Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Biochemistry ; 63(13): 1608-1620, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38864595

ABSTRACT

Riboswitches are RNA-regulating elements that mostly rely on structural changes to modulate gene expression at various levels. Recent studies have revealed that riboswitches may control several regulatory mechanisms cotranscriptionally, i.e., during the transcription elongation of the riboswitch or early in the coding region of the regulated gene. Here, we study the structure of the nascent thiamin pyrophosphate (TPP)-sensing thiC riboswitch in Escherichia coli by using biochemical and enzymatic conventional probing approaches. Our chemical (in-line and lead probing) and enzymatic (nucleases S1, A, T1, and RNase H) probing data provide a comprehensive model of how TPP binding modulates the structure of the thiC riboswitch. Furthermore, by using transcriptional roadblocks along the riboswitch sequence, we find that a certain portion of nascent RNA is needed to sense TPP that coincides with the formation of the P5 stem loop. Together, our data suggest that conventional techniques may readily be used to study cotranscriptional folding of nascent RNAs.


Subject(s)
Escherichia coli , Nucleic Acid Conformation , RNA Folding , Riboswitch , Thiamine Pyrophosphate , Riboswitch/genetics , Thiamine Pyrophosphate/metabolism , Thiamine Pyrophosphate/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Transcription, Genetic , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Gene Expression Regulation, Bacterial , Bacterial Proteins
2.
Nat Commun ; 15(1): 3955, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38729929

ABSTRACT

Widespread manganese-sensing transcriptional riboswitches effect the dependable gene regulation needed for bacterial manganese homeostasis in changing environments. Riboswitches - like most structured RNAs - are believed to fold co-transcriptionally, subject to both ligand binding and transcription events; yet how these processes are orchestrated for robust regulation is poorly understood. Through a combination of single-molecule and bulk approaches, we discover how a single Mn2+ ion and the transcribing RNA polymerase (RNAP), paused immediately downstream by a DNA template sequence, are coordinated by the bridging switch helix P1.1 in the representative Lactococcus lactis riboswitch. This coordination achieves a heretofore-overlooked semi-docked global conformation of the nascent RNA, P1.1 base pair stabilization, transcription factor NusA ejection, and RNAP pause extension, thereby enforcing transcription readthrough. Our work demonstrates how a central, adaptable RNA helix functions analogous to a molecular fulcrum of a first-class lever system to integrate disparate signals for finely balanced gene expression control.


Subject(s)
DNA-Directed RNA Polymerases , Gene Expression Regulation, Bacterial , Lactococcus lactis , Nucleic Acid Conformation , RNA, Bacterial , Riboswitch , Transcription, Genetic , Riboswitch/genetics , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/chemistry , Manganese/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Single Molecule Imaging
3.
bioRxiv ; 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38562847

ABSTRACT

Protein synthesis begins with the formation of a ribosome-mRNA complex. In bacteria, the 30S ribosomal subunit is recruited to many mRNAs through base pairing with the Shine Dalgarno (SD) sequence and RNA binding by ribosomal protein bS1. Translation can initiate on nascent mRNAs and RNA polymerase (RNAP) can promote recruitment of the pioneering 30S subunit. Here we examined ribosome recruitment to nascent mRNAs using cryo-EM, single-molecule fluorescence co-localization, and in-cell crosslinking mass spectrometry. We show that bS1 delivers the mRNA to the ribosome for SD duplex formation and 30S subunit activation. Additionally, bS1 mediates the stimulation of translation initiation by RNAP. Together, our work provides a mechanistic framework for how the SD duplex, ribosomal proteins and RNAP cooperate in 30S recruitment to mRNAs and establish transcription-translation coupling.

4.
Res Sq ; 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38352525

ABSTRACT

Widespread manganese-sensing transcriptional riboswitches effect the dependable gene regulation needed for bacterial manganese homeostasis in changing environments. Riboswitches - like most structured RNAs - are believed to fold co-transcriptionally, subject to both ligand binding and transcription events; yet how these processes are orchestrated for robust regulation is poorly understood. Through a combination of single molecule and bulk approaches, we discovered how a single Mn 2+ ion and the transcribing RNA polymerase (RNAP), paused immediately downstream by a DNA template sequence, are coordinated by the bridging switch helix P1.1 in the paradigmatic Lactococcus lactis riboswitch. This coordination achieves a heretofore-overlooked semi-docked global conformation of the nascent RNA, P1.1 base pair stabilization, transcription factor NusA ejection, and RNAP pause extension, thereby enforcing transcription readthrough. Our work demonstrates how a central, adaptable RNA helix functions analogous to a molecular fulcrum of a first-class lever system to integrate disparate signals for finely balanced gene expression control.

5.
Cell Chem Biol ; 31(1): 71-85, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38211587

ABSTRACT

Commensal and pathogenic bacteria continuously evolve to survive in diverse ecological niches by efficiently coordinating gene expression levels in their ever-changing environments. Regulation through the RNA transcript itself offers a faster and more cost-effective way to adapt than protein-based mechanisms and can be leveraged for diagnostic or antimicrobial purposes. However, RNA can fold into numerous intricate, not always functional structures that both expand and obscure the plethora of roles that regulatory RNAs serve within the cell. Here, we review the current knowledge of bacterial non-coding RNAs in relation to their folding pathways and interactions. We posit that co-transcriptional folding of these transcripts ultimately dictates their downstream functions. Elucidating the spatiotemporal folding of non-coding RNAs during transcription therefore provides invaluable insights into bacterial pathogeneses and predictive disease diagnostics. Finally, we discuss the implications of co-transcriptional folding andapplications of RNAs for therapeutics and drug targets.


Subject(s)
RNA, Long Noncoding , RNA , Bacteria/genetics , Bacteria/metabolism , Genes, Bacterial , RNA, Untranslated , Gene Expression , Gene Expression Regulation , RNA, Bacterial/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
6.
RNA ; 30(4): 381-391, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38253429

ABSTRACT

Bacterial riboswitches are molecular structures that play a crucial role in controlling gene expression to maintain cellular balance. The Escherichia coli lysC riboswitch has been previously shown to regulate gene expression through translation initiation and mRNA decay. Recent research suggests that lysC gene expression is also influenced by Rho-dependent transcription termination. Through a series of in silico, in vitro, and in vivo experiments, we provide experimental evidence that the lysC riboswitch directly and indirectly modulates Rho transcription termination. Our study demonstrates that Rho-dependent transcription termination plays a significant role in the cotranscriptional regulation of lysC expression. Together with previous studies, our work suggests that lysC expression is governed by a lysine-sensing riboswitch that regulates translation initiation, transcription termination, and mRNA degradation. Notably, both Rho and RNase E target the same region of the RNA molecule, implying that RNase E may degrade Rho-terminated transcripts, providing a means to selectively eliminate these incomplete messenger RNAs. Overall, this study sheds light on the complex regulatory mechanisms used by bacterial riboswitches, emphasizing the role of transcription termination in the control of gene expression and mRNA stability.


Subject(s)
Riboswitch , Riboswitch/genetics , Base Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Transcription, Genetic , Bacteria/genetics , Gene Expression Regulation, Bacterial , RNA, Bacterial/metabolism
7.
Nat Struct Mol Biol ; 30(7): 902-913, 2023 07.
Article in English | MEDLINE | ID: mdl-37264140

ABSTRACT

Folding of nascent transcripts can be modulated by the RNA polymerase (RNAP) that carries out their transcription, and vice versa. A pause of RNAP during transcription of a preQ1 riboswitch (termed que-PEC) is stabilized by a previously characterized template consensus sequence and the ligand-free conformation of the nascent RNA. Ligand binding to the riboswitch induces RNAP pause release and downstream transcription termination; however, the mechanism by which riboswitch folding modulates pausing is unclear. Here, we report single-particle cryo-electron microscopy reconstructions of que-PEC in ligand-free and ligand-bound states. In the absence of preQ1, the RNA transcript is in an unexpected hyper-translocated state, preventing downstream nucleotide incorporation. Strikingly, on ligand binding, the riboswitch rotates around its helical axis, expanding the surrounding RNAP exit channel and repositioning the transcript for elongation. Our study reveals the tight coupling by which nascent RNA structures and their ligands can functionally regulate the macromolecular transcription machinery.


Subject(s)
Escherichia coli Proteins , Riboswitch , RNA, Bacterial/chemistry , Ligands , Cryoelectron Microscopy , Escherichia coli Proteins/metabolism , DNA-Directed RNA Polymerases/metabolism , Transcription, Genetic , RNA Folding , Bacteria/metabolism , Nucleic Acid Conformation
8.
Expert Opin Ther Targets ; 27(6): 433-445, 2023.
Article in English | MEDLINE | ID: mdl-37364239

ABSTRACT

INTRODUCTION: The growth of antibiotic resistance among bacterial pathogens is an impending global threat that can only be averted through the development of novel antibacterial drugs. A promising answer could be the targeting of riboswitches, structured RNA elements found almost exclusively in bacteria. AREAS COVERED: This review examines the potential of riboswitches as novel antibacterial drug targets. The limited mechanisms of action of currently available antibiotics are summarized, followed by a delineation of the functional mechanisms of riboswitches. We then discuss the potential for developing novel approaches that target paradigmatic riboswitches in the context of their bacterial gene expression machinery. EXPERT OPINION: We highlight potential advantages of targeting riboswitches in their functional form, embedded within gene expression complexes critical for bacterial survival. We emphasize the benefits of this approach, including potentially higher species specificity and lower side effects.


Subject(s)
Riboswitch , Humans , Riboswitch/genetics , Anti-Bacterial Agents/pharmacology , Bacteria/genetics
9.
Nat Commun ; 13(1): 207, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017489

ABSTRACT

The archetypical transcriptional crcB fluoride riboswitch from Bacillus cereus is an intricately structured non-coding RNA element enhancing gene expression in response to toxic levels of fluoride. Here, we used single molecule FRET to uncover three dynamically interconverting conformations appearing along the transcription process: two distinct undocked states and one pseudoknotted docked state. We find that the fluoride anion specifically snap-locks the magnesium-induced, dynamically docked state. The long-range, nesting, single base pair A40-U48 acts as the main linchpin, rather than the multiple base pairs comprising the pseudoknot. We observe that the proximally paused RNA polymerase further fine-tunes the free energy to promote riboswitch docking. Finally, we show that fluoride binding at short transcript lengths is an early step toward partitioning folding into the docked conformation. These results reveal how the anionic fluoride ion cooperates with the magnesium-associated RNA to govern regulation of downstream genes needed for fluoride detoxification of the cell.


Subject(s)
Bacillus cereus/chemistry , Fluorides/chemistry , Magnesium/chemistry , RNA, Bacterial/chemistry , RNA, Messenger/chemistry , Riboswitch , Bacillus cereus/genetics , Bacillus cereus/metabolism , Base Pairing , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Fluorides/pharmacology , Gene Expression Regulation, Bacterial , Magnesium/metabolism , Molecular Docking Simulation , Nucleic Acid Conformation , Protein Binding , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thermodynamics
10.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Article in English | MEDLINE | ID: mdl-34740970

ABSTRACT

Cotranscriptional RNA folding is crucial for the timely control of biological processes, but because of its transient nature, its study has remained challenging. While single-molecule Förster resonance energy transfer (smFRET) is unique to investigate transient RNA structures, its application to cotranscriptional studies has been limited to nonnative systems lacking RNA polymerase (RNAP)-dependent features, which are crucial for gene regulation. Here, we present an approach that enables site-specific labeling and smFRET studies of kilobase-length transcripts within native bacterial complexes. By monitoring Escherichia coli nascent riboswitches, we reveal an inverse relationship between elongation speed and metabolite-sensing efficiency and show that pause sites upstream of the translation start codon delimit a sequence hotspot for metabolite sensing during transcription. Furthermore, we demonstrate a crucial role of the bacterial RNAP actively delaying the formation, within the hotspot sequence, of competing structures precluding metabolite binding. Our approach allows the investigation of cotranscriptional regulatory mechanisms in bacterial and eukaryotic elongation complexes.


Subject(s)
Escherichia coli Proteins/metabolism , Riboswitch/physiology , Single Molecule Imaging/methods , Transcription Elongation, Genetic , Carbocyanines , Escherichia coli , Escherichia coli Proteins/analysis , Fluorescence Resonance Energy Transfer , Fluorescent Dyes
11.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34782462

ABSTRACT

Cotranscriptional RNA folding is widely assumed to influence the timely control of gene expression, but our understanding remains limited. In bacteria, the fluoride (F-)-sensing riboswitch is a transcriptional control element essential to defend against toxic F- levels. Using this model riboswitch, we find that its ligand F- and essential bacterial transcription factor NusA compete to bind the cotranscriptionally folding RNA, opposing each other's modulation of downstream pausing and termination by RNA polymerase. Single-molecule fluorescence assays probing active transcription elongation complexes discover that NusA unexpectedly binds highly reversibly, frequently interrogating the complex for emerging, cotranscriptionally folding RNA duplexes. NusA thus fine-tunes the transcription rate in dependence of the ligand-responsive higher-order structure of the riboswitch. At the high NusA concentrations found intracellularly, this dynamic modulation is expected to lead to adaptive bacterial transcription regulation with fast response times.


Subject(s)
Escherichia coli Proteins/metabolism , Ligands , Riboswitch , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , RNA Folding , RNA, Bacterial/genetics , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/genetics
12.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Article in English | MEDLINE | ID: mdl-33850018

ABSTRACT

Bacterial messenger RNA (mRNA) synthesis by RNA polymerase (RNAP) and first-round translation by the ribosome are often coupled to regulate gene expression, yet how coupling is established and maintained is ill understood. Here, we develop biochemical and single-molecule fluorescence approaches to probe the dynamics of RNAP-ribosome interactions on an mRNA with a translational preQ1-sensing riboswitch in its 5' untranslated region. Binding of preQ1 leads to the occlusion of the ribosome binding site (RBS), inhibiting translation initiation. We demonstrate that RNAP poised within the mRNA leader region promotes ribosomal 30S subunit binding, antagonizing preQ1-induced RBS occlusion, and that the RNAP-30S bridging transcription factors NusG and RfaH distinctly enhance 30S recruitment and retention, respectively. We further find that, while 30S-mRNA interaction significantly impedes RNAP in the absence of translation, an actively translating ribosome promotes productive transcription. A model emerges wherein mRNA structure and transcription factors coordinate to dynamically modulate the efficiency of transcription-translation coupling.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Ribosomes/metabolism , Riboswitch/physiology , 5' Untranslated Regions , Binding Sites , DNA-Directed RNA Polymerases/physiology , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Peptide Elongation Factors/metabolism , Protein Biosynthesis/genetics , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Ribosomes/genetics , Riboswitch/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics
13.
Nucleic Acids Res ; 47(12): 6478-6487, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31045204

ABSTRACT

Riboswitches are cis-acting regulatory RNA biosensors that rival the efficiency of those found in proteins. At the heart of their regulatory function is the formation of a highly specific aptamer-ligand complex. Understanding how these RNAs recognize the ligand to regulate gene expression at physiological concentrations of Mg2+ ions and ligand is critical given their broad impact on bacterial gene expression and their potential as antibiotic targets. In this work, we used single-molecule FRET and biochemical techniques to demonstrate that Mg2+ ions act as fine-tuning elements of the amino acid-sensing lysC aptamer's ligand-free structure in the mesophile Bacillus subtilis. Mg2+ interactions with the aptamer produce encounter complexes with strikingly different sensitivities to the ligand in different, yet equally accessible, physiological ionic conditions. Our results demonstrate that the aptamer adapts its structure and folding landscape on a Mg2+-tunable scale to efficiently respond to changes in intracellular lysine of more than two orders of magnitude. The remarkable tunability of the lysC aptamer by sub-millimolar variations in the physiological concentration of Mg2+ ions suggests that some single-aptamer riboswitches have exploited the coupling of cellular levels of ligand and divalent metal ions to tightly control gene expression.


Subject(s)
Gene Expression Regulation, Bacterial , Magnesium/physiology , Riboswitch , Bacillus subtilis/chemistry , Bacillus subtilis/genetics , Fluorescence Resonance Energy Transfer , Ligands , Magnesium/analysis , RNA Folding , Transcription, Genetic
14.
PLoS Genet ; 15(5): e1008157, 2019 05.
Article in English | MEDLINE | ID: mdl-31136569

ABSTRACT

Most transcriptional activity of exponentially growing cells is carried out by the RNA Polymerase I (Pol I), which produces a ribosomal RNA (rRNA) precursor. In budding yeast, Pol I is a multimeric enzyme with 14 subunits. Among them, Rpa49 forms with Rpa34 a Pol I-specific heterodimer (homologous to PAF53/CAST heterodimer in human Pol I), which might be responsible for the specific functions of the Pol I. Previous studies provided insight in the involvement of Rpa49 in initiation, elongation, docking and releasing of Rrn3, an essential Pol I transcription factor. Here, we took advantage of the spontaneous occurrence of extragenic suppressors of the growth defect of the rpa49 null mutant to better understand the activity of Pol I. Combining genetic approaches, biochemical analysis of rRNA synthesis and investigation of the transcription rate at the individual gene scale, we characterized mutated residues of the Pol I as novel extragenic suppressors of the growth defect caused by the absence of Rpa49. When mapped on the Pol I structure, most of these mutations cluster within the jaw-lobe module, at an interface formed by the lobe in Rpa135 and the jaw made up of regions of Rpa190 and Rpa12. In vivo, the suppressor allele RPA135-F301S restores normal rRNA synthesis and increases Pol I density on rDNA genes when Rpa49 is absent. Growth of the Rpa135-F301S mutant is impaired when combined with exosome mutation rrp6Δ and it massively accumulates pre-rRNA. Moreover, Pol I bearing Rpa135-F301S is a hyper-active RNA polymerase in an in vitro tailed-template assay. We conclude that RNA polymerase I can be engineered to produce more rRNA in vivo and in vitro. We propose that the mutated area undergoes a conformational change that supports the DNA insertion into the cleft of the enzyme resulting in a super-active form of Pol I.


Subject(s)
Pol1 Transcription Initiation Complex Proteins/genetics , RNA Polymerase I/genetics , DNA, Ribosomal/genetics , Pol1 Transcription Initiation Complex Proteins/metabolism , RNA Precursors/genetics , RNA, Ribosomal , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic
15.
RNA Biol ; 16(8): 1066-1073, 2019 08.
Article in English | MEDLINE | ID: mdl-31081713

ABSTRACT

Transcriptional pauses have been reported in bacterial riboswitches and, in some cases, their specific positioning has been shown to be important for gene regulation. Here, we show that a hairpin structure in the Escherichia coli thiamin pyrophosphate (TPP) thiC riboswitch is involved in transcriptional pausing and ligand sensitivity. Using in vitro transcription kinetic experiments, we show that all three major transcriptional pauses in the thiC riboswitch are affected by NusA, a transcriptional factor known to stimulate hairpin-stabilized pauses. Using a truncated region of the riboswitch, we isolated the hairpin structure responsible for stabilization of the most upstream pause. Destabilization of this structure led to a weaker pause and a decreased NusA effect. In the context of the full-length riboswitch, this same mutation also led to a weaker pause, as well as a decreased TPP binding affinity. Our work suggests that RNA structures involved in transcriptional pausing in riboswitches are important for ligand sensitivity, most likely by increasing the time allowed to the ligand for binding to the riboswitch.


Subject(s)
Bacterial Proteins/genetics , Escherichia coli Proteins/genetics , Riboswitch/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/genetics , Nucleic Acid Conformation , Thiamine Pyrophosphate/genetics , Transcription Factors/genetics
16.
Methods ; 162-163: 3-11, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30951833

ABSTRACT

RNA structures and their dynamic fluctuations lie at the heart of understanding key biological process such as transcription, splicing, translation and RNA decay. While conventional bulk assays have proven to identify and characterize key pathway intermediates, the generally dynamic nature of RNA structures renders the information obtained from time and ensemble averaging techniques necessarily lacking in critical details. Here we detail Single-Molecule Kinetic Analysis of RNA Transient Structure (SiM-KARTS), a method that readily monitors structural fluctuations of single RNA molecules through the repetitive interaction of fluorescent probes with an unlabeled, surface-immobilized RNA target of virtually any length and in any biological context. In addition, we demonstrate the broad applicability of SiM-KARTS by kinetically fingerprinting the binding of cognate tRNA ligand to single immobilized T-box riboswitch molecules. SiM-KARTS represents a valuable tool for probing biologically relevant structure and interaction features of potentially many diverse RNA metabolic pathways.


Subject(s)
Nucleic Acid Conformation , RNA, Transfer/metabolism , Riboswitch , Single Molecule Imaging/methods , Bacillus subtilis/genetics , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Image Processing, Computer-Assisted , Kinetics , Markov Chains , Microscopy, Fluorescence/methods , Oligonucleotides/chemistry , Oligonucleotides/metabolism , RNA Probes/chemistry , RNA Probes/metabolism , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Transfer/chemistry , Software
17.
RNA Biol ; 16(9): 1077-1085, 2019 09.
Article in English | MEDLINE | ID: mdl-30328748

ABSTRACT

Riboswitches are dynamic RNA motifs that are mostly embedded in the 5'-untranslated regions of bacterial mRNAs, where they regulate gene expression transcriptionally or translationally by undergoing conformational changes upon binding of a small metabolite or ion. Due to the small size of typical ligands, relatively little free energy is available from ligand binding to overcome the often high energetic barrier of reshaping RNA structure. Instead, most riboswitches appear to take advantage of the directional and hierarchical folding of RNA by employing the ligand as a structural 'linchpin' to adjust the kinetic partitioning between alternate folds. In this model, even small, local structural and kinetic effects of ligand binding can cascade into global RNA conformational changes affecting gene expression. Single-molecule (SM) microscopy tools are uniquely suited to study such kinetically controlled RNA folding since they avoid the ensemble averaging of bulk techniques that loses sight of unsynchronized, transient, and/or multi-state kinetic behavior. This review summarizes how SM methods have begun to unravel riboswitch-mediated gene regulation.


Subject(s)
Gene Expression Regulation, Bacterial/genetics , RNA Folding/genetics , Riboswitch/genetics , Single Molecule Imaging/methods , Bacteria/genetics , Fluorescence Resonance Energy Transfer/methods , Kinetics , Microscopy, Fluorescence/methods , Optical Tweezers
18.
Nucleic Acids Res ; 45(12): 7474-7486, 2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28520932

ABSTRACT

Riboswitches are regulatory elements that control gene expression by altering RNA structure upon the binding of specific metabolites. Although Bacillus subtilis riboswitches have been shown to control premature transcription termination, less is known about regulatory mechanisms employed by Escherichia coli riboswitches, which are predicted to regulate mostly at the level of translation initiation. Here, we present experimental evidence suggesting that the majority of known E. coli riboswitches control transcription termination by using the Rho transcription factor. In the case of the thiamin pyrophosphate-dependent thiM riboswitch, we find that Rho-dependent transcription termination is triggered as a consequence of translation repression. Using in vitro and in vivo assays, we show that the Rho-mediated regulation relies on RNA target elements located at the beginning of thiM coding region. Gene reporter assays indicate that relocating Rho target elements to a different gene induces transcription termination, demonstrating that such elements are modular domains controlling Rho. Our work provides strong evidence that translationally regulating riboswitches also regulate mRNA levels through an indirect control mechanism ensuring tight control of gene expression.


Subject(s)
Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Protein Biosynthesis , Rho Factor/genetics , Riboswitch , Transcription Termination, Genetic , Base Sequence , Escherichia coli/metabolism , Genes, Reporter , Nucleic Acid Conformation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rho Factor/metabolism , Thiamine Pyrophosphate/metabolism
19.
Nat Commun ; 8: 13892, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28071751

ABSTRACT

On the basis of nascent transcript sequencing, it has been postulated but never demonstrated that transcriptional pausing at translation start sites is important for gene regulation. Here we show that the Escherichia coli thiamin pyrophosphate (TPP) thiC riboswitch contains a regulatory pause site in the translation initiation region that acts as a checkpoint for thiC expression. By biochemically probing nascent transcription complexes halted at defined positions, we find a narrow transcriptional window for metabolite binding, in which the downstream boundary is delimited by the checkpoint. We show that transcription complexes at the regulatory pause site favour the formation of a riboswitch intramolecular lock that strongly prevents TPP binding. In contrast, cotranscriptional metabolite binding increases RNA polymerase pausing and induces Rho-dependent transcription termination at the checkpoint. Early transcriptional pausing may provide a general mechanism, whereby transient transcriptional windows directly coordinate the sensing of environmental cues and bacterial mRNA regulation.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Riboswitch/genetics , Bacterial Proteins/metabolism , Codon, Initiator , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Mutation , Protein Biosynthesis , Protein Conformation , Ribonuclease H/genetics , Ribonuclease H/metabolism , Thiamine Pyrophosphate/metabolism , Transcription, Genetic
20.
Methods Mol Biol ; 1334: 109-14, 2015.
Article in English | MEDLINE | ID: mdl-26404146

ABSTRACT

The study of biologically significant and native structures is vital to characterize RNA-based regulatory mechanisms. Riboswitches are cis-acting RNA molecules that are involved in the biosynthesis and transport of cellular metabolites. Because riboswitches regulate gene expression by modulating their structure, it is vital to employ native probing assays to determine how native riboswitch structures perform highly efficient and specific ligand recognition. By employing RNase H probing, it is possible to determine the accessibility of specific RNA domains in various structural contexts. Herein, we describe how to employ RNase H probing to characterize nascent mRNA riboswitch molecules as a way to obtain information regarding the riboswitch regulation control mechanism.


Subject(s)
Molecular Biology/methods , RNA, Messenger/chemistry , Ribonuclease H/chemistry , Riboswitch/genetics , Gene Expression Regulation, Bacterial , Ligands , Nucleic Acid Conformation , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...