the synthesized RNA at a specific length triggers the transition to the elongation phase. Mitochondria recruit a single-subunit RNAP and one or two auxiliary factors to initiate transcription. Previous studies have revealed the molecular architectures of yeast1 and human2 mitochondrial RNAP initiation complexes (ICs). Here we provide a comprehensive, stepwise mechanism of transcription initiation by solving high-resolution cryogenic electron microscopy (cryo-EM) structures of yeast mitochondrial RNAP and the transcription factor Mtf1 catalysing two- to eight-nucleotide RNA synthesis at single-nucleotide addition steps. The growing RNA-DNA is accommodated in the polymerase cleft by template scrunching and non-template reorganization, creating stressed intermediates. During early initiation, non-template strand scrunching and unscrunching destabilize the short two- and three-nucleotide RNAs, triggering abortive synthesis. Subsequently, the non-template reorganizes into a base-stacked staircase-like structure supporting processive five- to eight-nucleotide RNA synthesis. The expanded non-template staircase and highly scrunched template in IC8 destabilize the promoter interactions with Mtf1 to facilitate initiation bubble collapse and promoter escape for the transition from initiation to the elongation complex (EC). The series of transcription initiation steps, each guided by the interplay of multiple structural components, reveal a finely tuned mechanism for potential regulatory control.


Subject(s)
Mitochondria , Saccharomyces cerevisiae , Transcription Initiation, Genetic , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/ultrastructure , Nucleotides/metabolism , RNA/biosynthesis , RNA/ultrastructure , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Cryoelectron Microscopy , DNA/metabolism , DNA/ultrastructure
4.
Nature ; 613(7945): 783-789, 2023 01.
Article in English | MEDLINE | ID: mdl-36631609

ABSTRACT

Efficient and accurate termination is required for gene transcription in all living organisms1,2. Cellular RNA polymerases in both bacteria and eukaryotes can terminate their transcription through a factor-independent termination pathway3,4-called intrinsic termination transcription in bacteria-in which RNA polymerase recognizes terminator sequences, stops nucleotide addition and releases nascent RNA spontaneously. Here we report a set of single-particle cryo-electron microscopy structures of Escherichia coli transcription intrinsic termination complexes representing key intermediate states of the event. The structures show how RNA polymerase pauses at terminator sequences, how the terminator RNA hairpin folds inside RNA polymerase, and how RNA polymerase rewinds the transcription bubble to release RNA and then DNA. These macromolecular snapshots define a structural mechanism for bacterial intrinsic termination and a pathway for RNA release and DNA collapse that is relevant for factor-independent termination by all RNA polymerases.


Subject(s)
DNA, Bacterial , DNA-Directed RNA Polymerases , Escherichia coli , RNA, Bacterial , Transcription Termination, Genetic , Cryoelectron Microscopy , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/ultrastructure , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Bacterial/ultrastructure , Terminator Regions, Genetic/genetics , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , DNA, Bacterial/ultrastructure
5.
Nature ; 614(7947): 367-374, 2023 02.
Article in English | MEDLINE | ID: mdl-36697824

ABSTRACT

Rho is a ring-shaped hexameric ATP-dependent molecular motor. Together with the transcription elongation factor NusG, Rho mediates factor-dependent transcription termination and transcription-translation-coupling quality control in Escherichia coli1-4. Here we report the preparation of complexes that are functional in factor-dependent transcription termination from Rho, NusG, RNA polymerase (RNAP), and synthetic nucleic acid scaffolds, and we report cryogenic electron microscopy structures of the complexes. The structures show that functional factor-dependent pre-termination complexes contain a closed-ring Rho hexamer; have RNA threaded through the central channel of Rho; have 60 nucleotides of RNA interacting sequence-specifically with the exterior of Rho and 6 nucleotides of RNA interacting sequence-specifically with the central channel of Rho; have Rho oriented relative to RNAP such that ATP-dependent translocation by Rho exerts mechanical force on RNAP; and have NusG bridging Rho and RNAP. The results explain five decades of research on Rho and provide a foundation for understanding Rho's function.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Transcription Factors , Transcription Termination, Genetic , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , Escherichia coli/chemistry , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/ultrastructure , RNA/chemistry , RNA/genetics , RNA/metabolism , RNA/ultrastructure , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcription Factors/ultrastructure
6.
Nature ; 610(7931): 394-401, 2022 10.
Article in English | MEDLINE | ID: mdl-36171293

ABSTRACT

Filoviruses, including Ebola virus, pose an increasing threat to the public health. Although two therapeutic monoclonal antibodies have been approved to treat the Ebola virus disease1,2, there are no approved broadly reactive drugs to control diverse filovirus infection. Filovirus has a large polymerase (L) protein and the cofactor viral protein 35 (VP35), which constitute the basic functional unit responsible for virus genome RNA synthesis3. Owing to its conservation, the L-VP35 polymerase complex is a promising target for broadly reactive antiviral drugs. Here we determined the structure of Ebola virus L protein in complex with tetrameric VP35 using cryo-electron microscopy (state 1). Structural analysis revealed that Ebola virus L possesses a filovirus-specific insertion element that is essential for RNA synthesis, and that VP35 interacts extensively with the N-terminal region of L by three protomers of the VP35 tetramer. Notably, we captured the complex structure in a second conformation with the unambiguous priming loop and supporting helix away from polymerase active site (state 2). Moreover, we demonstrated that the century-old drug suramin could inhibit the activity of the Ebola virus polymerase in an enzymatic assay. The structure of the L-VP35-suramin complex reveals that suramin can bind at the highly conserved NTP entry channel to prevent substrates from entering the active site. These findings reveal the mechanism of Ebola virus replication and may guide the development of more powerful anti-filovirus drugs.


Subject(s)
Cryoelectron Microscopy , DNA-Directed RNA Polymerases , Ebolavirus , Viral Regulatory and Accessory Proteins , Antiviral Agents/pharmacology , Catalytic Domain , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , Ebolavirus/enzymology , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Humans , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA, Viral/biosynthesis , Suramin/chemistry , Suramin/metabolism , Suramin/pharmacology , Suramin/therapeutic use , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/metabolism , Viral Regulatory and Accessory Proteins/ultrastructure , Virus Replication
7.
Commun Biol ; 4(1): 874, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34267316

ABSTRACT

Cryo-EM maps are valuable sources of information for protein structure modeling. However, due to the loss of contrast at high frequencies, they generally need to be post-processed to improve their interpretability. Most popular approaches, based on global B-factor correction, suffer from limitations. For instance, they ignore the heterogeneity in the map local quality that reconstructions tend to exhibit. Aiming to overcome these problems, we present DeepEMhancer, a deep learning approach designed to perform automatic post-processing of cryo-EM maps. Trained on a dataset of pairs of experimental maps and maps sharpened using their respective atomic models, DeepEMhancer has learned how to post-process experimental maps performing masking-like and sharpening-like operations in a single step. DeepEMhancer was evaluated on a testing set of 20 different experimental maps, showing its ability to reduce noise levels and obtain more detailed versions of the experimental maps. Additionally, we illustrated the benefits of DeepEMhancer on the structure of the SARS-CoV-2 RNA polymerase.


Subject(s)
Cryoelectron Microscopy/instrumentation , DNA-Directed RNA Polymerases/ultrastructure , Deep Learning , SARS-CoV-2/ultrastructure , Viral Proteins/ultrastructure
8.
STAR Protoc ; 2(2): 100431, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33870232

ABSTRACT

In yeast mitochondria, transcription initiation requires assembly of mitochondrial RNA polymerase and transcription initiation factor MTF1 at the DNA promoter initiation site. This protocol describes the purification of the component proteins and assembly of partially melted and fully melted initiation complex states. Both states co-exist in equilibrium in the same sample as seen by cryoelectron microscopy (cryo-EM) and allow elucidation of MTF1's structural roles in controlling the transition into elongation. We further outline how analysis of the complex by light scattering, thermal shift assay, and ultrafiltration assay exhibits reproducible results. For complete details on the use and execution of this protocol, please refer to De Wijngaert et al. (2021).


Subject(s)
Cryoelectron Microscopy/methods , DNA-Directed RNA Polymerases , Mitochondrial Proteins , Saccharomyces cerevisiae Proteins , Transcription Factors , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/ultrastructure , Mitochondrial Ribosomes , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcription Factors/ultrastructure
9.
Nat Commun ; 12(1): 796, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542236

ABSTRACT

RNA polymerases (RNAPs) synthesize RNA from NTPs, whereas DNA polymerases synthesize DNA from 2'dNTPs. DNA polymerases select against NTPs by using steric gates to exclude the 2'OH, but RNAPs have to employ alternative selection strategies. In single-subunit RNAPs, a conserved Tyr residue discriminates against 2'dNTPs, whereas selectivity mechanisms of multi-subunit RNAPs remain hitherto unknown. Here, we show that a conserved Arg residue uses a two-pronged strategy to select against 2'dNTPs in multi-subunit RNAPs. The conserved Arg interacts with the 2'OH group to promote NTP binding, but selectively inhibits incorporation of 2'dNTPs by interacting with their 3'OH group to favor the catalytically-inert 2'-endo conformation of the deoxyribose moiety. This deformative action is an elegant example of an active selection against a substrate that is a substructure of the correct substrate. Our findings provide important insights into the evolutionary origins of biopolymers and the design of selective inhibitors of viral RNAPs.


Subject(s)
Bacterial Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , Deoxyribonucleotides/metabolism , Deoxyribose/metabolism , Arginine/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/ultrastructure , Crystallography, X-Ray , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/isolation & purification , DNA-Directed RNA Polymerases/ultrastructure , Escherichia coli/enzymology , Escherichia coli/genetics , Kinetics , Molecular Docking Simulation , Promoter Regions, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Substrate Specificity , Thermus thermophilus/enzymology , Thermus thermophilus/genetics
10.
Nat Commun ; 12(1): 528, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33483500

ABSTRACT

Ribosomal RNA (rRNA) is most highly expressed in rapidly growing bacteria and is drastically downregulated under stress conditions by the global transcriptional regulator DksA and the alarmone ppGpp. Here, we determined cryo-electron microscopy structures of the Escherichia coli RNA polymerase (RNAP) σ70 holoenzyme during rRNA promoter recognition with and without DksA/ppGpp. RNAP contacts the UP element using dimerized α subunit carboxyl-terminal domains and scrunches the template DNA with the σ finger and ß' lid to select the transcription start site favorable for rapid promoter escape. Promoter binding induces conformational change of σ domain 2 that opens a gate for DNA loading and ejects σ1.1 from the RNAP cleft to facilitate open complex formation. DksA/ppGpp binding also opens the DNA loading gate, which is not coupled to σ1.1 ejection and impedes open complex formation. These results provide a molecular basis for the exceptionally active rRNA transcription and its vulnerability to DksA/ppGpp.


Subject(s)
Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Promoter Regions, Genetic/genetics , RNA, Ribosomal/genetics , Transcription, Genetic , Cryoelectron Microscopy , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Guanosine Tetraphosphate/metabolism , Holoenzymes/chemistry , Holoenzymes/metabolism , Holoenzymes/ultrastructure , Protein Conformation , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Ribosomal/metabolism , Sigma Factor/chemistry , Sigma Factor/metabolism , Sigma Factor/ultrastructure , Transcription Initiation Site
11.
Viral Immunol ; 34(1): 18-26, 2021.
Article in English | MEDLINE | ID: mdl-32429800

ABSTRACT

The resolution revolution of cryo-electron microscopy (cryo-EM) has made a significant impact on the structural analysis of the Pneumoviridae multifunctional RNA polymerases. In recent months, several high-resolution structures of apo RNA polymerases of Pneumoviridae, which includes the human respiratory syncytial virus (HRSV) and human metapneumovirus (HMPV), have been determined by single-particle cryo-EM. These structures illustrated high similarities and minor differences between the Pneumoviridae polymerases and revealed the potential mechanisms of the Pneumoviridae RNA synthesis.


Subject(s)
Cryoelectron Microscopy , DNA-Directed RNA Polymerases/ultrastructure , Pneumovirus/enzymology , Humans , Respiratory Syncytial Virus, Human/enzymology
12.
Nat Commun ; 11(1): 6284, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33293519

ABSTRACT

The MerR-family proteins represent a unique family of bacteria transcription factors (TFs), which activate transcription in a manner distinct from canonical ones. Here, we report a cryo-EM structure of a B. subtilis transcription activation complex comprising B. subtilis six-subunit (2αßß'ωε) RNA Polymerase (RNAP) core enzyme, σA, a promoter DNA, and the ligand-bound B. subtilis BmrR, a prototype of MerR-family TFs. The structure reveals that RNAP and BmrR recognize the upstream promoter DNA from opposite faces and induce four significant kinks from the -35 element to the -10 element of the promoter DNA in a cooperative manner, which restores otherwise inactive promoter activity by shortening the length of promoter non-optimal -35/-10 spacer. Our structure supports a DNA-distortion and RNAP-non-contact paradigm of transcriptional activation by MerR TFs.


Subject(s)
Bacillus subtilis/genetics , Bacterial Proteins/metabolism , Drug Resistance, Multiple, Bacterial/genetics , Gene Expression Regulation, Bacterial , Trans-Activators/metabolism , Transcriptional Activation , Bacillus subtilis/drug effects , Bacterial Proteins/ultrastructure , Cryoelectron Microscopy , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , Promoter Regions, Genetic/genetics , Trans-Activators/ultrastructure
13.
Nat Commun ; 11(1): 6420, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33339820

ABSTRACT

In bacteria, transcription complexes stalled on DNA represent a major source of roadblocks for the DNA replication machinery that must be removed in order to prevent damaging collisions. Gram-positive bacteria contain a transcription factor HelD that is able to remove and recycle stalled complexes, but it was not known how it performed this function. Here, using single particle cryo-electron microscopy, we have determined the structures of Bacillus subtilis RNA polymerase (RNAP) elongation and HelD complexes, enabling analysis of the conformational changes that occur in RNAP driven by HelD interaction. HelD has a 2-armed structure which penetrates deep into the primary and secondary channels of RNA polymerase. One arm removes nucleic acids from the active site, and the other induces a large conformational change in the primary channel leading to removal and recycling of the stalled polymerase, representing a novel mechanism for recycling transcription complexes in bacteria.


Subject(s)
Bacillus subtilis/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , Transcription, Genetic , Bacterial Proteins/ultrastructure , Cryoelectron Microscopy , DNA-Directed RNA Polymerases/ultrastructure , Imaging, Three-Dimensional , Models, Molecular , Protein Binding , Transcription Elongation, Genetic
14.
Nat Commun ; 11(1): 6419, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33339823

ABSTRACT

RNA synthesis is central to life, and RNA polymerase (RNAP) depends on accessory factors for recovery from stalled states and adaptation to environmental changes. Here, we investigated the mechanism by which a helicase-like factor HelD recycles RNAP. We report a cryo-EM structure of a complex between the Mycobacterium smegmatis RNAP and HelD. The crescent-shaped HelD simultaneously penetrates deep into two RNAP channels that are responsible for nucleic acids binding and substrate delivery to the active site, thereby locking RNAP in an inactive state. We show that HelD prevents non-specific interactions between RNAP and DNA and dissociates stalled transcription elongation complexes. The liberated RNAP can either stay dormant, sequestered by HelD, or upon HelD release, restart transcription. Our results provide insights into the architecture and regulation of the highly medically-relevant mycobacterial transcription machinery and define HelD as a clearing factor that releases RNAP from nonfunctional complexes with nucleic acids.


Subject(s)
Bacterial Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , Mycobacterium smegmatis/enzymology , Nucleic Acids/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Catalytic Domain , Cryoelectron Microscopy , DNA, Bacterial/chemistry , DNA, Bacterial/metabolism , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/ultrastructure , Models, Molecular , Protein Binding , Protein Domains
15.
Nucleic Acids Res ; 48(16): 9195-9203, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32810236

ABSTRACT

G-quadruplex (GQ) is formed at various regions of DNA, including telomeres of chromosomes and regulatory regions of oncogenes. Since GQ is important in both gene regulation and genome instability, the biological and medical implications of this abnormal DNA structure have been intensively studied. Its formation mechanisms, however, are not clearly understood yet. We report single-molecule fluorescence experiments to monitor the cotranscriptional GQ formation coupled with R-loop formation using T7 RNA polymerase. The GQ is formed very rarely per single-round transcription. R-loop formation precedes and facilitates GQ formation. Once formed, some GQs are extremely stable, resistant even to RNase H treatment, and accumulate in multiple-round transcription conditions. On the other hand, GQ existing in the non-template strand promotes the R-loop formation in the next rounds of transcription. Our study clearly shows the existence of a positive feedback mechanism of GQ and R-loop formations, which may possibly contribute to gene regulation and genome instability.


Subject(s)
DNA/ultrastructure , G-Quadruplexes , R-Loop Structures/genetics , Single Molecule Imaging/methods , DNA-Directed RNA Polymerases/ultrastructure , Fluorescence , Fluorescence Resonance Energy Transfer , Humans , Telomere/ultrastructure , Viral Proteins/ultrastructure
16.
Proc Natl Acad Sci U S A ; 117(27): 15642-15649, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32571927

ABSTRACT

The RNA polymerase (RNAP) trigger loop (TL) is a mobile structural element of the RNAP active center that, based on crystal structures, has been proposed to cycle between an "unfolded"/"open" state that allows an NTP substrate to enter the active center and a "folded"/"closed" state that holds the NTP substrate in the active center. Here, by quantifying single-molecule fluorescence resonance energy transfer between a first fluorescent probe in the TL and a second fluorescent probe elsewhere in RNAP or in DNA, we detect and characterize TL closing and opening in solution. We show that the TL closes and opens on the millisecond timescale; we show that TL closing and opening provides a checkpoint for NTP complementarity, NTP ribo/deoxyribo identity, and NTP tri/di/monophosphate identity, and serves as a target for inhibitors; and we show that one cycle of TL closing and opening typically occurs in each nucleotide addition cycle in transcription elongation.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , DNA/chemistry , RNA/chemistry , Transcription, Genetic , Catalytic Domain , DNA/genetics , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/ultrastructure , Escherichia coli/genetics , Fluorescence Resonance Energy Transfer , Models, Molecular , Nucleotides , Protein Conformation , Protein Folding , RNA/genetics , Single Molecule Imaging
17.
Nat Commun ; 10(1): 3916, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31477705

ABSTRACT

Transcription by RNA polymerase V (Pol V) in plants is required for RNA-directed DNA methylation, leading to transcriptional gene silencing. Global chromatin association of Pol V requires components of the DDR complex DRD1, DMS3 and RDM1, but the assembly process of this complex and the underlying mechanism for Pol V recruitment remain unknown. Here we show that all DDR complex components co-localize with Pol V, and we report the cryoEM structures of two complexes associated with Pol V recruitment-DR (DMS3-RDM1) and DDR' (DMS3-RDM1-DRD1 peptide), at 3.6 Å and 3.5 Å resolution, respectively. RDM1 dimerization at the center frames the assembly of the entire complex and mediates interactions between DMS3 and DRD1 with a stoichiometry of 1 DRD1:4 DMS3:2 RDM1. DRD1 binding to the DR complex induces a drastic movement of a DMS3 coiled-coil helix bundle. We hypothesize that both complexes are functional intermediates that mediate Pol V recruitment.


Subject(s)
Arabidopsis Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , RNA, Plant/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/ultrastructure , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/ultrastructure , Cryoelectron Microscopy , DNA, Plant/genetics , DNA, Plant/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/ultrastructure , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/ultrastructure , Gene Expression Regulation, Plant , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Binding , Protein Conformation , RNA, Plant/chemistry , RNA, Plant/genetics
18.
Nucleic Acids Res ; 47(17): 9423-9432, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31392983

ABSTRACT

Bacteriophage T4 middle promoters are activated through a process called σ appropriation, which requires the concerted effort of two T4-encoded transcription factors: AsiA and MotA. Despite extensive biochemical and genetic analyses, puzzle remains, in part, because of a lack of precise structural information for σ appropriation complex. Here, we report a single-particle cryo-electron microscopy (cryo-EM) structure of an intact σ appropriation complex, comprising AsiA, MotA, Escherichia coli RNA polymerase (RNAP), σ70 and a T4 middle promoter. As expected, AsiA binds to and remodels σ region 4 to prevent its contact with host promoters. Unexpectedly, AsiA undergoes a large conformational change, takes over the job of σ region 4 and provides an anchor point for the upstream double-stranded DNA. Because σ region 4 is conserved among bacteria, other transcription factors may use the same strategy to alter the landscape of transcription immediately. Together, the structure provides a foundation for understanding σ appropriation and transcription activation.


Subject(s)
DNA-Binding Proteins/ultrastructure , DNA-Directed RNA Polymerases/ultrastructure , Transcription Factors/ultrastructure , Viral Proteins/ultrastructure , Bacteriophage T4/chemistry , Bacteriophage T4/genetics , Bacteriophage T4/ultrastructure , DNA , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Escherichia coli/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , Promoter Regions, Genetic/genetics , Protein Conformation , Transcription Factors/chemistry , Transcription Factors/genetics , Viral Proteins/genetics
19.
Nat Commun ; 10(1): 3048, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31296855

ABSTRACT

Bacteriophages typically hijack the host bacterial transcriptional machinery to regulate their own gene expression and that of the host bacteria. The structural basis for bacteriophage protein-mediated transcription regulation-in particular transcription antitermination-is largely unknown. Here we report the 3.4 Å and 4.0 Å cryo-EM structures of two bacterial transcription elongation complexes (P7-NusA-TEC and P7-TEC) comprising the bacteriophage protein P7, a master host-transcription regulator encoded by bacteriophage Xp10 of the rice pathogen Xanthomonas oryzae pv. Oryzae (Xoo) and discuss the mechanisms by which P7 modulates the host bacterial RNAP. The structures together with biochemical evidence demonstrate that P7 prevents transcription termination by plugging up the RNAP RNA-exit channel and impeding RNA-hairpin formation at the intrinsic terminator. Moreover, P7 inhibits transcription initiation by restraining RNAP-clamp motions. Our study reveals the structural basis for transcription antitermination by phage proteins and provides insights into bacterial transcription regulation.


Subject(s)
Bacterial Proteins/metabolism , Bacteriophages/genetics , Transcriptional Elongation Factors/metabolism , Viral Proteins/metabolism , Xanthomonas/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/ultrastructure , Cryoelectron Microscopy , DNA-Directed RNA Polymerases/isolation & purification , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/ultrastructure , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Viral , Host Microbial Interactions/genetics , Oryza/microbiology , Protein Structure, Secondary , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Terminator Regions, Genetic/genetics , Transcription, Genetic , Transcriptional Elongation Factors/isolation & purification , Transcriptional Elongation Factors/ultrastructure , Viral Proteins/isolation & purification , Viral Proteins/ultrastructure , Xanthomonas/virology
20.
Nat Commun ; 10(1): 2342, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138817

ABSTRACT

Recent history is punctuated by the emergence of highly pathogenic coronaviruses such as SARS- and MERS-CoV into human circulation. Upon infecting host cells, coronaviruses assemble a multi-subunit RNA-synthesis complex of viral non-structural proteins (nsp) responsible for the replication and transcription of the viral genome. Here, we present the 3.1 Å resolution structure of the SARS-CoV nsp12 polymerase bound to its essential co-factors, nsp7 and nsp8, using single particle cryo-electron microscopy. nsp12 possesses an architecture common to all viral polymerases as well as a large N-terminal extension containing a kinase-like fold and is bound by two nsp8 co-factors. This structure illuminates the assembly of the coronavirus core RNA-synthesis machinery, provides key insights into nsp12 polymerase catalysis and fidelity and acts as a template for the design of novel antiviral therapeutics.


Subject(s)
Coenzymes/ultrastructure , DNA-Directed RNA Polymerases/ultrastructure , Severe acute respiratory syndrome-related coronavirus/ultrastructure , Viral Nonstructural Proteins/ultrastructure , Cryoelectron Microscopy , Genome, Viral , Severe acute respiratory syndrome-related coronavirus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...