Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
PLoS Genet ; 17(10): e1009878, 2021 10.
Article in English | MEDLINE | ID: mdl-34710092

ABSTRACT

In bacteria, small non-coding RNAs (sRNAs) bind to target mRNAs and regulate their translation and/or stability. In the polycistronic galETKM operon of Escherichia coli, binding of the Spot 42 sRNA to the operon transcript leads to the generation of galET mRNA. The mechanism of this regulation has remained unclear. We show that sRNA-mRNA base pairing at the beginning of the galK gene leads to both transcription termination and transcript cleavage within galK, and generates galET mRNAs with two different 3'-OH ends. Transcription termination requires Rho, and transcript cleavage requires the endonuclease RNase E. The sRNA-mRNA base-paired segments required for generating the two galET species are different, indicating different sequence requirements for the two events. The use of two targets in an mRNA, each of which causes a different outcome, appears to be a novel mode of action for a sRNA. Considering the prevalence of potential sRNA targets at cistron junctions, the generation of new mRNA species by the mechanisms reported here might be a widespread mode of bacterial gene regulation.


Subject(s)
Endoribonucleases/genetics , Escherichia coli/genetics , RNA, Messenger/genetics , RNA, Small Untranslated/genetics , Transcription Termination, Genetic/physiology , Transcription, Genetic/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/genetics , Operon/genetics , RNA, Bacterial/genetics
2.
Mol Cell ; 81(17): 3589-3603.e13, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34324863

ABSTRACT

Transcription elongation has emerged as a regulatory hub in gene expression of metazoans. A major control point occurs during early elongation before RNA polymerase II (Pol II) is released into productive elongation. Prior research has linked BRD4 with transcription elongation. Here, we use rapid BET protein and BRD4-selective degradation along with quantitative genome-wide approaches to investigate direct functions of BRD4 in Pol II transcription regulation. Notably, as an immediate consequence of acute BRD4 loss, promoter-proximal pause release is impaired, and transcriptionally engaged Pol II past this checkpoint undergoes readthrough transcription. An integrated proteome-wide analysis uncovers elongation and 3'-RNA processing factors as core BRD4 interactors. BRD4 ablation disrupts the recruitment of general 3'-RNA processing factors at the 5'-control region, which correlates with RNA cleavage and termination defects. These studies, performed in human cells, reveal a BRD4-mediated checkpoint and begin to establish a molecular link between 5'-elongation control and 3'-RNA processing.


Subject(s)
Cell Cycle Proteins/physiology , Nuclear Proteins/physiology , Transcription Elongation, Genetic/physiology , Transcription Factors/physiology , Animals , Cell Cycle Proteins/metabolism , Cell Line , Gene Expression , Histones/metabolism , Humans , Mice , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/physiology , Transcription Factors/metabolism , Transcription Termination, Genetic/physiology , Transcription, Genetic/genetics , Transcription, Genetic/physiology
3.
Cell Rep ; 33(4): 108319, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33113359

ABSTRACT

Many RNA polymerases terminate transcription using allosteric/intrinsic mechanisms, whereby protein alterations or nucleotide sequences promote their release from DNA. RNA polymerase II (Pol II) is somewhat different based on its behavior at protein-coding genes where termination additionally requires endoribonucleolytic cleavage and subsequent 5'→3' exoribonuclease activity. The Pol-II-transcribed small nuclear RNAs (snRNAs) also undergo endoribonucleolytic cleavage by the Integrator complex, which promotes their transcriptional termination. Here, we confirm the involvement of Integrator but show that Integrator-independent processes can terminate snRNA transcription both in its absence and naturally. This is often associated with exosome degradation of snRNA precursors that long-read sequencing analysis reveals as frequently terminating at T-runs located downstream of some snRNAs. This finding suggests a unifying vulnerability of RNA polymerases to such sequences given their well-known roles in terminating Pol III and bacterial RNA polymerase.


Subject(s)
RNA Polymerase II/metabolism , RNA, Small Nuclear/metabolism , Transcription Termination, Genetic/physiology , Humans
4.
Life Sci Alliance ; 3(10)2020 10.
Article in English | MEDLINE | ID: mdl-32747416

ABSTRACT

DDX5, XRN2, and PRMT5 have been shown to resolve DNA/RNA hybrids (R-loops) at RNA polymerase II transcription termination sites at few genomic loci. Herein, we perform genome-wide R-loop mapping using classical DNA/RNA immunoprecipitation and high-throughput sequencing (DRIP-seq) of loci regulated by DDX5, XRN2, and PRMT5. We observed hundreds to thousands of R-loop gains and losses at transcribed loci in DDX5-, XRN2-, and PRMT5-deficient U2OS cells. R-loop gains were characteristic of highly transcribed genes located at gene-rich regions, whereas R-loop losses were observed in low-density gene areas. DDX5, XRN2, and PRMT5 shared many R-loop gain loci at transcription termination sites, consistent with their coordinated role in RNA polymerase II transcription termination. DDX5-depleted cells had unique R-loop gain peaks near the transcription start site that did not overlap with those of siXRN2 and siPRMT5 cells, suggesting a role for DDX5 in transcription initiation independent of XRN2 and PRMT5. Moreover, we observed that the accumulated R-loops at certain loci in siDDX5, siXRN2, and siPRMT5 cells near the transcription start site of genes led to antisense intergenic transcription. Our findings define unique and shared roles of DDX5, XRN2, and PRMT5 in DNA/RNA hybrid regulation.


Subject(s)
DEAD-box RNA Helicases/metabolism , Exoribonucleases/metabolism , Protein-Arginine N-Methyltransferases/metabolism , R-Loop Structures/genetics , Cell Line , DEAD-box RNA Helicases/genetics , DNA/genetics , Exoribonucleases/genetics , Genomics/methods , Humans , Immunoprecipitation/methods , Nucleic Acid Hybridization/genetics , Protein-Arginine N-Methyltransferases/genetics , R-Loop Structures/physiology , RNA/genetics , RNA Polymerase II/genetics , Transcription Termination, Genetic/physiology , Transcription, Genetic/genetics
5.
RNA ; 26(10): 1334-1344, 2020 10.
Article in English | MEDLINE | ID: mdl-32546512

ABSTRACT

Fission yeast Erh1 exists in a complex with RNA-binding protein Mmi1. Deletion of erh1 up-regulates the phosphate homeostasis gene pho1, which is normally repressed by transcription in cis of a 5' flanking prt lncRNA. Here we present evidence that de-repression of pho1 by erh1Δ is achieved through precocious 3'-processing/termination of prt lncRNA synthesis, to wit: (i) erh1Δ does not affect the activity of the prt or pho1 promoters per se; (ii) de-repression by erh1Δ depends on CPF (cleavage and polyadenylation factor) subunits Ctf1, Dis2, Ssu72, Swd22, and Ppn1 and on termination factor Rhn1; (iii) de-repression requires synthesis by the Asp1 IPP kinase of inositol 1-pyrophosphates (1-IPPs); (iv) de-repression is effaced by mutating Thr4 of the RNA polymerase II CTD to alanine; and (v) erh1Δ exerts an additive effect on pho1 de-repression in combination with mutating CTD Ser7 to alanine and with deletion of the IPP pyrophosphatase Aps1. These findings point to Erh1 as an antagonist of lncRNA termination in the prt-pho1 axis. In contrast, in mmi1Δ cells there is a reduction in pho1 mRNA and increase in the formation of a prt-pho1 read-through transcript, consistent with Mmi1 being an agonist of prt termination. We envision that Erh1 acts as a brake on Mmi1's ability to promote CPF-dependent termination during prt lncRNA synthesis. Consistent with this idea, erh1Δ de-repression of pho1 was eliminated by mutating the Mmi1-binding sites in the prt lncRNA.


Subject(s)
Acid Phosphatase/genetics , Carrier Proteins/genetics , Gene Expression Regulation, Fungal/genetics , RNA, Long Noncoding/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Transcription Termination, Genetic/physiology , Inositol Phosphates/genetics , Promoter Regions, Genetic/genetics , RNA Polymerase II/genetics , RNA, Messenger/genetics
6.
Nat Commun ; 11(1): 1063, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32102997

ABSTRACT

Mediator is a coregulatory complex that regulates transcription of Pol II-dependent genes. Previously, we showed that human Mediator subunit MED26 plays a role in the recruitment of Super Elongation Complex (SEC) or Little Elongation Complex (LEC) to regulate the expression of certain genes. MED26 plays a role in recruiting SEC to protein-coding genes including c-myc and LEC to small nuclear RNA (snRNA) genes. However, how MED26 engages SEC or LEC to regulate distinct genes is unclear. Here, we provide evidence that MED26 recruits LEC to modulate transcription termination of non-polyadenylated transcripts including snRNAs and mRNAs encoding replication-dependent histone (RDH) at Cajal bodies. Our findings indicate that LEC recruited by MED26 promotes efficient transcription termination by Pol II through interaction with CBC-ARS2 and NELF/DSIF, and promotes 3' end processing by enhancing recruitment of Integrator or Heat Labile Factor to snRNA or RDH genes, respectively.


Subject(s)
Gene Expression Regulation/genetics , Mediator Complex/genetics , RNA, Small Nuclear/genetics , Transcription Termination, Genetic/physiology , Transcriptional Elongation Factors/genetics , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Nuclear Proteins/metabolism , RNA Cap-Binding Proteins/metabolism , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism
7.
Br J Radiol ; 93(1115): 20190966, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-31944860

ABSTRACT

The significance of canonical DNA non-homologous end-joining (c-NHEJ) for DNA double strand break (DSB) repair has increased from lower organisms to higher eukaryotes, and plays the predominant role in human cells. Ku, the c-NHEJ end-binding component, binds DSBs with high efficiency enabling c-NHEJ to be the first choice DSB repair pathway, although alternative pathways can ensue after regulated steps to remove Ku. Indeed, radiation-induced DSBs are repaired rapidly in human cells. However, an important question is the fidelity with which radiation-induced DSBs are repaired, which is essential for assessing any harmful impacts caused by radiation exposure. Indeed, is compromised fidelity a price we pay for high capacity repair. Two subpathways of c-NHEJ have been revealed; a fast process that does not require nucleases or significant chromatin changes and a slower process that necessitates resection factors, and potentially more significant chromatin changes at the DSB. Recent studies have also shown that DSBs within transcriptionally active regions are repaired by specialised mechanisms, and the response at such DSBs encompasses a process of transcriptional arrest. Here, we consider the limitations of c-NHEJ that might result in DSB misrepair. We consider the common IR-induced misrepair events and discuss how they might arise via the distinct subpathways of c-NHEJ.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair/physiology , DNA Mismatch Repair/physiology , Chromatin/physiology , DNA/radiation effects , G1 Phase/genetics , G2 Phase/genetics , Humans , Ku Autoantigen/physiology , Resting Phase, Cell Cycle/genetics , Transcription Termination, Genetic/physiology , Transcriptional Activation/physiology
8.
Genes Dev ; 34(1-2): 132-145, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31805520

ABSTRACT

The allosteric and torpedo models have been used for 30 yr to explain how transcription terminates on protein-coding genes. The former invokes termination via conformational changes in the transcription complex and the latter proposes that degradation of the downstream product of poly(A) signal (PAS) processing is important. Here, we describe a single mechanism incorporating features of both models. We show that termination is completely abolished by rapid elimination of CPSF73, which causes very extensive transcriptional readthrough genome-wide. This is because CPSF73 functions upstream of modifications to the elongation complex and provides an entry site for the XRN2 torpedo. Rapid depletion of XRN2 enriches these events that we show are underpinned by protein phosphatase 1 (PP1) activity, the inhibition of which extends readthrough in the absence of XRN2. Our results suggest a combined allosteric/torpedo mechanism, in which PP1-dependent slowing down of polymerases over termination regions facilitates their pursuit/capture by XRN2 following PAS processing.


Subject(s)
Cleavage And Polyadenylation Specificity Factor/metabolism , Transcription Termination, Genetic/physiology , Cell Line , Cleavage And Polyadenylation Specificity Factor/genetics , Exoribonucleases/metabolism , Gene Deletion , HCT116 Cells , Humans , RNA/metabolism , RNA Polymerase II/metabolism , Receptors, Neuropeptide Y/metabolism , Ribonuclease H/metabolism
9.
Nat Commun ; 10(1): 3728, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31427601

ABSTRACT

Discovery of CRISPR-Cas systems is one of paramount importance in the field of microbiology. Currently, how CRISPR-Cas systems are finely regulated remains to be defined. Here we use small regulatory RNA (sRNA) library to screen sRNAs targeting type I-F CRISPR-Cas system through proximity ligation by T4 RNA ligase and find 34 sRNAs linking to CRISPR loci. Among 34 sRNAs for potential regulators of CRISPR, sRNA pant463 and PhrS enhance CRISPR loci transcription, while pant391 represses their transcription. We identify PhrS as a regulator of CRISPR-Cas by binding CRISPR leaders to suppress Rho-dependent transcription termination. PhrS-mediated anti-termination facilitates CRISPR locus transcription to generate CRISPR RNA (crRNA) and subsequently promotes CRISPR-Cas adaptive immunity against bacteriophage invasion. Furthermore, this also exists in type I-C/-E CRISPR-Cas, suggesting general regulatory mechanisms in bacteria kingdom. Our findings identify sRNAs as important regulators of CRISPR-Cas, extending roles of sRNAs in controlling bacterial physiology by promoting CRISPR-Cas adaptation priming.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Escherichia coli/genetics , Pseudomonas aeruginosa/genetics , RNA, Bacterial/biosynthesis , RNA, Small Untranslated/genetics , Rho Factor/antagonists & inhibitors , Transcription Termination, Genetic/physiology , Bacteriophages/genetics , CRISPR-Cas Systems/genetics , High-Throughput Screening Assays , RNA, Bacterial/genetics
10.
PLoS Genet ; 15(5): e1008141, 2019 05.
Article in English | MEDLINE | ID: mdl-31120886

ABSTRACT

Stop-codon read-through refers to the phenomenon that a ribosome goes past the stop codon and continues translating into the otherwise untranslated region (UTR) of a transcript. Recent ribosome-profiling experiments in eukaryotes uncovered widespread stop-codon read-through that also varies among tissues, prompting the adaptive hypothesis that stop-codon read-through is an important, regulated mechanism for generating proteome diversity. Here we propose and test a competing hypothesis that stop-codon read-through arises mostly from molecular errors and is largely nonadaptive. The error hypothesis makes distinct predictions about the probability of read-through, frequency of sequence motifs for read-through, and conservation of the read-through region, each of which is supported by genome-scale data from yeasts and fruit flies. Thus, except for the few cases with demonstrated functions, stop-codon read-through is generally nonadaptive. This finding, along with other molecular errors recently quantified, reveals a much less precise or orderly cellular life than is commonly thought.


Subject(s)
Codon, Terminator/genetics , Transcription Termination, Genetic/physiology , Transcription, Genetic/genetics , 3' Untranslated Regions , Animals , Drosophila/genetics , Evolution, Molecular , Ribosomes , Yeasts/genetics
11.
Nat Commun ; 10(1): 1545, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30948716

ABSTRACT

Extrinsic transcription termination typically involves remodeling of RNA polymerase by an accessory helicase. In yeast this is accomplished by the Sen1 helicase homologous to human senataxin (SETX). To gain insight into these processes we develop a DNA scaffold construct compatible with magnetic-trapping assays and from which S. cerevisiae RNA polymerase II (Pol II), as well as E. coli RNA polymerase (ecRNAP), can efficiently initiate transcription without transcription factors, elongate, and undergo extrinsic termination. By stalling Pol II TECs on the construct we can monitor Sen1-induced termination in real-time, revealing the formation of an intermediate in which the Pol II transcription bubble appears half-rewound. This intermediate requires ~40 sec to form and lasts ~20 sec prior to final dissociation of the stalled Pol II. The experiments enabled by the scaffold construct permit detailed statistical and kinetic analysis of Pol II interactions with a range of cofactors in a multi-round, high-throughput fashion.


Subject(s)
DNA Helicases/physiology , Escherichia coli/genetics , RNA Helicases/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Transcription Termination, Genetic/physiology , Transcription, Genetic , DNA Helicases/genetics , DNA Helicases/metabolism , Escherichia coli/metabolism , Models, Molecular , RNA Helicases/genetics , RNA Helicases/metabolism , RNA Polymerase II/metabolism , RNA Polymerase II/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
12.
Nat Commun ; 10(1): 1207, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872584

ABSTRACT

In bacteria, transcription-coupled repair of DNA lesions initiates after the Mfd protein removes RNA polymerases (RNAPs) stalled at the lesions. The bacterial RNA helicase, Rho, is a transcription termination protein that dislodges the elongation complexes. Here, we show that Rho dislodges the stalled RNAPs at DNA lesions. Strains defective in both Rho and Mfd are susceptible to DNA-damaging agents and are inefficient in repairing or propagating UV-damaged DNA. In vitro transcription assays show that Rho dissociates the stalled elongation complexes at the DNA lesions. We conclude that Rho-dependent termination recycles stalled RNAPs, which might facilitate DNA repair and other DNA-dependent processes essential for bacterial cell survival. We surmise that Rho might compete with, or augment, the Mfd function.


Subject(s)
DNA Repair/physiology , DNA-Directed RNA Polymerases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Transcription Termination, Genetic/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA Damage/drug effects , DNA Damage/radiation effects , DNA, Bacterial/metabolism , Escherichia coli Proteins/genetics , Mitomycin/pharmacology , Mutation , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , RNA, Bacterial/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Ultraviolet Rays/adverse effects
13.
PLoS Genet ; 15(2): e1007646, 2019 02.
Article in English | MEDLINE | ID: mdl-30742606

ABSTRACT

Putrescine belongs to the large group of polyamines, an essential class of metabolites that exists throughout all kingdoms of life. The Salmonella speF gene encodes an inducible ornithine decarboxylase that produces putrescine from ornithine. Putrescine can be also synthesized from arginine in a parallel metabolic pathway. Here, we show that speF expression is controlled at multiple levels through regulatory elements contained in a long leader sequence. At the heart of this regulation is a short open reading frame, orf34, which is required for speF production. Translation of orf34 interferes with Rho-dependent transcription termination and helps to unfold an inhibitory RNA structure sequestering speF ribosome-binding site. Two consecutive arginine codons in the conserved domain of orf34 provide a third level of speF regulation. Uninterrupted translation of orf34 under conditions of high arginine allows the formation of a speF mRNA structure that is degraded by RNase G, whereas ribosome pausing at the consecutive arginine codons in the absence of arginine enables the formation of an alternative structure that is resistant to RNase G. Thus, the rate of ribosome progression during translation of the upstream ORF influences the dynamics of speF mRNA folding and putrescine production. The identification of orf34 and its regulatory functions provides evidence for the evolutionary conservation of ornithine decarboxylase regulatory elements and putrescine production.


Subject(s)
Arginine/metabolism , Polyamines/metabolism , RNA, Messenger/genetics , Salmonella/genetics , Salmonella/metabolism , Base Sequence , Open Reading Frames/genetics , Ornithine/metabolism , Ornithine Decarboxylase/metabolism , Protein Biosynthesis/genetics , Protein Domains/physiology , Transcription Termination, Genetic/physiology
14.
Cell Rep ; 25(1): 259-269.e5, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30282034

ABSTRACT

Termination of RNA polymerase II (Pol II) transcription is a key step that is important for 3' end formation of functional mRNA, mRNA release, and Pol II recycling. Even so, the underlying termination mechanism is not yet understood. Here, we demonstrate that the conserved and essential termination factor Seb1 is found on Pol II near the end of the RNA exit channel and the Rpb4/7 stalk. Furthermore, the Seb1 interaction surface with Pol II largely overlaps with that of the elongation factor Spt5. Notably, Seb1 co-transcriptional recruitment is dependent on Spt5 dephosphorylation by the conserved PP1 phosphatase Dis2, which also dephosphorylates threonine 4 within the Pol II heptad repeated C-terminal domain. We propose that Dis2 orchestrates the transition from elongation to termination phase during the transcription cycle by mediating elongation to termination factor exchange and dephosphorylation of Pol II C-terminal domain.


Subject(s)
Peptide Elongation Factors/genetics , RNA Polymerase II/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Transcription Factors/genetics , Transcription Termination, Genetic/physiology , Transcription, Genetic/genetics
15.
Nat Commun ; 9(1): 4364, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341288

ABSTRACT

Termination of RNA polymerase II (RNAPII) transcription is a fundamental step of gene expression that is critical for determining the borders between genes. In budding yeast, termination at protein-coding genes is initiated by the cleavage/polyadenylation machinery, whereas termination of most noncoding RNA (ncRNA) genes occurs via the Nrd1-Nab3-Sen1 (NNS) pathway. Here, we find that NNS-like transcription termination is not conserved in fission yeast. Rather, genome-wide analyses show global recruitment of mRNA 3' end processing factors at the end of ncRNA genes, including snoRNAs and snRNAs, and that this recruitment coincides with high levels of Ser2 and Tyr1 phosphorylation on the RNAPII C-terminal domain. We also find that termination of mRNA and ncRNA transcription requires the conserved Ysh1/CPSF-73 and Dhp1/XRN2 nucleases, supporting widespread cleavage-dependent transcription termination in fission yeast. Our findings thus reveal that a common mode of transcription termination can produce functionally and structurally distinct types of polyadenylated and non-polyadenylated RNAs.


Subject(s)
RNA/genetics , Schizosaccharomyces/genetics , Transcription Termination, Genetic/physiology , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Helicases/physiology , RNA Helicases/genetics , RNA Helicases/metabolism , RNA Helicases/physiology , RNA Polymerase II/metabolism , RNA Polymerase II/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , Species Specificity
16.
Microbiol Spectr ; 6(5)2018 09.
Article in English | MEDLINE | ID: mdl-30191803

ABSTRACT

Transcription is a discontinuous process, where each nucleotide incorporation cycle offers a decision between elongation, pausing, halting, or termination. Many cis-acting regulatory RNAs, such as riboswitches, exert their influence over transcription elongation. Through such mechanisms, certain RNA elements can couple physiological or environmental signals to transcription attenuation, a process where cis-acting regulatory RNAs directly influence formation of transcription termination signals. However, through another regulatory mechanism called processive antitermination (PA), RNA polymerase can bypass termination sites over much greater distances than transcription attenuation. PA mechanisms are widespread in bacteria, although only a few classes have been discovered overall. Also, although traditional, signal-responsive riboswitches have not yet been discovered to promote PA, it is increasingly clear that small RNA elements are still oftentimes required. In some instances, small RNA elements serve as loading sites for cellular factors that promote PA. In other instances, larger, more complicated RNA elements participate in PA in unknown ways, perhaps even acting alone to trigger PA activity. These discoveries suggest that what is now needed is a systematic exploration of PA in bacteria, to determine how broadly these transcription elongation mechanisms are utilized, to reveal the diversity in their molecular mechanisms, and to understand the general logic behind their cellular applications. This review covers the known examples of PA regulatory mechanisms and speculates that they may be broadly important to bacteria.


Subject(s)
Bacterial Proteins/physiology , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA-Binding Proteins/physiology , Transcription Factors/metabolism , Transcription, Genetic , Bacterial Proteins/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli Proteins , Nucleotides/metabolism , Operon , Peptide Elongation Factors , RNA-Binding Proteins/genetics , Trans-Activators , Transcription Termination, Genetic/physiology
17.
Mol Cell ; 71(6): 911-922.e4, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30122535

ABSTRACT

NusG/Spt5 proteins are the only transcription factors utilized by all cellular organisms. In enterobacteria, NusG antagonizes the transcription termination activity of Rho, a hexameric helicase, during the synthesis of ribosomal and actively translated mRNAs. Paradoxically, NusG helps Rho act on untranslated transcripts, including non-canonical antisense RNAs and those arising from translational stress; how NusG fulfills these disparate functions is unknown. Here, we demonstrate that NusG activates Rho by assisting helicase isomerization from an open-ring, RNA-loading state to a closed-ring, catalytically active translocase. A crystal structure of closed-ring Rho in complex with NusG reveals the physical basis for this activation and further explains how Rho is excluded from translationally competent RNAs. This study demonstrates how a universally conserved transcription factor acts to modulate the activity of a ring-shaped ATPase motor and establishes how the innate sequence bias of a termination factor can be modulated to silence pervasive, aberrant transcription.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , Escherichia coli Proteins/physiology , Peptide Elongation Factors/physiology , Transcription Factors/physiology , Transcription Termination, Genetic/physiology , Transcriptional Elongation Factors/physiology , Bacterial Proteins , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Models, Molecular , Peptide Elongation Factors/metabolism , Protein Conformation , RNA, Bacterial , Rho Factor/metabolism , Rho Factor/physiology , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcription, Genetic/physiology
18.
Mol Cell ; 69(1): 48-61.e6, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29304333

ABSTRACT

The carboxy-terminal domain (CTD) of RNA polymerase (Pol) II is composed of a repetition of YSPTSPS heptads and functions as a loading platform for protein complexes that regulate transcription, splicing, and maturation of RNAs. Here, we studied mammalian CTD mutants to analyze the function of tyrosine1 residues in the transcription cycle. Mutation of 3/4 of the tyrosine residues (YFFF mutant) resulted in a massive read-through transcription phenotype in the antisense direction of promoters as well as in the 3' direction several hundred kilobases downstream of genes. The YFFF mutant shows reduced Pol II at promoter-proximal pause sites, a loss of interaction with the Mediator and Integrator complexes, and impaired recruitment of these complexes to chromatin. Consistent with these observations, Pol II loading at enhancers and maturation of snRNAs are altered in the YFFF context genome-wide. We conclude that tyrosine1 residues of the CTD control termination of transcription by Pol II.


Subject(s)
RNA Polymerase II/genetics , RNA, Messenger/biosynthesis , Transcription Termination, Genetic/physiology , Transcription, Genetic/physiology , Tyrosine/genetics , Cell Line, Tumor , Chromatin/metabolism , Humans , Mutation/genetics , Promoter Regions, Genetic/genetics , RNA Polymerase II/metabolism , RNA, Small Nuclear/genetics
19.
Gene ; 641: 292-296, 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-29111204

ABSTRACT

OBJECTIVE: The association between mutations in the USP26 gene and male infertility has been studied intensively. However, the biological function of the mutant proteins remains to be elucidated. To confirm the effects of the reported mutations, we analyse the enzyme activity of USP26 between the wild-type and the variants from a molecular perspective. METHODS: Using pGEX-USP26 as a template, site-directed mutagenesis was conducted to generate nineteen USP26 mutant plasmids. Using Ub-Met-ß-gal and GST-Ub52 as model substrates, a USP cleavage assay was conducted to assess the enzymatic activities of the mutants. RESULTS: The enzyme activity of the Q156H mutant disappeared, but the other 18 mutants had the same activity as the wild type. E174# and E189# were terminal mutants, but they still had the same activity as the wild type. When we constructed the transcription terminal mutants E174#(1-522 bp), E174#(523-2742 bp), E189#(1-567 bp) and E189#(568-2742 bp) artificially, the enzyme activity of these four mutants disappeared. CONCLUSIONS: We have successfully constructed nineteen mutants of USP26. The enzyme activity of the Q156H mutant disappeared, but the enzyme activities of the other 18 mutants were the same as that of the wild type.


Subject(s)
Cysteine Endopeptidases/genetics , Mutation/genetics , Escherichia coli/genetics , Mutagenesis, Site-Directed/methods , Mutant Proteins/genetics , Plasmids/genetics , Recombinant Fusion Proteins/genetics , Transcription Termination, Genetic/physiology
20.
Genes Dev ; 31(18): 1870-1879, 2017 09 15.
Article in English | MEDLINE | ID: mdl-29021241

ABSTRACT

Transcription termination determines the ends of transcriptional units and thereby ensures the integrity of the transcriptome and faithful gene regulation. Studies in yeast and human cells have identified the exoribonuclease XRN2 as a key termination factor for protein-coding genes. Here we performed a genome-wide investigation of RNA polymerase II (Pol II) transcription termination in XRN2-deficient Caenorhabditis elegans and observed two distinct modes of termination. Although a subset of genes requires XRN2, termination of other genes appears both independent of, and refractory to, XRN2. XRN2 independence is not merely a consequence of failure to recruit XRN2, since XRN2 is present on-and promotes Pol II accumulation near the polyadenylation sites of-both gene classes. Unexpectedly, promoters instruct the choice of termination mode, but XRN2-independent termination additionally requires a compatible region downstream from the 3' end cleavage site. Hence, different termination mechanisms may work with different configurations of Pol II complexes dictated by promoters.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Exoribonucleases/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Transcription Termination, Genetic/physiology , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Exoribonucleases/genetics , RNA Interference , RNA Polymerase II/genetics , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...