Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.296
Filter
Add more filters

Publication year range
1.
Cell ; 167(1): 111-121.e13, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27662085

ABSTRACT

Bacterial small RNAs (sRNAs) have been implicated in various aspects of post-transcriptional gene regulation. Here, we demonstrate that sRNAs also act at the level of transcription termination. We use the rpoS gene, which encodes a general stress sigma factor σ(S), as a model system, and show that sRNAs DsrA, ArcZ, and RprA bind the rpoS 5'UTR to suppress premature Rho-dependent transcription termination, both in vitro and in vivo. sRNA-mediated antitermination markedly stimulates transcription of rpoS during the transition to the stationary phase of growth, thereby facilitating a rapid adjustment of bacteria to global metabolic changes. Next generation RNA sequencing and bioinformatic analysis indicate that Rho functions as a global "attenuator" of transcription, acting at the 5'UTR of hundreds of bacterial genes, and that its suppression by sRNAs is a widespread mode of bacterial gene regulation.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , RNA, Small Untranslated/metabolism , Sigma Factor/metabolism , Transcription Termination, Genetic , 5' Untranslated Regions
2.
Mol Cell ; 83(22): 4158-4173.e7, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37949068

ABSTRACT

Sporulating bacteria can retreat into long-lasting dormant spores that preserve the capacity to germinate when propitious. However, how the revival transcriptional program is memorized for years remains elusive. We revealed that in dormant spores, core RNA polymerase (RNAP) resides in a central chromosomal domain, where it remains bound to a subset of intergenic promoter regions. These regions regulate genes encoding for most essential cellular functions, such as rRNAs and tRNAs. Upon awakening, RNAP recruits key transcriptional components, including sigma factor, and progresses to express the adjacent downstream genes. Mutants devoid of spore DNA-compacting proteins exhibit scattered RNAP localization and subsequently disordered firing of gene expression during germination. Accordingly, we propose that the spore chromosome is structured to preserve the transcriptional program by halting RNAP, prepared to execute transcription at the auspicious time. Such a mechanism may sustain long-term transcriptional programs in diverse organisms displaying a quiescent life form.


Subject(s)
Bacillus subtilis , Spores, Bacterial , Spores, Bacterial/genetics , Spores, Bacterial/metabolism , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Sigma Factor/genetics , Sigma Factor/metabolism , Promoter Regions, Genetic , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism
3.
Nature ; 627(8003): 424-430, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38418874

ABSTRACT

Mycobacterium tuberculosis (Mtb) is a bacterial pathogen that causes tuberculosis (TB), an infectious disease that is responsible for major health and economic costs worldwide1. Mtb encounters diverse environments during its life cycle and responds to these changes largely by reprogramming its transcriptional output2. However, the mechanisms of Mtb transcription and how they are regulated remain poorly understood. Here we use a sequencing method that simultaneously determines both termini of individual RNA molecules in bacterial cells3 to profile the Mtb transcriptome at high resolution. Unexpectedly, we find that most Mtb transcripts are incomplete, with their 5' ends aligned at transcription start sites and 3' ends located 200-500 nucleotides downstream. We show that these short RNAs are mainly associated with paused RNA polymerases (RNAPs) rather than being products of premature termination. We further show that the high propensity of Mtb RNAP to pause early in transcription relies on the binding of the σ-factor. Finally, we show that a translating ribosome promotes transcription elongation, revealing a potential role for transcription-translation coupling in controlling Mtb gene expression. In sum, our findings depict a mycobacterial transcriptome that prominently features incomplete transcripts resulting from RNAP pausing. We propose that the pausing phase constitutes an important transcriptional checkpoint in Mtb that allows the bacterium to adapt to environmental changes and could be exploited for TB therapeutics.


Subject(s)
Gene Expression Regulation, Bacterial , Mycobacterium tuberculosis , RNA, Bacterial , Transcriptome , DNA-Directed RNA Polymerases/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , RNA, Bacterial/analysis , RNA, Bacterial/biosynthesis , RNA, Bacterial/genetics , Transcriptome/genetics , Tuberculosis/microbiology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcription Initiation Site , Sigma Factor/metabolism , Ribosomes/metabolism , Protein Biosynthesis
4.
Nature ; 614(7946): 168-174, 2023 02.
Article in English | MEDLINE | ID: mdl-36423657

ABSTRACT

CRISPR defence systems such as the well-known DNA-targeting Cas9 and the RNA-targeting type III systems are widespread in prokaryotes1,2. The latter orchestrates a complex antiviral response that is initiated through the synthesis of cyclic oligoadenylates after recognition of foreign RNA3-5. Among the large set of proteins that are linked to type III systems and predicted to bind cyclic oligoadenylates6,7, a CRISPR-associated Lon protease (CalpL) stood out to us. CalpL contains a sensor domain of the SAVED family7 fused to a Lon protease effector domain. However, the mode of action of this effector is unknown. Here we report the structure and function of CalpL and show that this soluble protein forms a stable tripartite complex with two other proteins, CalpT and CalpS, that are encoded on the same operon. After activation by cyclic tetra-adenylate (cA4), CalpL oligomerizes and specifically cleaves the MazF homologue CalpT, which releases the extracytoplasmic function σ factor CalpS from the complex. Our data provide a direct connection between CRISPR-based detection of foreign nucleic acids and transcriptional regulation. Furthermore, the presence of a SAVED domain that binds cyclic tetra-adenylate in a CRISPR effector reveals a link to the cyclic-oligonucleotide-based antiphage signalling system.


Subject(s)
Bacteria , Bacteriophages , CRISPR-Associated Proteins , CRISPR-Cas Systems , Nucleotides, Cyclic , Protease La , Bacteria/enzymology , Bacteria/immunology , Bacteria/metabolism , Bacteria/virology , Bacteriophages/immunology , Bacteriophages/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/physiology , Cyclic AMP/analogs & derivatives , Cyclic AMP/chemistry , Enzyme Activation , Gene Expression Regulation, Bacterial , Nucleotides, Cyclic/immunology , Nucleotides, Cyclic/metabolism , Operon , Protease La/chemistry , Protease La/metabolism , RNA, Viral , Sigma Factor , Transcription, Genetic
5.
Mol Cell ; 81(1): 139-152.e10, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33217319

ABSTRACT

The bacterium Francisella tularensis (Ft) is one of the most infectious agents known. Ft virulence is controlled by a unique combination of transcription regulators: the MglA-SspA heterodimer, PigR, and the stress signal, ppGpp. MglA-SspA assembles with the σ70-associated RNAP holoenzyme (RNAPσ70), forming a virulence-specialized polymerase. These factors activate Francisella pathogenicity island (FPI) gene expression, which is required for virulence, but the mechanism is unknown. Here we report FtRNAPσ70-promoter-DNA, FtRNAPσ70-(MglA-SspA)-promoter DNA, and FtRNAPσ70-(MglA-SspA)-ppGpp-PigR-promoter DNA cryo-EM structures. Structural and genetic analyses show MglA-SspA facilitates σ70 binding to DNA to regulate virulence and virulence-enhancing genes. Our Escherichia coli RNAPσ70-homodimeric EcSspA structure suggests this is a general SspA-transcription regulation mechanism. Strikingly, our FtRNAPσ70-(MglA-SspA)-ppGpp-PigR-DNA structure reveals ppGpp binding to MglA-SspA tethers PigR to promoters. PigR in turn recruits FtRNAP αCTDs to DNA UP elements. Thus, these studies unveil a unique mechanism for Ft pathogenesis involving a virulence-specialized RNAP that employs two (MglA-SspA)-based strategies to activate virulence genes.


Subject(s)
DNA-Directed RNA Polymerases , Francisella tularensis , Promoter Regions, Genetic , Sigma Factor , Virulence Factors , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Francisella tularensis/genetics , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , Sigma Factor/genetics , Sigma Factor/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
6.
Mol Cell ; 81(1): 8-9, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33417856

ABSTRACT

Travis et al. (2020) reveal how Francisella tularensis uses stress-induced ppGpp to activate its virulent pathogenesis program by tethering an αCTD-DNA organizer (PigR) to a σ-organizing heterodimer (MglA-SspA), highlighting the remarkable diversity of transcriptional mechanisms in under-studied bacteria.


Subject(s)
Francisella tularensis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Francisella tularensis/genetics , Francisella tularensis/metabolism , Gene Expression Regulation, Bacterial , Guanosine Pentaphosphate , Sigma Factor/genetics , Virulence
7.
Mol Cell ; 81(14): 2887-2900.e5, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34171298

ABSTRACT

WhiB7 represents a distinct subclass of transcription factors in the WhiB-Like (Wbl) family, a unique group of iron-sulfur (4Fe-4S] cluster-containing proteins exclusive to the phylum of Actinobacteria. In Mycobacterium tuberculosis (Mtb), WhiB7 interacts with domain 4 of the primary sigma factor (σA4) in the RNA polymerase holoenzyme and activates genes involved in multiple drug resistance and redox homeostasis. Here, we report crystal structures of the WhiB7:σA4 complex alone and bound to its target promoter DNA at 1.55-Å and 2.6-Å resolution, respectively. These structures show how WhiB7 regulates gene expression by interacting with both σA4 and the AT-rich sequence upstream of the -35 promoter DNA via its C-terminal DNA-binding motif, the AT-hook. By combining comparative structural analysis of the two high-resolution σA4-bound Wbl structures with molecular and biochemical approaches, we identify the structural basis of the functional divergence between the two distinct subclasses of Wbl proteins in Mtb.


Subject(s)
Bacterial Proteins/metabolism , Iron-Sulfur Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Transcription Factors/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial/genetics , Iron-Sulfur Proteins/genetics , Mycobacterium tuberculosis/genetics , Promoter Regions, Genetic/genetics , Sigma Factor/genetics , Sigma Factor/metabolism , Transcription Factors/genetics
8.
Mol Cell ; 81(14): 2875-2886.e5, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34171296

ABSTRACT

In pathogenic mycobacteria, transcriptional responses to antibiotics result in induced antibiotic resistance. WhiB7 belongs to the Actinobacteria-specific family of Fe-S-containing transcription factors and plays a crucial role in inducible antibiotic resistance in mycobacteria. Here, we present cryoelectron microscopy structures of Mycobacterium tuberculosis transcriptional regulatory complexes comprising RNA polymerase σA-holoenzyme, global regulators CarD and RbpA, and WhiB7, bound to a WhiB7-regulated promoter. The structures reveal how WhiB7 interacts with σA-holoenzyme while simultaneously interacting with an AT-rich sequence element via its AT-hook. Evidently, AT-hooks, rare elements in bacteria yet prevalent in eukaryotes, bind to target AT-rich DNA sequences similarly to the nuclear chromosome binding proteins. Unexpectedly, a subset of particles contained a WhiB7-stabilized closed promoter complex, revealing this intermediate's structure, and we apply kinetic modeling and biochemical assays to rationalize how WhiB7 activates transcription. Altogether, our work presents a comprehensive view of how WhiB7 serves to activate gene expression leading to antibiotic resistance.


Subject(s)
Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial/genetics , Intrinsic Factor/genetics , Mycobacterium tuberculosis/genetics , Transcription Factors/genetics , Transcriptional Activation/genetics , Anti-Bacterial Agents/pharmacology , Cryoelectron Microscopy/methods , DNA-Directed RNA Polymerases/genetics , Gene Expression Regulation, Bacterial/genetics , Mycobacterium tuberculosis/drug effects , Promoter Regions, Genetic/genetics , Sigma Factor/genetics
9.
Annu Rev Microbiol ; 77: 625-644, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37437215

ABSTRACT

Responding to environmental cues is a prerequisite for survival in the microbial world. Extracytoplasmic function σ factors (ECFs) represent the third most abundant and by far the most diverse type of bacterial signal transduction. While archetypal ECFs are controlled by cognate anti-σ factors, comprehensive comparative genomics efforts have revealed a much higher abundance and regulatory diversity of ECF regulation than previously appreciated. They have also uncovered a diverse range of anti-σ factor-independent modes of controlling ECF activity, including fused regulatory domains and phosphorylation-dependent mechanisms. While our understanding of ECF diversity is comprehensive for well-represented and heavily studied bacterial phyla-such as Proteobacteria, Firmicutes, and Actinobacteria (phylum Actinomycetota)-our current knowledge about ECF-dependent signaling in the vast majority of underrepresented phyla is still far from complete. In particular, the dramatic extension of bacterial diversity in the course of metagenomic studies represents both a new challenge and an opportunity in expanding the world of ECF-dependent signal transduction.


Subject(s)
Bacterial Proteins , Sigma Factor , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Sigma Factor/genetics , Sigma Factor/metabolism , Bacteria/genetics , Bacteria/metabolism , Signal Transduction , Gene Expression Regulation, Bacterial
11.
Cell ; 154(3): 623-36, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23911325

ABSTRACT

The decision to initiate DNA replication is a critical step in the cell cycle of all organisms. Cells often delay replication in the face of stressful conditions, but the underlying mechanisms remain incompletely defined. Here, we demonstrate in Caulobacter crescentus that proteotoxic stress induces a cell-cycle arrest by triggering the degradation of DnaA, the conserved replication initiator. A depletion of available Hsp70 chaperone, DnaK, either through genetic manipulation or heat shock, induces synthesis of the Lon protease, which can directly degrade DnaA. Unexpectedly, we find that unfolded proteins, which accumulate following a loss of DnaK, also allosterically activate Lon to degrade DnaA, thereby ensuring a cell-cycle arrest. Our work reveals a mechanism for regulating DNA replication under adverse growth conditions. Additionally, our data indicate that unfolded proteins can actively and directly alter substrate recognition by cellular proteases.


Subject(s)
Bacterial Proteins/metabolism , Caulobacter crescentus/cytology , Caulobacter crescentus/physiology , DNA Replication , DNA-Binding Proteins/metabolism , Protease La/metabolism , Bacterial Proteins/genetics , Caulobacter crescentus/genetics , DNA, Bacterial/metabolism , DNA-Binding Proteins/genetics , Escherichia coli/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response , Molecular Chaperones/metabolism , Protein Folding , Sigma Factor/metabolism , Stress, Physiological
12.
Mol Cell ; 77(3): 443-445, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32032510

ABSTRACT

A recent paper by Gallagher et al. (2020) demonstrates that c-di-GMP controls spore formation in Streptomyces venezuelae through sequestering the sporulation sigma factor σWhiG and presents the crystal structure of a ternary complex between c-di-GMP, σWhiG, and its anti-sigma factor, RsiG.


Subject(s)
Streptomyces , Bacterial Proteins , Cell Differentiation , Cyclic GMP/analogs & derivatives , Sigma Factor
13.
Mol Cell ; 77(3): 586-599.e6, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31810759

ABSTRACT

Streptomyces are our primary source of antibiotics, produced concomitantly with the transition from vegetative growth to sporulation in a complex developmental life cycle. We previously showed that the signaling molecule c-di-GMP binds BldD, a master repressor, to control initiation of development. Here we demonstrate that c-di-GMP also intervenes later in development to control differentiation of the reproductive hyphae into spores by arming a novel anti-σ (RsiG) to bind and sequester a sporulation-specific σ factor (σWhiG). We present the structure of the RsiG-(c-di-GMP)2-σWhiG complex, revealing an unusual, partially intercalated c-di-GMP dimer bound at the RsiG-σWhiG interface. RsiG binds c-di-GMP in the absence of σWhiG, employing a novel E(X)3S(X)2R(X)3Q(X)3D motif repeated on each helix of a coiled coil. Further studies demonstrate that c-di-GMP is essential for RsiG to inhibit σWhiG. These findings reveal a newly described control mechanism for σ-anti-σ complex formation and establish c-di-GMP as the central integrator of Streptomyces development.


Subject(s)
Cyclic GMP/analogs & derivatives , Sigma Factor/metabolism , Streptomyces/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Cyclic GMP/metabolism , Cyclic GMP/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Protein Domains , RNA, Bacterial/metabolism , Spores, Bacterial/metabolism , Streptomyces/genetics
14.
Cell ; 148(4): 679-89, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22341441

ABSTRACT

Understanding the pathway and kinetic mechanisms of transcription initiation is essential for quantitative understanding of gene regulation, but initiation is a multistep process, the features of which can be obscured in bulk analysis. We used a multiwavelength single-molecule fluorescence colocalization approach, CoSMoS, to define the initiation pathway at an activator-dependent bacterial σ(54) promoter that recapitulates characteristic features of eukaryotic promoters activated by enhancer binding proteins. The experiments kinetically characterize all major steps of the initiation process, revealing heretofore unknown features, including reversible formation of two closed complexes with greatly differing stabilities, multiple attempts for each successful formation of an open complex, and efficient release of σ(54) from the polymerase core at the start of transcript synthesis. Open complexes are committed to transcription, suggesting that regulation likely targets earlier steps in the mechanism. CoSMoS is a powerful, generally applicable method to elucidate the mechanisms of transcription and other multistep biochemical processes.


Subject(s)
Promoter Regions, Genetic , Sigma Factor/metabolism , Spectrum Analysis/methods , Transcription, Genetic , Base Sequence , DNA-Directed RNA Polymerases/metabolism , Gene Expression Regulation, Bacterial , Kinetics , Molecular Sequence Data , Salmonella typhimurium/metabolism
15.
Cell ; 151(5): 1005-16, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23178120

ABSTRACT

Phage G1 gp67 is a 23 kDa protein that binds to the Staphylococcus aureus (Sau) RNA polymerase (RNAP) σ(A) subunit and blocks cell growth by inhibiting transcription. We show that gp67 has little to no effect on transcription from most promoters but is a potent inhibitor of ribosomal RNA transcription. A 2.0-Å-resolution crystal structure of the complex between gp67 and Sau σ(A) domain 4 (σ(A)(4)) explains how gp67 joins the RNAP promoter complex through σ(A)(4) without significantly affecting σ(A)(4) function. Our results indicate that gp67 forms a complex with RNAP at most, if not all, σ(A)-dependent promoters, but selectively inhibits promoters that depend on an interaction between upstream DNA and the RNAP α-subunit C-terminal domain (αCTD). Thus, we reveal a promoter-specific transcription inhibition mechanism by which gp67 interacts with the RNAP promoter complex through one subunit (σ(A)), and selectively affects the function of another subunit (αCTD) depending on promoter usage.


Subject(s)
Growth Inhibitors/metabolism , Myoviridae/metabolism , Promoter Regions, Genetic , Staphylococcus aureus/growth & development , Staphylococcus aureus/virology , Viral Proteins/metabolism , Base Sequence , Crystallography, X-Ray , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/metabolism , Models, Molecular , Molecular Sequence Data , RNA, Bacterial/genetics , RNA, Ribosomal/genetics , Sigma Factor/metabolism , Staphylococcus aureus/genetics , Transcription, Genetic
16.
Mol Cell ; 74(4): 785-800.e7, 2019 05 16.
Article in English | MEDLINE | ID: mdl-30948267

ABSTRACT

Antibiotics can induce mutations that cause antibiotic resistance. Yet, despite their importance, mechanisms of antibiotic-promoted mutagenesis remain elusive. We report that the fluoroquinolone antibiotic ciprofloxacin (cipro) induces mutations by triggering transient differentiation of a mutant-generating cell subpopulation, using reactive oxygen species (ROS). Cipro-induced DNA breaks activate the Escherichia coli SOS DNA-damage response and error-prone DNA polymerases in all cells. However, mutagenesis is limited to a cell subpopulation in which electron transfer together with SOS induce ROS, which activate the sigma-S (σS) general-stress response, which allows mutagenic DNA-break repair. When sorted, this small σS-response-"on" subpopulation produces most antibiotic cross-resistant mutants. A U.S. Food and Drug Administration (FDA)-approved drug prevents σS induction, specifically inhibiting antibiotic-promoted mutagenesis. Further, SOS-inhibited cell division, which causes multi-chromosome cells, promotes mutagenesis. The data support a model in which within-cell chromosome cooperation together with development of a "gambler" cell subpopulation promote resistance evolution without risking most cells.


Subject(s)
Anti-Bacterial Agents/adverse effects , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Mutagenesis/genetics , Cell Division/drug effects , Ciprofloxacin/adverse effects , DNA Damage/drug effects , DNA-Directed DNA Polymerase/genetics , Drug Resistance, Bacterial/drug effects , Escherichia coli/drug effects , Escherichia coli/pathogenicity , Gene Expression Regulation, Bacterial/drug effects , Mutagenesis/drug effects , Mutation , Reactive Oxygen Species/metabolism , SOS Response, Genetics/drug effects , Sigma Factor/genetics
17.
Proc Natl Acad Sci U S A ; 121(10): e2321910121, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38422018

ABSTRACT

Bacteroidota are abundant members of the human gut microbiota that shape the enteric landscape by modulating host immunity and degrading dietary- and host-derived glycans. These processes are mediated in part by Outer Membrane Vesicles (OMVs). Here, we developed a high-throughput screen to identify genes required for OMV biogenesis and its regulation in Bacteroides thetaiotaomicron (Bt). We identified a family of Dual membrane-spanning anti-sigma factors (Dma) that control OMV biogenesis. We conducted molecular and multiomic analyses to demonstrate that deletion of Dma1, the founding member of the Dma family, modulates OMV production by controlling the activity of the ECF21 family sigma factor, Das1, and its downstream regulon. Dma1 has a previously uncharacterized domain organization that enables Dma1 to span both the inner and outer membrane of Bt. Phylogenetic analyses reveal that this common feature of the Dma family is restricted to the phylum Bacteroidota. This study provides mechanistic insights into the regulation of OMV biogenesis in human gut bacteria.


Subject(s)
Bacteroides thetaiotaomicron , Humans , Bacteroides thetaiotaomicron/genetics , Sigma Factor , Phylogeny
18.
PLoS Genet ; 20(3): e1011142, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38457455

ABSTRACT

Succinate is a potent immune signalling molecule that is present in the mammalian gut and within macrophages. Both of these infection niches are colonised by the pathogenic bacterium Salmonella enterica serovar Typhimurium during infection. Succinate is a C4-dicarboyxlate that can serve as a source of carbon for bacteria. When succinate is provided as the sole carbon source for in vitro cultivation, Salmonella and other enteric bacteria exhibit a slow growth rate and a long lag phase. This growth inhibition phenomenon was known to involve the sigma factor RpoS, but the genetic basis of the repression of bacterial succinate utilisation was poorly understood. Here, we use an experimental evolution approach to isolate fast-growing mutants during growth of S. Typhimurium on succinate containing minimal medium. Our approach reveals novel RpoS-independent systems that inhibit succinate utilisation. The CspC RNA binding protein restricts succinate utilisation, an inhibition that is antagonised by high levels of the small regulatory RNA (sRNA) OxyS. We discovered that the Fe-S cluster regulatory protein IscR inhibits succinate utilisation by repressing the C4-dicarboyxlate transporter DctA. Furthermore, the ribose operon repressor RbsR is required for the complete RpoS-driven repression of succinate utilisation, suggesting a novel mechanism of RpoS regulation. Our discoveries shed light on the redundant regulatory systems that tightly regulate the utilisation of succinate. We speculate that the control of central carbon metabolism by multiple regulatory systems in Salmonella governs the infection niche-specific utilisation of succinate.


Subject(s)
Bacterial Proteins , Succinic Acid , Animals , Bacterial Proteins/metabolism , Succinic Acid/metabolism , Salmonella typhimurium/genetics , Succinates/metabolism , Carbon/metabolism , Sigma Factor/genetics , Sigma Factor/metabolism , Gene Expression Regulation, Bacterial , Mammals/metabolism
19.
Proc Natl Acad Sci U S A ; 121(13): e2400584121, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38502707

ABSTRACT

When faced with starvation, the bacterium Bacillus subtilis transforms itself into a dormant cell type called a "spore". Sporulation initiates with an asymmetric division event, which requires the relocation of the core divisome components FtsA and FtsZ, after which the sigma factor σF is exclusively activated in the smaller daughter cell. Compartment-specific activation of σF requires the SpoIIE phosphatase, which displays a biased localization on one side of the asymmetric division septum and associates with the structural protein DivIVA, but the mechanism by which this preferential localization is achieved is unclear. Here, we isolated a variant of DivIVA that indiscriminately activates σF in both daughter cells due to promiscuous localization of SpoIIE, which was corrected by overproduction of FtsA and FtsZ. We propose that the core components of the redeployed cell division machinery drive the asymmetric localization of DivIVA and SpoIIE to trigger the initiation of the sporulation program.


Subject(s)
Bacillus subtilis , Bacterial Proteins , Bacillus subtilis/metabolism , Transcriptional Activation , Bacterial Proteins/metabolism , Spores, Bacterial/genetics , Spores, Bacterial/metabolism , Cell Division/genetics , Sigma Factor/genetics , Sigma Factor/metabolism
20.
PLoS Genet ; 20(6): e1011127, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38829907

ABSTRACT

The cell envelope fortifies bacterial cells against antibiotics and other insults. Species in the Mycobacteriales order have a complex envelope that includes an outer layer of mycolic acids called the mycomembrane (MM) and a cell wall composed of peptidoglycan and arabinogalactan. This envelope architecture is unique among bacteria and contributes significantly to the virulence of pathogenic Mycobacteriales like Mycobacterium tuberculosis. Characterization of pathways that govern envelope biogenesis in these organisms is therefore critical in understanding their biology and for identifying new antibiotic targets. To better understand MM biogenesis, we developed a cell sorting-based screen for mutants defective in the surface exposure of a porin normally embedded in the MM of the model organism Corynebacterium glutamicum. The results revealed a requirement for the conserved σD envelope stress response in porin export and identified MarP as the site-1 protease, respectively, that activate the response by cleaving the membrane-embedded anti-sigma factor. A reporter system revealed that the σD pathway responds to defects in mycolic acid and arabinogalactan biosynthesis, suggesting that the stress response has the unusual property of being induced by activating signals that arise from defects in the assembly of two distinct envelope layers. Our results thus provide new insights into how C. glutamicum and related bacteria monitor envelope integrity and suggest a potential role for members of the σD regulon in protein export to the MM.


Subject(s)
Cell Membrane , Cell Wall , Corynebacterium glutamicum , Mycolic Acids , Sigma Factor , Cell Wall/metabolism , Cell Wall/genetics , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Mycolic Acids/metabolism , Sigma Factor/metabolism , Sigma Factor/genetics , Cell Membrane/metabolism , Stress, Physiological , Porins/metabolism , Porins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Galactans/metabolism , Gene Expression Regulation, Bacterial , Peptidoglycan/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL