Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Nature ; 626(8000): 891-896, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38326611

ABSTRACT

Transcription elongation stalls at lesions in the DNA template1. For the DNA lesion to be repaired, the stalled transcription elongation complex (EC) has to be removed from the damaged site2. Here we show that translation, which is coupled to transcription in bacteria, actively dislodges stalled ECs from the damaged DNA template. By contrast, paused, but otherwise elongation-competent, ECs are not dislodged by the ribosome. Instead, they are helped back into processive elongation. We also show that the ribosome slows down when approaching paused, but not stalled, ECs. Our results indicate that coupled ribosomes functionally and kinetically discriminate between paused ECs and stalled ECs, ensuring the selective destruction of only the latter. This functional discrimination is controlled by the RNA polymerase's catalytic domain, the Trigger Loop. We show that the transcription-coupled DNA repair helicase UvrD, proposed to cause backtracking of stalled ECs3, does not interfere with ribosome-mediated dislodging. By contrast, the transcription-coupled DNA repair translocase Mfd4 acts synergistically with translation, and dislodges stalled ECs that were not destroyed by the ribosome. We also show that a coupled ribosome efficiently destroys misincorporated ECs that can cause conflicts with replication5. We propose that coupling to translation is an ancient and one of the main mechanisms of clearing non-functional ECs from the genome.


Subject(s)
DNA-Directed RNA Polymerases , Escherichia coli , Protein Biosynthesis , Transcription, Genetic , Catalytic Domain , DNA Helicases/metabolism , DNA Repair , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Kinetics , Ribosomes/metabolism , Templates, Genetic , Transcription Elongation, Genetic , Genome, Bacterial
2.
Mol Cell ; 72(2): 263-274.e5, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30244835

ABSTRACT

Antibiotic-resistant bacterial pathogens pose an urgent healthcare threat, prompting a demand for new medicines. We report the mode of action of the natural ansamycin antibiotic kanglemycin A (KglA). KglA binds bacterial RNA polymerase at the rifampicin-binding pocket but maintains potency against RNA polymerases containing rifampicin-resistant mutations. KglA has antibiotic activity against rifampicin-resistant Gram-positive bacteria and multidrug-resistant Mycobacterium tuberculosis (MDR-M. tuberculosis). The X-ray crystal structures of KglA with the Escherichia coli RNA polymerase holoenzyme and Thermus thermophilus RNA polymerase-promoter complex reveal an altered-compared with rifampicin-conformation of KglA within the rifampicin-binding pocket. Unique deoxysugar and succinate ansa bridge substituents make additional contacts with a separate, hydrophobic pocket of RNA polymerase and preclude the formation of initial dinucleotides, respectively. Previous ansa-chain modifications in the rifamycin series have proven unsuccessful. Thus, KglA represents a key starting point for the development of a new class of ansa-chain derivatized ansamycins to tackle rifampicin resistance.


Subject(s)
Biological Products/pharmacology , Drug Resistance, Bacterial/drug effects , Mycobacterium tuberculosis/drug effects , Rifabutin/pharmacology , Rifampin/pharmacology , Rifamycins/pharmacology , Antitubercular Agents/pharmacology , DNA-Directed RNA Polymerases/genetics , Drug Resistance, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Humans , Microbial Sensitivity Tests/methods , Mutation/drug effects , Mutation/genetics , Mycobacterium tuberculosis/genetics , Thermus thermophilus/drug effects , Thermus thermophilus/genetics
3.
Mol Microbiol ; 120(4): 539-546, 2023 10.
Article in English | MEDLINE | ID: mdl-37856403

ABSTRACT

The flow of genetic information from the chromosome to protein in all living organisms consists of two steps: (1) copying information coded in DNA into an mRNA intermediate via transcription by RNA polymerase, followed by (2) translation of this mRNA into a polypeptide by the ribosome. Unlike eukaryotes, where transcription and translation are separated by a nuclear envelope, in bacterial cells, these two processes occur within the same compartment. This means that a pioneering ribosome starts translation on nascent mRNA that is still being actively transcribed by RNA polymerase. This tethering via mRNA is referred to as 'coupling' of transcription and translation (CTT). CTT raises many questions regarding physical interactions and potential mutual regulation between these large (ribosome is ~2.5 MDa and RNA polymerase is 0.5 MDa) and powerful molecular machines. Accordingly, we will discuss some recently discovered structural and functional aspects of CTT.


Subject(s)
Protein Biosynthesis , Transcription, Genetic , DNA-Directed RNA Polymerases/metabolism , Ribosomes/metabolism , RNA, Messenger/metabolism
4.
Nucleic Acids Res ; 49(15): 8777-8784, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34365509

ABSTRACT

Transcribing RNA polymerase (RNAP) can fall into backtracking, phenomenon when the 3' end of the transcript disengages from the template DNA. Backtracking is caused by sequences of the nucleic acids or by misincorporation of erroneous nucleotides. To resume productive elongation backtracked complexes have to be resolved through hydrolysis of RNA. There is currently no consensus on the mechanism of catalysis of this reaction by Escherichia coli RNAP. Here we used Salinamide A, that we found inhibits RNAP catalytic domain Trigger Loop (TL), to show that the TL is required for RNA cleavage during proofreading of misincorporation events but plays little role during cleavage in sequence-dependent backtracked complexes. Results reveal that backtracking caused by misincorporation is distinct from sequence-dependent backtracking, resulting in different conformations of the 3' end of RNA within the active center. We show that the TL is required to transfer the 3' end of misincorporated transcript from cleavage-inefficient 'misincorporation site' into the cleavage-efficient 'backtracked site', where hydrolysis takes place via transcript-assisted catalysis and is largely independent of the TL. These findings resolve the controversy surrounding mechanism of RNA hydrolysis by E. coli RNA polymerase and indicate that the TL role in RNA cleavage has diverged among bacteria.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , RNA, Messenger/metabolism , Transcription Elongation, Genetic , Catalytic Domain , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/chemistry , Depsipeptides/chemistry , Depsipeptides/pharmacology , Escherichia coli/enzymology , Escherichia coli/genetics , Hydrolysis , RNA Cleavage
5.
Proc Natl Acad Sci U S A ; 117(15): 8462-8467, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32238560

ABSTRACT

In bacteria, the first two steps of gene expression-transcription and translation-are spatially and temporally coupled. Uncoupling may lead to the arrest of transcription through RNA polymerase backtracking, which interferes with replication forks, leading to DNA double-stranded breaks and genomic instability. How transcription-translation coupling mitigates these conflicts is unknown. Here we show that, unlike replication, translation is not inhibited by arrested transcription elongation complexes. Instead, the translating ribosome actively pushes RNA polymerase out of the backtracked state, thereby reactivating transcription. We show that the distance between the two machineries upon their contact on mRNA is smaller than previously thought, suggesting intimate interactions between them. However, this does not lead to the formation of a stable functional complex between the enzymes, as was once proposed. Our results reveal an active, energy-driven mechanism that reactivates backtracked elongation complexes and thus helps suppress their interference with replication.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Escherichia coli/enzymology , RNA, Messenger/metabolism , Ribosomes/metabolism , Transcription, Genetic , Transcriptional Elongation Factors/metabolism , Base Sequence , DNA-Directed RNA Polymerases/genetics , Escherichia coli/genetics , RNA, Messenger/genetics , Ribosomes/genetics , Transcriptional Elongation Factors/genetics
6.
PLoS Pathog ; 16(7): e1008672, 2020 07.
Article in English | MEDLINE | ID: mdl-32706832

ABSTRACT

Most clinical MRSA (methicillin-resistant S. aureus) isolates exhibit low-level ß-lactam resistance (oxacillin MIC 2-4 µg/ml) due to the acquisition of a novel penicillin binding protein (PBP2A), encoded by mecA. However, strains can evolve high-level resistance (oxacillin MIC ≥256 µg/ml) by an unknown mechanism. Here we have developed a robust system to explore the basis of the evolution of high-level resistance by inserting mecA into the chromosome of the methicillin-sensitive S. aureus SH1000. Low-level mecA-dependent oxacillin resistance was associated with increased expression of anaerobic respiratory and fermentative genes. High-level resistant derivatives had acquired mutations in either rpoB (RNA polymerase subunit ß) or rpoC (RNA polymerase subunit ß') and these mutations were shown to be responsible for the observed resistance phenotype. Analysis of rpoB and rpoC mutants revealed decreased growth rates in the absence of antibiotic, and alterations to, transcription elongation. The rpoB and rpoC mutations resulted in decreased expression to parental levels, of anaerobic respiratory and fermentative genes and specific upregulation of 11 genes including mecA. There was however no direct correlation between resistance and the amount of PBP2A. A mutational analysis of the differentially expressed genes revealed that a member of the S. aureus Type VII secretion system is required for high level resistance. Interestingly, the genomes of two of the high level resistant evolved strains also contained missense mutations in this same locus. Finally, the set of genetically matched strains revealed that high level antibiotic resistance does not incur a significant fitness cost during pathogenesis. Our analysis demonstrates the complex interplay between antibiotic resistance mechanisms and core cell physiology, providing new insight into how such important resistance properties evolve.


Subject(s)
Bacterial Proteins/genetics , DNA-Directed RNA Polymerases/genetics , Gene Expression Regulation, Bacterial/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Penicillin-Binding Proteins/genetics , beta-Lactam Resistance/genetics , Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects
7.
Nucleic Acids Res ; 48(14): 7914-7923, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32652039

ABSTRACT

Bacterial RNA polymerase is a potent target for antibiotics, which utilize a plethora of different modes of action, some of which are still not fully understood. Ureidothiophene (Urd) was found in a screen of a library of chemical compounds for ability to inhibit bacterial transcription. The mechanism of Urd action is not known. Here, we show that Urd inhibits transcription at the early stage of closed complex formation by blocking interaction of RNA polymerase with the promoter -10 element, while not affecting interactions with -35 element or steps of transcription after promoter closed complex formation. We show that mutation in the region 1.2 of initiation factor σ decreases sensitivity to Urd. The results suggest that Urd may directly target σ region 1.2, which allosterically controls the recognition of -10 element by σ region 2. Alternatively, Urd may block conformational changes of the holoenzyme required for engagement with -10 promoter element, although by a mechanism distinct from that of antibiotic fidaxomycin (lipiarmycin). The results suggest a new mode of transcription inhibition involving the regulatory domain of σ subunit, and potentially pinpoint a novel target for development of new antibacterials.


Subject(s)
Anti-Bacterial Agents/pharmacology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Promoter Regions, Genetic , Thiophenes/pharmacology , Transcription Initiation, Genetic/drug effects , Anti-Bacterial Agents/chemistry , Bacteria/enzymology , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Sigma Factor/antagonists & inhibitors , Sigma Factor/chemistry , Thiophenes/chemistry
8.
Antimicrob Agents Chemother ; 65(12): e0086421, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34606341

ABSTRACT

Rifamycins, such as rifampicin (Rif), are potent inhibitors of bacterial RNA polymerase (RNAP) and are widely used antibiotics. Rifamycin resistance is usually associated with mutations in RNAP that preclude rifamycin binding. However, some bacteria have a type of ADP-ribosyl transferases, Arr, which ADP-ribosylate rifamycin molecules, thus inactivating their antimicrobial activity. Here, we directly show that ADP-ribosylation abolishes inhibition of transcription by rifampicin, the most widely used rifamycin antibiotic. We also show that a natural rifamycin, kanglemycin A (KglA), which has a unique sugar moiety at the ansa chain close to the Arr modification site, does not bind to Arr from Mycobacterium smegmatis and thus is not susceptible to inactivation. We, found, however, that kanglemycin A can still be ADP-ribosylated by the Arr of an emerging pathogen, Mycobacterium abscessus. Interestingly, the only part of Arr that exhibits no homology between the species is the part that sterically clashes with the sugar moiety of kanglemycin A in M. smegmatis Arr. This suggests that M. abscessus has encountered KglA or rifamycin with a similar sugar modification in the course of evolution. The results show that KglA could be an effective antimicrobial against some of the Arr-encoding bacteria.


Subject(s)
Rifamycins , ADP-Ribosylation , Microbial Sensitivity Tests , Rifampin/pharmacology , Rifamycins/pharmacology
9.
Mol Cell ; 52(2): 248-54, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24095282

ABSTRACT

HipA of Escherichia coli is a eukaryote-like serine-threonine kinase that inhibits cell growth and induces persistence (multidrug tolerance). Previously, it was proposed that HipA inhibits cell growth by the phosphorylation of the essential translation factor EF-Tu. Here, we provide evidence that EF-Tu is not a target of HipA. Instead, a genetic screen reveals that the overexpression of glutamyl-tRNA synthetase (GltX) suppresses the toxicity of HipA. We show that HipA phosphorylates conserved Ser(239) near the active center of GltX and inhibits aminoacylation, a unique example of an aminoacyl-tRNA synthetase being inhibited by a toxin encoded by a toxin-antitoxin locus. HipA only phosphorylates tRNA(Glu)-bound GltX, which is consistent with the earlier finding that the regulatory motif containing Ser(239) changes configuration upon tRNA binding. These results indicate that HipA mediates persistence by the generation of "hungry" codons at the ribosomal A site that trigger the synthesis of (p)ppGpp, a hypothesis that we verify experimentally.


Subject(s)
Drug Tolerance , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Protein Serine-Threonine Kinases/metabolism , Adenosine Triphosphate/metabolism , Aminoacylation , Anti-Bacterial Agents/pharmacology , Binding Sites/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Glutamate-tRNA Ligase/chemistry , Glutamate-tRNA Ligase/genetics , Glutamate-tRNA Ligase/metabolism , Guanosine Pentaphosphate/metabolism , Models, Genetic , Models, Molecular , Mutation , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factor Tu/metabolism , Phosphorylation , Protein Biosynthesis , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , RNA, Transfer, Glu/genetics , RNA, Transfer, Glu/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Serine/chemistry , Serine/genetics , Serine/metabolism
10.
Int J Mol Sci ; 22(24)2021 Dec 14.
Article in English | MEDLINE | ID: mdl-34948223

ABSTRACT

Insertions of transposable elements (TEs) in eukaryotic genomes are usually associated with repressive chromatin, which spreads to neighbouring genomic sequences. In ovaries of Drosophila melanogaster, the Piwi-piRNA pathway plays a key role in the transcriptional silencing of TEs considered to be exerted mostly through the establishment of H3K9me3 histone marks recruiting Heterochromatin Protein 1a (HP1a). Here, using RNA-seq, we investigated the expression of TEs and the adjacent genomic regions upon Piwi and HP1a germline knockdowns sharing a similar genetic background. We found that the depletion of Piwi and HP1a led to the derepression of only partially overlapping TE sets. Several TEs were silenced predominantly by HP1a, whereas the upregulation of some other TEs was more pronounced upon Piwi knockdown and, surprisingly, was diminished upon a Piwi/HP1a double-knockdown. We revealed that HP1a loss influenced the expression of thousands of protein-coding genes mostly not adjacent to TE insertions and, in particular, downregulated a putative transcriptional factor required for TE activation. Nevertheless, our results indicate that Piwi and HP1a cooperatively exert repressive effects on the transcription of euchromatic loci flanking the insertions of some Piwi-regulated TEs. We suggest that this mechanism controls the silencing of a small set of TE-adjacent tissue-specific genes, preventing their inappropriate expression in ovaries.


Subject(s)
Argonaute Proteins/metabolism , Chromobox Protein Homolog 5/metabolism , DNA Transposable Elements , Drosophila Proteins/metabolism , Germ Cells/metabolism , Ovary/metabolism , RNA-Seq , Animals , Argonaute Proteins/genetics , Chromobox Protein Homolog 5/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Female
11.
Nucleic Acids Res ; 45(3): 1105-1113, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28180286

ABSTRACT

The transcription error rate estimated from mistakes in end product RNAs is 10−3­10−5. We analyzed the fidelity of nascent RNAs from all actively transcribing elongation complexes (ECs) in Escherichia coli and Saccharomyces cerevisiae and found that 1­3% of all ECs in wild-type cells, and 5­7% of all ECs in cells lacking proofreading factors are, in fact, misincorporated complexes. With the exception of a number of sequence-dependent hotspots, most misincorporations are distributed relatively randomly. Misincorporation at hotspots does not appear to be stimulated by pausing. Since misincorporation leads to a strong pause of transcription due to backtracking, our findings indicate that misincorporation could be a major source of transcriptional pausing and lead to conflicts with other RNA polymerases and replication in bacteria and eukaryotes. This observation implies that physical resolution of misincorporated complexes may be the main function of the proofreading factors Gre and TFIIS. Although misincorporation mechanisms between bacteria and eukaryotes appear to be conserved, the results suggest the existence of a bacteria-specific mechanism(s) for reducing misincorporation in protein-coding regions. The links between transcription fidelity, human disease, and phenotypic variability in genetically-identical cells can be explained by the accumulation of misincorporated complexes, rather than mistakes in mature RNA.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Transcription, Genetic , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Models, Genetic , RNA/genetics , RNA/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcriptional Elongation Factors/metabolism
12.
Trends Biochem Sci ; 39(3): 121-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24507752

ABSTRACT

Fic enzymes post-translationally modify proteins through AMPylation, UMPylation, phosphorylation, or phosphocholination. They have been identified across all domains of life, and they target a myriad of proteins such as eukaryotic GTPases, unstructured protein segments, and bacterial enzymes. Consequently, they play crucial roles in eukaryotic signal transduction, drug tolerance, bacterial pathogenicity, and the bacterial stress response. Structurally, they consist of an all α-helical core domain that supports and scaffolds a structurally conserved active-site loop, which catalyses the transfer of various parts of a nucleotide cofactor to proteins. Despite their diverse substrates and targets, they retain a conserved active site and reaction chemistry. This catalytic variety came to light only recently with the crystal structures of different Fic enzymes.


Subject(s)
Bacteria , Bacterial Proteins , GTP Phosphohydrolases , Protein Modification, Translational/physiology , Transferases , Animals , Bacteria/enzymology , Bacteria/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Protein Structure, Secondary , Protein Structure, Tertiary , Transferases/chemistry , Transferases/genetics , Transferases/metabolism
13.
Methods ; 120: 76-84, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28434904

ABSTRACT

The identification of the protein-coding regions of a genome is straightforward due to the universality of start and stop codons. However, the boundaries of the transcribed regions, conditional operon structures, non-coding RNAs and the dynamics of transcription, such as pausing of elongation, are non-trivial to identify, even in the comparatively simple genomes of prokaryotes. Traditional methods for the study of these areas, such as tiling arrays, are noisy, labour-intensive and lack the resolution required for densely-packed bacterial genomes. Recently, deep sequencing has become increasingly popular for the study of the transcriptome due to its lower costs, higher accuracy and single nucleotide resolution. These methods have revolutionised our understanding of prokaryotic transcriptional dynamics. Here, we review the deep sequencing and data analysis techniques that are available for the study of transcription in prokaryotes, and discuss the bioinformatic considerations of these analyses.


Subject(s)
Gene Expression Profiling/methods , Genome, Bacterial/genetics , High-Throughput Nucleotide Sequencing/methods , RNA, Bacterial/genetics , Sequence Analysis, RNA/methods , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Gene Expression Profiling/instrumentation , High-Throughput Nucleotide Sequencing/instrumentation , Open Reading Frames/genetics , Operon/genetics , Prokaryotic Cells/chemistry , Prokaryotic Cells/enzymology , Prokaryotic Cells/metabolism , RNA, Bacterial/isolation & purification , Sequence Analysis, RNA/instrumentation , Terminator Regions, Genetic/genetics , Transcription Initiation Site , Transcription, Genetic , Transcriptome/genetics
14.
Nucleic Acids Res ; 44(6): 2577-92, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26609136

ABSTRACT

Coupled transcription and translation in bacteria are tightly regulated. Some small RNAs (sRNAs) control aspects of this coupling by modifying ribosome access or inducing degradation of the message. Here, we show that sRNA IsrA (IS61 or McaS) specifically associates with core enzyme of RNAP in vivo and in vitro, independently of σ factor and away from the main nucleic-acids-binding channel of RNAP. We also show that, in the cells, IsrA exists as ribonucleoprotein particles (sRNPs), which involve a defined set of proteins including Hfq, S1, CsrA, ProQ and PNPase. Our findings suggest that IsrA might be directly involved in transcription or can participate in regulation of gene expression by delivering proteins associated with it to target mRNAs through its interactions with transcribing RNAP and through regions of sequence-complementarity with the target. In this eukaryotic-like model only in the context of a complex with its target, IsrA and its associated proteins become active. In this manner, in the form of sRNPs, bacterial sRNAs could regulate a number of targets with various outcomes, depending on the set of associated proteins.


Subject(s)
DNA-Directed RNA Polymerases/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , RNA, Bacterial/genetics , RNA, Small Untranslated/genetics , Ribonucleoproteins/genetics , Base Sequence , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Host Factor 1 Protein/genetics , Host Factor 1 Protein/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Molecular Sequence Data , Polyribonucleotide Nucleotidyltransferase/genetics , Polyribonucleotide Nucleotidyltransferase/metabolism , Protein Biosynthesis , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Untranslated/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribonucleoproteins/metabolism , Sigma Factor/genetics , Sigma Factor/metabolism , Transcription, Genetic
15.
J Nat Prod ; 80(5): 1558-1562, 2017 05 26.
Article in English | MEDLINE | ID: mdl-28398740

ABSTRACT

The madurastatins are pentapeptide siderophores originally described as containing an unusual salicylate-capped N-terminal aziridine ring. Isolation of madurastatin C1 (1) (also designated MBJ-0034), from Actinomadura sp. DEM31376 (itself isolated from a deep sea sediment), prompted structural reevaluation of the madurastatin siderophores, in line with the recent work of Thorson and Shaaban. NMR spectroscopy in combination with partial synthesis allowed confirmation of the structure of madurastatin C1 (1) as containing an N-terminal 2-(2-hydroxyphenyl)oxazoline in place of the originally postulated aziridine, while absolute stereochemistry was determined via Harada's advanced Marfey's method. Therefore, this work further supports Thorson and Shaaban's proposed structural revision of the madurastatin class of siderophores (madurastatins A1 (2), B1 (3), C1 (1), and MBJ-0036 (4)) as N-terminal 2-(2-hydroxyphenyl)oxazolines.


Subject(s)
Aziridines/chemistry , Oligopeptides/chemistry , Peptides/chemistry , Piperidones/chemistry , Siderophores/chemistry , Magnetic Resonance Spectroscopy , Molecular Structure , Stereoisomerism
16.
Nucleic Acids Res ; 43(13): 6299-308, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26038312

ABSTRACT

Regulation of transcription elongation is based on response of RNA polymerase (RNAP) to various pause signals and is modulated by various accessory factors. Here we report that a 7 kDa protein p7 encoded by bacteriophage Xp10 acts as an elongation processivity factor of RNAP of host bacterium Xanthomonas oryzae, a major rice pathogen. Our data suggest that p7 stabilizes the upstream DNA duplex of the elongation complex thus disfavouring backtracking and promoting forward translocated states of the elongation complex. The p7-induced 'pushing' of RNAP and modification of RNAP contacts with the upstream edge of the transcription bubble lead to read-through of various types of pauses and termination signals and generally increase transcription processivity and elongation rate, contributing for transcription of an extremely long late genes operon of Xp10. Forward translocation was observed earlier upon the binding of unrelated bacterial elongation factor NusG, suggesting that this may be a general pathway of regulation of transcription elongation.


Subject(s)
Bacteriophages , DNA-Directed RNA Polymerases/metabolism , Transcription Elongation, Genetic , Viral Proteins/metabolism , DNA/metabolism , Transcription Termination, Genetic , Xanthomonas/enzymology
17.
Nucleic Acids Res ; 43(3): 1529-36, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25605801

ABSTRACT

Collisions between paused transcription elongation complexes and replication forks inevitably happen, which may lead to collapse of replication fork and could be detrimental to cells. Bacterial transcription factor DksA and its cofactor alarmone ppGpp were proposed to contribute to prevention of such collisions, although the mechanism of this activity remains elusive. Here we show that DksA/ppGpp do not destabilise transcription elongation complexes or inhibit their backtracking, as was proposed earlier. Instead, we show, both in vitro and in vivo, that DksA/ppGpp increase fidelity of transcription elongation by slowing down misincorporation events. As misincorporation events cause temporary pauses, contribution to fidelity suggests the mechanism by which DksA/ppGpp contribute to prevention of collisions of transcription elongation complexes with replication forks. DksA is only the second known accessory factor, after transcription factor Gre, that increases fidelity of RNA synthesis in bacteria.


Subject(s)
Escherichia coli Proteins/physiology , Escherichia coli/physiology , Pyrophosphatases/physiology , Transcription, Genetic/physiology
18.
EMBO J ; 31(3): 630-9, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22124324

ABSTRACT

Pausing of transcription is an important step of regulation of gene expression in bacteria and eukaryotes. Here we uncover a factor-independent mechanism of transcription pausing, which is determined by the ability of the elongating RNA polymerase to recognize the sequence of the RNA-DNA hybrid. We show that, independently of thermodynamic stability of the elongation complex, RNA polymerase directly 'senses' the shape and/or identity of base pairs of the RNA-DNA hybrid. Recognition of the RNA-DNA hybrid sequence delays translocation by RNA polymerase, and thus slows down the nucleotide addition cycle through 'in pathway' mechanism. We show that this phenomenon is conserved among bacterial and eukaryotic RNA polymerases, and is involved in regulatory pauses, such as a pause regulating the production of virulence factors in some bacteria and a pause regulating transcription/replication of HIV-1. The results indicate that recognition of RNA-DNA hybrid sequence by multi-subunit RNA polymerases is involved in transcription regulation and may determine the overall rate of transcription elongation.


Subject(s)
DNA/chemistry , Nucleic Acid Hybridization , RNA/chemistry , Transcription, Genetic , Bacteria/pathogenicity , Base Sequence , Biocatalysis , DNA-Directed RNA Polymerases/metabolism , HIV-1/pathogenicity , Molecular Sequence Data , Virulence
19.
Methods ; 86: 51-9, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26080048

ABSTRACT

The various properties of RNA polymerase (RNAP) complexes with nucleic acids during different stages of transcription involve various types of regulation and different cross-talk with other cellular entities and with fellow RNAP molecules. The interactions of transcriptional apparatus with the translational machinery have been focused mainly in terms of outcomes of gene expression, whereas the study of the physical interaction of the ribosome and the RNAP remains obscure partly due to the lack of a system that allows such observations. In this article we will describe the methodology needed to set up a pure, transcription-coupled-to-translation system in which the translocation of the ribosome can be performed in a step-wise manner towards RNAP allowing investigation of the interactions between the two machineries at colliding and non-colliding distances. In the same time RNAP can be put in various types of states, such as paused, roadblocked, backtracked, etc. The experimental system thus allows studying the effects of the ribosome on different aspects of transcription elongation and the effects by RNAP on translation.


Subject(s)
DNA-Directed RNA Polymerases/genetics , Protein Biosynthesis , RNA, Messenger/genetics , Transcription, Genetic , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , Escherichia coli , Molecular Biology/methods , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , Ribosomes/genetics , Ribosomes/metabolism
20.
Nucleic Acids Res ; 42(17): 10987-99, 2014.
Article in English | MEDLINE | ID: mdl-25190458

ABSTRACT

Transcription by RNA polymerase may be interrupted by pauses caused by backtracking or misincorporation that can be resolved by the conserved bacterial Gre-factors. However, the consequences of such pausing in the living cell remain obscure. Here, we developed molecular biology and transcriptome sequencing tools in the human pathogen Streptococcus pneumoniae and provide evidence that transcription elongation is rate-limiting on highly expressed genes. Our results suggest that transcription elongation may be a highly regulated step of gene expression in S. pneumoniae. Regulation is accomplished via long-living elongation pauses and their resolution by elongation factor GreA. Interestingly, mathematical modeling indicates that long-living pauses cause queuing of RNA polymerases, which results in 'transcription traffic jams' on the gene and thus blocks its expression. Together, our results suggest that long-living pauses and RNA polymerase queues caused by them are a major problem on highly expressed genes and are detrimental for cell viability. The major and possibly sole function of GreA in S. pneumoniae is to prevent formation of backtracked elongation complexes.


Subject(s)
Bacterial Proteins/physiology , Gene Expression Regulation, Bacterial , Streptococcus pneumoniae/genetics , Transcription Elongation, Genetic , Transcriptional Elongation Factors/physiology , Models, Genetic , Promoter Regions, Genetic , Streptococcus pneumoniae/cytology , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/metabolism , Transcription Initiation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL