Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 284
Filter
1.
Proc Natl Acad Sci U S A ; 119(10): e2117930119, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35239434

ABSTRACT

SignificanceWhile most small, regulatory RNAs are thought to be "noncoding," a few have been found to also encode a small protein. Here we describe a 164-nucleotide RNA that encodes a 28-amino acid, amphipathic protein, which interacts with aerobic glycerol-3-phosphate dehydrogenase and increases dehydrogenase activity but also base pairs with two mRNAs to reduce expression. The coding and base-pairing sequences overlap, and the two regulatory functions compete.


Subject(s)
Carbon/metabolism , Escherichia coli/metabolism , RNA, Bacterial/physiology , Culture Media , Escherichia coli/growth & development , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Galactose/metabolism , Glycerol/metabolism , Glycerolphosphate Dehydrogenase/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Protein Biosynthesis , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Messenger/metabolism
2.
BMC Plant Biol ; 21(1): 155, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33771101

ABSTRACT

BACKGROUND: Anastomosis group 1 IA (AG1-IA) of Rhizoctonia solani is the major agent of banded leaf and sheath blight (BLSB) disease that causes severe yield loss in many worldwide crops. MicroRNAs (miRNAs) are ~ 22 nt non-coding RNAs that negatively regulate gene expression levels by mRNA degradation or translation inhibition. A better understanding of miRNA function during AG1-IA infection can expedite to elucidate the molecular mechanisms of fungi-host interactions. RESULTS: In this study, we sequenced three small RNA libraries obtained from the mycelium of AG1-IA isolate, non-infected maize sheath and mixed maize sheath 3 days after inoculation. In total, 137 conserved and 34 novel microRNA-like small RNAs (milRNAs) were identified from the pathogen. Among these, one novel and 17 conserved milRNAs were identified as potential virulence-associated (VA) milRNAs. Subsequently, the prediction of target genes for these milRNAs was performed in both AG1-IA and maize, while functional annotation of these targets suggested a link to pathogenesis-related biological processes. Further, expression patterns of these virulence-associated milRNAs demonstrated that theyparticipate in the virulence of AG1-IA. Finally, regulation of one maize targeting gene, GRMZM2G412674 for Rhi-milRNA-9829-5p, was validated by dual-luciferase assay and identified to play a positive role in BLSB resistance in two maize mutants. These results suggest the global differentially expressed milRNAs of R. solani AG1-IA that participate in the regulation of target genes in both AG1-IA and maize to reinforce its pathogenicity. CONCLUSIONS: Our data have provided a comprehensive overview of the VA-milRNAs of R. solani and identified that they are probably the virulence factors by directly interfered in host targeting genes. These results offer new insights on the molecular mechanisms of R.solani-maize interactions during the process of infection.


Subject(s)
MicroRNAs/physiology , Plant Diseases/microbiology , Rhizoctonia/pathogenicity , Zea mays/microbiology , Gene Expression Profiling , Gene Expression Regulation, Plant , High-Throughput Nucleotide Sequencing , Plant Diseases/genetics , RNA, Bacterial/physiology , Rhizoctonia/genetics , Virulence/genetics , Zea mays/genetics
3.
Mol Plant Pathol ; 22(2): 255-270, 2021 02.
Article in English | MEDLINE | ID: mdl-33314618

ABSTRACT

Erwinia amylovora is the causative agent of the devastating disease fire blight of pome fruit trees. After infection of host plant leaves at apple shoot tips, E. amylovora cells form biofilms in xylem vessels, restrict water flow, and cause wilting symptoms. Although E. amylovora is well known to be able to cause systemic infection, how biofilm cells of E. amylovora transit from the sessile mode of growth in xylem to the planktonic mode of growth in cortical parenchyma remains unknown. Increasing evidence has suggested the important modulatory roles of Hfq-dependent small RNAs (sRNAs) in the pathogenesis of E. amylovora. Here, we demonstrate that the sRNA RprA acts as a positive regulator of amylovoran exopolysaccharide production, the type III secretion system (T3SS), and flagellar-dependent motility, and as a negative regulator of levansucrase activity and cellulose production. We also show that RprA affects the promoter activity of multiple virulence factor genes and regulates hrpS, a critical T3SS regulator, at the posttranscriptional level. We determined that rprA expression can be activated by the Rcs phosphorelay, and that expression is active during T3SS-mediated host infection in an immature pear fruit infection model. We further showed that overexpression of rprA activated the in vitro dispersal of E. amylovora cells from biofilms. Thus, our investigation of the varied role of RprA in affecting E. amylovora virulence provides important insights into the functions of this sRNA in biofilm control and systemic infection.


Subject(s)
Erwinia amylovora/metabolism , RNA, Bacterial/physiology , Virulence Factors/metabolism , Biofilms , Cellulose/genetics , Erwinia amylovora/genetics , Erwinia amylovora/pathogenicity , Hexosyltransferases/genetics , Movement , Polysaccharides, Bacterial/genetics , Promoter Regions, Genetic , Type III Secretion Systems/genetics , Virulence Factors/genetics
4.
Appl Environ Microbiol ; 87(3)2021 01 15.
Article in English | MEDLINE | ID: mdl-33158897

ABSTRACT

Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen that undergoes swarming motility in response to semisolid conditions with amino acids as a nitrogen source. With a genome encoding hundreds of potential intergenic small RNAs (sRNAs), P. aeruginosa can easily adapt to different conditions and stresses. We previously identified 20 sRNAs that were differentially expressed (DE) under swarming conditions. Here, these sRNAs were overexpressed in strain PAO1 and were subjected to an array of phenotypic screens. Overexpression of the PrrH sRNA resulted in decreased swimming motility, whereas a ΔprrH mutant had decreased cytotoxicity and increased pyoverdine production. Overexpression of the previously uncharacterized PA2952.1 sRNA resulted in decreased swarming and swimming motilities, increased gentamicin and tobramycin resistance under swarming conditions, and increased trimethoprim susceptibility. Transcriptome sequencing (RNA-Seq) and proteomic analysis were performed on the wild type (WT) overexpressing PA2952.1 compared to the empty vector control under swarming conditions, and these revealed the differential expression (absolute fold change [FC] ≥ 1.5) of 784 genes and the differential abundance (absolute FC ≥ 1.25) of 59 proteins. Among these were found 73 transcriptional regulators, two-component systems, and sigma and anti-sigma factors. Downstream effectors included downregulated pilus and flagellar genes, the upregulated efflux pump MexGHI-OpmD, and the upregulated arn operon. Genes involved in iron and zinc uptake were generally upregulated, and certain pyoverdine genes were upregulated. Overall, the sRNAs PA2952.1 and PrrH appeared to be involved in regulating virulence-related programs in P. aeruginosa, including iron acquisition and motility.IMPORTANCE Due to the rising incidence of multidrug-resistant (MDR) strains and the difficulty of eliminating P. aeruginosa infections, it is important to understand the regulatory mechanisms that allow this bacterium to adapt to and thrive under a variety of conditions. Small RNAs (sRNAs) are one regulatory mechanism that allows bacteria to change the amount of protein synthesized. In this study, we overexpressed 20 different sRNAs in order to investigate how this might affect different bacterial behaviors. We found that one of the sRNAs, PrrH, played a role in swimming motility and virulence phenotypes, indicating a potentially important role in clinical infections. Another sRNA, PA2952.1, affected other clinically relevant phenotypes, including motility and antibiotic resistance. RNA-Seq and proteomics of the strain overexpressing PA2952.1 revealed the differential expression of 784 genes and 59 proteins, with a total of 73 regulatory factors. This substantial dysregulation indicates an important role for the sRNA PA2952.1.


Subject(s)
Iron/metabolism , Pseudomonas aeruginosa/genetics , RNA, Bacterial/physiology , Virulence , Bacterial Proteins/genetics , Cell Line , Cell Survival , Genes, Bacterial , Humans , Proteomics , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/physiology , Zinc/metabolism
5.
J Bacteriol ; 203(2)2020 12 18.
Article in English | MEDLINE | ID: mdl-33077637

ABSTRACT

Staphylococcus aureus capsule polysaccharide is an important antiphagocytic virulence factor. The cap genes are regulated at the promoter element (Pcap) upstream of the cap operon. Pcap, which consists of a dominant SigB-dependent promoter and a weaker upstream SigA-dependent promoter, is activated by global regulator MgrA. How MgrA activates capsule is unclear. Here, we showed that MgrA directly bound to the Pcap region and affected the SigA-dependent promoter. Interestingly, an electrophoretic mobility shift assay showed that MgrA bound to a large region of Pcap, mainly downstream of the SigA-dependent promoter. We further showed that the ArlRS two-component system and the Agr quorum sensing system activated capsule primarily through MgrA in the early growth phases.IMPORTANCE The virulence of Staphylococcus aureus depends on the expression of various virulence factors, which is governed by a complex regulatory network. We have been using capsule as a model virulence factor to study virulence gene regulation in S. aureus MgrA is one of the regulators of capsule and has a major effect on capsule production. However, how MgrA regulates capsule genes is not understood. In this study, we were able to define the mechanism involving MgrA regulation of capsule. In addition, we also delineated the role of MgrA in capsule regulatory pathways involving the key virulence regulators Agr and Arl. This study further advances our understanding of virulence gene regulation in S. aureus, an important human pathogen.


Subject(s)
Bacterial Capsules/chemistry , Immunoglobulin A, Secretory/physiology , Polysaccharides, Bacterial/physiology , Promoter Regions, Genetic/physiology , Staphylococcus aureus/physiology , Virulence Factors/physiology , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Electrophoretic Mobility Shift Assay , Immunoblotting , Immunoglobulin A, Secretory/genetics , Mutation , Polysaccharides, Bacterial/genetics , RNA, Bacterial/isolation & purification , RNA, Bacterial/physiology , Real-Time Polymerase Chain Reaction , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Reverse Transcription , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Virulence/genetics , Virulence Factors/genetics
6.
Elife ; 92020 08 03.
Article in English | MEDLINE | ID: mdl-32744240

ABSTRACT

Negative feedback regulation, that is the ability of a gene to repress its own synthesis, is the most abundant regulatory motif known to biology. Frequently reported for transcriptional regulators, negative feedback control relies on binding of a transcription factor to its own promoter. Here, we report a novel mechanism for gene autoregulation in bacteria relying on small regulatory RNA (sRNA) and the major endoribonuclease, RNase E. TIER-seq analysis (transiently-inactivating-an-endoribonuclease-followed-by-RNA-seq) revealed ~25,000 RNase E-dependent cleavage sites in Vibrio cholerae, several of which resulted in the accumulation of stable sRNAs. Focusing on two examples, OppZ and CarZ, we discovered that these sRNAs are processed from the 3' untranslated region (3' UTR) of the oppABCDF and carAB operons, respectively, and base-pair with their own transcripts to inhibit translation. For OppZ, this process also triggers Rho-dependent transcription termination. Our data show that sRNAs from 3' UTRs serve as autoregulatory elements allowing negative feedback control at the post-transcriptional level.


Subject(s)
3' Untranslated Regions/physiology , Gene Expression Regulation, Bacterial , RNA, Bacterial/physiology , RNA, Small Untranslated/physiology , Vibrio cholerae/genetics , Endoribonucleases/metabolism , Feedback, Physiological , Protein Biosynthesis , RNA-Seq , Rho Factor/metabolism , Terminator Regions, Genetic , Vibrio cholerae/enzymology
7.
Mol Microbiol ; 114(3): 363-366, 2020 09.
Article in English | MEDLINE | ID: mdl-32367584

ABSTRACT

Regulation at the post-transcriptional level is an important mode of gene expression control in bacteria. Small RNA regulators (sRNAs) that act via intramolecular base-pairing with target mRNAs are key players in this process and most often sequester the target's ribosome binding site (RBS) to down-regulate translation initiation. Over the past few years, several exceptions from this mechanism have been reported, revealing that sRNAs are able to influence translation initiation from a distance. In this issue of Molecular Microbiology, Azam and Vanderpool show that repression of the manY mRNA by the sRNA SgrS relies on an unconventional mechanism involving a translational enhancer element and ribosomal protein S1. Binding of S1 to an AU-rich sequence within the 5' untranslated region of the manY transcript promotes translation of the mRNA, and base-pairing of SgrS to the same site can interfere with this process. Therefore, instead of blocking translation initiation by occluding the manY RBS, SgrS reduces ManY synthesis by inhibiting S1-dependent translation activation. These findings increase the base-pairing window for sRNA-mediated gene expression control in bacteria and highlight the role of ribosomal protein S1 for translation initiation.


Subject(s)
Bacteria/genetics , Peptide Chain Initiation, Translational , RNA, Bacterial/physiology , RNA, Small Untranslated/genetics , Ribosomal Proteins/genetics , 5' Untranslated Regions , Base Pairing/genetics , Binding Sites , Enhancer Elements, Genetic , Gene Expression Regulation, Bacterial , Protein Biosynthesis , RNA, Messenger/genetics , Ribosomes/physiology
8.
J Microbiol ; 58(6): 499-506, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32279276

ABSTRACT

RaoN is a Salmonella-specific small RNA that is encoded in the cspH-envE intergenic region on Salmonella pathogenicity island-11. We previously reported that RaoN is induced under conditions of acid and oxidative stress combined with nutrient limitation, contributing to the intramacrophage growth of Salmonella enterica serovar Typhimurium. However, the role of RaoN in nitrosative stress response and virulence has not yet been elucidated. Here we show that the raoN mutant strain has increased susceptibility to nitrosative stress by using a nitric oxide generating acidified nitrite. Extending previous research on the role of RaoN in oxidative stress resistance, we found that NADPH oxidase inhibition restores the growth of the raoN mutant in LPS-treated J774A.1 macrophages. Flow cytometry analysis further revealed that the inactivation of raoN leads to an increase in the intracellular level of reactive oxygen species (ROS) in Salmonella-infected macrophages, suggesting that RaoN is involved in the inhibition of NADPH oxidase-mediated ROS production by mechanisms not yet resolved. Moreover, we evaluated the effect of raoN mutation on the virulence in murine systemic infection and determined that the raoN mutant is less virulent than the wild-type strain following oral inoculation. In conclusion, small regulatory RNA RaoN controls nitrosative-oxidative stress resistance and is required for virulence of Salmonella in mice.


Subject(s)
Oxidative Stress , RNA, Bacterial/physiology , RNA, Small Untranslated/physiology , Salmonella Infections/microbiology , Animals , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , RAW 264.7 Cells , Salmonella typhimurium/pathogenicity , Virulence
9.
BMC Vet Res ; 15(1): 319, 2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31488137

ABSTRACT

BACKGROUND: Small non-coding RNAs (sRNAs) regulate bacterial gene expression at the post-transcriptional level. STnc640 is a type of sRNA that was identified in Salmonella Typhimurium. RESULTS: In this study, STnc640 in Salmonella Enteritidis was confirmed to be an Hfq-dependent sRNA. TargetRNA software analysis showed that fimbrial genes fimA and bcfA were likely to be the target genes of STnc640. To investigate the target mRNAs and function of STnc640 in pathogenicity, we constructed the deletion mutant strain 50336△stnc640 and the complemented strain 50336△stnc640/pstnc640 in Salmonella Enteritidis 50336. The RT-qPCR results showed that the mRNA level of fimA was decreased, while bcfA was unchanged in 50336△stnc640 compared with that in the wild type (WT) strain. The adhesion ability of 50336△stnc640 to Caco-2 cells was increased compared to the 50336 WT strain. The virulence of 50336△stnc640 was enhanced in a one-day-old chicken model of S. Enteritidis disease as determined by quantifying the 50% lethal dose (LD50) of the bacterial strains. CONCLUSIONS: The results demonstrate that STnc640 contributes to the virulence of Salmonella Enteritidis.


Subject(s)
Antigens, Bacterial/genetics , Fimbriae Proteins/genetics , Gene Expression Regulation, Bacterial , RNA, Bacterial/physiology , RNA, Small Untranslated/physiology , Salmonella enteritidis/genetics , Salmonella enteritidis/pathogenicity , Animals , Bacterial Adhesion/genetics , Caco-2 Cells , Chickens , Female , Humans , Male , Poultry Diseases/virology , Salmonella Infections, Animal/virology , Sequence Deletion , Virulence/genetics
10.
J Bacteriol ; 201(22)2019 11 15.
Article in English | MEDLINE | ID: mdl-31451541

ABSTRACT

Coxiella burnetii is an obligate intracellular gammaproteobacterium and zoonotic agent of Q fever. We previously identified 15 small noncoding RNAs (sRNAs) of C. burnetii One of them, CbsR12 (Coxiella burnetiismall RNA 12), is highly transcribed during axenic growth and becomes more prominent during infection of cultured mammalian cells. Secondary structure predictions of CbsR12 revealed four putative CsrA-binding sites in stem loops with consensus AGGA/ANGGA motifs. We subsequently determined that CbsR12 binds to recombinant C. burnetii CsrA-2, but not CsrA-1, proteins in vitro Moreover, through a combination of in vitro and cell culture assays, we identified several in trans mRNA targets of CbsR12. Of these, we determined that CbsR12 binds and upregulates translation of carA transcripts coding for carbamoyl phosphate synthetase A, an enzyme that catalyzes the first step of pyrimidine biosynthesis. In addition, CbsR12 binds and downregulates translation of metK transcripts coding for S-adenosylmethionine synthetase, a component of the methionine cycle. Furthermore, we found that CbsR12 binds to and downregulates the quantity of cvpD transcripts, coding for a type IVB effector protein, in mammalian cell culture. Finally, we found that CbsR12 is necessary for expansion of Coxiella-containing vacuoles and affects growth rates in a dose-dependent manner in the early phase of infecting THP-1 cells. This is the first characterization of a trans-acting sRNA of C. burnetii and the first example of a bacterial sRNA that regulates both CarA and MetK synthesis. CbsR12 is one of only a few identified trans-acting sRNAs that interacts with CsrA.IMPORTANCE Regulation of metabolism and virulence in C. burnetii is not well understood. Here, we show that C. burnetii small RNA 12 (CbsR12) is highly transcribed in the metabolically active large-cell variant compared to the nonreplicative small-cell variant. We show that CbsR12 directly regulates several genes involved in metabolism, along with a type IV effector gene, in trans In addition, we demonstrate that CbsR12 binds to CsrA-2 in vitro and induces autoaggregation and biofilm formation when transcribed ectopically in Escherichia coli, consistent with other CsrA-sequestering sRNAs. These results implicate CbsR12 in the indirect regulation of a number of genes via CsrA-mediated regulatory activities. The results also support CbsR12 as a crucial regulatory component early on in a mammalian cell infection.


Subject(s)
Coxiella burnetii/genetics , Q Fever/microbiology , RNA, Bacterial/physiology , RNA, Small Untranslated/physiology , RNA-Binding Proteins/metabolism , Vacuoles/metabolism , Animals , Axenic Culture , Bacterial Proteins/metabolism , Chlorocebus aethiops , Coxiella burnetii/growth & development , Coxiella burnetii/metabolism , Humans , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , THP-1 Cells , Vero Cells
11.
Science ; 365(6456): 919-922, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31346137

ABSTRACT

Rhizobial infection and root nodule formation in legumes require recognition of signal molecules produced by the bacteria and their hosts. Here, we show that rhizobial transfer RNA (tRNA)-derived small RNA fragments (tRFs) are signal molecules that modulate host nodulation. Three families of rhizobial tRFs were confirmed to regulate host genes associated with nodule initiation and development through hijacking the host RNA-interference machinery that involves ARGONAUTE 1. Silencing individual tRFs with the use of short tandem target mimics or by overexpressing their targets represses root hair curling and nodule formation, whereas repressing these targets with artificial microRNAs identical to the respective tRFs or mutating these targets with CRISPR-Cas9 promotes nodulation. Our findings thus uncover a bacterial small RNA-mediated mechanism for prokaryote-eukaryote interaction and may pave the way for enhancing nodulation efficiency in legumes.


Subject(s)
Bradyrhizobium/physiology , Gene Expression Regulation, Plant , Glycine max/microbiology , Host Microbial Interactions/genetics , Plant Root Nodulation/genetics , RNA, Bacterial/physiology , RNA, Small Untranslated/physiology , RNA, Transfer/physiology , Argonaute Proteins/genetics , Bradyrhizobium/genetics , CRISPR-Cas Systems , Nitrogen Fixation , Nucleic Acid Conformation , Plant Proteins/genetics , Plant Roots/metabolism , Plant Roots/microbiology , RNA Interference , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Small Untranslated/chemistry , RNA, Small Untranslated/genetics , RNA, Transfer/chemistry , RNA, Transfer/genetics , Glycine max/genetics , Glycine max/metabolism
12.
Environ Microbiol ; 21(8): 2933-2947, 2019 08.
Article in English | MEDLINE | ID: mdl-31106966

ABSTRACT

The biofilm formation by Pseudomonas aeruginosa highly increases the bacterial resistance to antimicrobial agents and host immune clearance. The biofilm formation is positively regulated by two small RNAs, RsmY and RsmZ. Previously, we reported that mutation in the polynucleotide phosphorylase (PNPase) coding gene pnp increases the levels of RsmY/Z. However, in this study, we found that the biofilm formation is decreased in the pnp mutant, which is due to a defect in rhamnolipids production. The rhamnolipids production is regulated by the RhlI-RhlR quorum sensing system. We found that PNPase influences the translation of RhlI through its 5'-untranslated region (UTR) and identified that the sRNA P27 is responsible for the translational repression. In vitro translation experiments demonstrated that P27 directly represses the translation of the rhlI mRNA through its 5'UTR in an Hfq-dependent manner. Point mutations in the rhlI 5'UTR or P27, which abolish the pairing between the two RNAs restore the rhlI expression and rhamnolipids production as well as the biofilm formation in the pnp mutant. Overall, our results reveal a novel layer of regulation of the Rhl quorum sensing system by the sRNA P27.


Subject(s)
Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Ligases/genetics , Pseudomonas aeruginosa/genetics , Quorum Sensing , RNA, Bacterial/physiology , RNA, Small Untranslated/physiology , Transcription Factors/genetics , Biofilms/growth & development , Glycolipids/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , Protein Biosynthesis , Pseudomonas aeruginosa/enzymology , Quorum Sensing/genetics , RNA Processing, Post-Transcriptional
13.
Biochimie ; 164: 3-16, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30995539

ABSTRACT

Prokaryotes encounter constant and often brutal modifications to their environment. In order to survive, they need to maintain fitness, which includes adapting their protein expression patterns. Many factors control gene expression but this review focuses on just one, namely antisense RNAs (asRNAs), a class of non-coding RNAs (ncRNAs) characterized by their location in cis and their perfect complementarity with their targets. asRNAs were considered for a long time to be trivial and only to be found on mobile genetic elements. However, recent advances in methodology have revealed that their abundance and potential activities have been underestimated. This review aims to illustrate the role of asRNA in various physiologically crucial functions in both archaea and bacteria, which can be regrouped in three categories: cell maintenance, horizontal gene transfer and virulence. A literature survey of asRNAs demonstrates the difficulties to characterize and assign a role to asRNAs. With the aim of facilitating this task, we describe recent technological advances that could be of interest to identify new asRNAs and to discover their function.


Subject(s)
Archaea , Bacteria , Bacterial Physiological Phenomena/genetics , Gene Transfer, Horizontal/genetics , RNA, Antisense , Virulence/genetics , Archaea/genetics , Archaea/pathogenicity , Archaea/physiology , Bacteria/genetics , Bacteria/pathogenicity , Gene Expression Regulation, Archaeal , Gene Expression Regulation, Bacterial , RNA, Antisense/genetics , RNA, Antisense/physiology , RNA, Archaeal/genetics , RNA, Archaeal/physiology , RNA, Bacterial/genetics , RNA, Bacterial/physiology
14.
Biochimie ; 162: 144-155, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31002843

ABSTRACT

RyhB is a key regulator of iron level in Escherichia coli (E. coli), which assists in conserving iron for life-sustaining cellular functions when cytoplasmic levels of the ferrous form of iron is limited. RyhB affects glucose metabolism. Seventy percent of the genes that are regulated by RyhB are related to metabolism. We demonstrated for the first time that the activity of the pentose phosphate pathway increased upon ryhB activation using a13C stable isotope-based technique called METAFoR (Metabolic flux ratio analysis). U-13C glucose-based studies showed that the reversible exchange activity of serine and glycine was enhanced by flux redistribution, which further favors NADPH formation. In addition, Entner-Doudoroff (ED) pathway activity was inhibited in the ryhB-defective cells. Quantitative physiology-based experiments highlighted a significant increase in the levels of reactive oxygen species (ROS) in ryhB-induced W3100 E. coli cells in batch culture. A simultaneous decrease in NADH/NAD+ and NADPH/NADP+ ratios outlined the potentially direct roles of NADH and NADPH in antagonizing the excess ROS formed after ryhB activation. Our observations offer a new perspective regarding the roles of RyhB and highlight that this small RNA can significantly affect cell metabolism in addition to its role as a regulator of gene expression.


Subject(s)
Escherichia coli/metabolism , Iron/metabolism , NAD/metabolism , Pentose Phosphate Pathway , RNA, Bacterial/physiology , RNA, Small Untranslated/physiology , Reactive Oxygen Species/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial
15.
J Appl Microbiol ; 127(4): 968-984, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30927378

ABSTRACT

Synthetic biology requires the design and implementation of novel enzymes, genetic circuits or even entire cells, which can be controlled by the user. RNA-based regulatory elements have many important functional properties in this regard, such as their modular nature and their ability to respond to specific external stimuli. These properties have led to the widespread exploration of their use as gene regulation devices in synthetic biology. In this review, we focus on two major types of RNA elements: riboswitches and RNA thermometers (RNATs). We describe their general structure and function, before discussing their potential uses in synthetic biology (e.g. in the production of biofuels and biodegradable plastics). We also discuss their limitations, and novel strategies to implement RNA-based regulatory devices in biotechnological applications. We close with a description of some common model organisms used in synthetic biology, with a focus on the current applications and limitations of RNA-based regulation.


Subject(s)
Biotechnology/methods , Gene Expression Regulation , RNA/physiology , Synthetic Biology/methods , Bacteria/genetics , Bacteria/metabolism , RNA/chemistry , RNA, Bacterial/chemistry , RNA, Bacterial/physiology , Riboswitch , Temperature
16.
J Fish Dis ; 42(2): 277-291, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30488970

ABSTRACT

Vibrio alginolyticus as an important pathogen in aquaculture can encounter the oxidative stress produced by the immune system during infection. Previous studies showed that sRNAs have important functions in response to oxidative stress in bacteria; however, less of sRNAs related to oxidative stress response were identified in V. alginolyticus. In this study, a total of 749 novel sRNAs were identified by RNA sequencing; among them, 128 sRNAs were up- or downregulated in response to oxidative stress. In addition, 1,870 genes exhibited variation on mRNA levels in oxidative stress response. By analysing the target genes of the sRNAs, we concluded that these sRNAs could regulate expressions of genes responsible for iron transport, catalase, GSH-dependent defence system, electron transferred and stress response. Moreover, the functions of the sRNAs are also seemed related to the pathogenicity in V. alginolyticus. Based on the results, we constructed the oxidative stress model in V. alginolyticus. This study provides us the first outlook of sRNAs function in oxidative stress response in V. alginolyticus. Furthermore, this study can help us to prevent and control this important opportunistic pathogen in aquaculture.


Subject(s)
Oxidative Stress/physiology , RNA, Bacterial/physiology , RNA, Small Untranslated/physiology , Vibrio alginolyticus/physiology , Gene Expression Profiling , Oxidative Stress/genetics , RNA, Bacterial/genetics , RNA, Messenger , RNA, Small Untranslated/genetics , Sequence Analysis, RNA , Vibrio alginolyticus/genetics
17.
Mol Cell ; 72(3): 541-552.e6, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388413

ABSTRACT

Numerous classes of riboswitches have been found to regulate bacterial gene expression in response to physiological cues, offering new paths to antibacterial drugs. As common studies of isolated riboswitches lack the functional context of the transcription machinery, we here combine single-molecule, biochemical, and simulation approaches to investigate the coupling between co-transcriptional folding of the pseudoknot-structured preQ1 riboswitch and RNA polymerase (RNAP) pausing. We show that pausing at a site immediately downstream of the riboswitch requires a ligand-free pseudoknot in the nascent RNA, a precisely spaced sequence resembling the pause consensus, and electrostatic and steric interactions with the RNAP exit channel. While interactions with RNAP stabilize the native fold of the riboswitch, binding of the ligand signals RNAP release from the pause. Our results demonstrate that the nascent riboswitch and its ligand actively modulate the function of RNAP and vice versa, a paradigm likely to apply to other cellular RNA transcripts.


Subject(s)
DNA-Directed RNA Polymerases/physiology , Nucleoside Q/physiology , Riboswitch/physiology , Aptamers, Nucleotide , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Fluorescence Resonance Energy Transfer/methods , Gene Expression Regulation, Bacterial , Ligands , Nucleic Acid Conformation , Nucleoside Q/metabolism , Protein Folding , RNA Folding , RNA, Bacterial/physiology , Riboswitch/genetics , Single Molecule Imaging , Transcription, Genetic/physiology
18.
World J Microbiol Biotechnol ; 34(11): 171, 2018 Nov 09.
Article in English | MEDLINE | ID: mdl-30413889

ABSTRACT

Bacterial genomes contain a huge amount of different genes. These genes are spatiotemporally expressed to accomplish some required functions within the organism. Inside the cell, any step of gene expression may be modulated at four possible places such as transcription initiation, translation regulation, mRNA stability and protein stability. To achieve this, there is a necessity of strong regulators either natural or synthetic which can fine-tune gene expression regarding the required function. In recent years, riboswitches as metabolite responsive control elements residing in the untranslated regions of certain messenger RNAs, have been known to control gene expression at transcription or translation level. Importantly, these control elements do not prescribe the involvement of protein factors for metabolite binding. However, they own their particular properties to sense intramolecular metabolites (ligands). Herein, we highlighted current important bacterial riboswitches, their applications to support genetic control, ligand-binding domain mechanisms and current progress in synthetic riboswitches.


Subject(s)
Gene Expression Regulation, Bacterial , RNA, Bacterial/physiology , Riboswitch/physiology , Aptamers, Nucleotide/metabolism , Aptamers, Peptide/metabolism , Bacteria/genetics , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Genes, Bacterial/genetics , Genes, Bacterial/physiology , Glycine/metabolism , Ligands , Pyrimidinones/metabolism , Pyrroles/metabolism , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , Riboswitch/genetics
19.
Microbiol Spectr ; 6(5)2018 09.
Article in English | MEDLINE | ID: mdl-30191804

ABSTRACT

Diverse mechanisms and functions of posttranscriptional regulation by small regulatory RNAs and RNA-binding proteins have been described in bacteria. In contrast, little is known about the spatial organization of RNAs in bacterial cells. In eukaryotes, subcellular localization and transport of RNAs play important roles in diverse physiological processes, such as embryonic patterning, asymmetric cell division, epithelial polarity, and neuronal plasticity. It is now clear that bacterial RNAs also can accumulate at distinct sites in the cell. However, due to the small size of bacterial cells, RNA localization and localization-associated functions are more challenging to study in bacterial cells, and the underlying molecular mechanisms of transcript localization are less understood. Here, we review the emerging examples of RNAs localized to specific subcellular locations in bacteria, with indications that subcellular localization of transcripts might be important for gene expression and regulatory processes. Diverse mechanisms for bacterial RNA localization have been suggested, including close association to their genomic site of transcription, or to the localizations of their protein products in translation-dependent or -independent processes. We also provide an overview of the state of the art of technologies to visualize and track bacterial RNAs, ranging from hybridization-based approaches in fixed cells to in vivo imaging approaches using fluorescent protein reporters and/or RNA aptamers in single living bacterial cells. We conclude with a discussion of open questions in the field and ongoing technological developments regarding RNA imaging in eukaryotic systems that might likewise provide novel insights into RNA localization in bacteria.


Subject(s)
Molecular Imaging/methods , RNA Transport , RNA, Bacterial/ultrastructure , Staining and Labeling/methods , Aptamers, Nucleotide/chemistry , Bacteria/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Fluorescent Dyes , Gene Expression Regulation, Bacterial , Green Fluorescent Proteins , In Situ Hybridization, Fluorescence/methods , Microscopy, Fluorescence , Protein Transport , RNA Transport/physiology , RNA, Bacterial/genetics , RNA, Bacterial/physiology , RNA-Binding Proteins/physiology , Recombinant Fusion Proteins , Ribonucleases , Transcription, Genetic
20.
Microbiol Spectr ; 6(4)2018 07.
Article in English | MEDLINE | ID: mdl-30003867

ABSTRACT

Bacterial regulatory RNAs are key players in adaptation to changing environmental conditions and response to diverse cellular stresses. However, while regulatory RNAs of bacterial pathogens have been intensely studied under defined conditions in vitro, characterization of their role during the infection of eukaryotic host organisms is lagging behind. This review summarizes our current understanding of the contribution of the different classes of regulatory RNAs and RNA-binding proteins to bacterial virulence and illustrates their role in infection by reviewing the mechanisms of some prominent representatives of each class. Emerging technologies are described that bear great potential for global, unbiased studies of virulence-related RNAs in bacterial model and nonmodel pathogens in the future. The review concludes by deducing common principles of RNA-mediated gene expression control of virulence programs in different pathogens, and by defining important open questions for upcoming research in the field.


Subject(s)
Bacteria/pathogenicity , Host Microbial Interactions/physiology , RNA, Bacterial/physiology , RNA-Binding Proteins/physiology , Virulence Factors/physiology , Adaptation, Physiological , Bacteria/genetics , Bacterial Physiological Phenomena , Bacterial Proteins/physiology , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Expression Regulation, Bacterial , Genes, Bacterial/genetics , RNA, Bacterial/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/physiology , Virulence/genetics , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL