Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
PNAS Nexus ; 3(4): pgae154, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38650860

ABSTRACT

In response to nutrient deprivation, bacteria activate a conserved stress response pathway called the stringent response (SR). During SR activation in Caulobacter crescentus, SpoT synthesizes the secondary messengers guanosine 5'-diphosphate 3'-diphosphate and guanosine 5'-triphosphate 3'-diphosphate (collectively known as (p)ppGpp), which affect transcription by binding RNA polymerase (RNAP) to down-regulate anabolic genes. (p)ppGpp also impacts the expression of anabolic genes by controlling the levels and activities of their transcriptional regulators. In Caulobacter, a major regulator of anabolic genes is the transcription factor CdnL. If and how CdnL is controlled during the SR and why that might be functionally important are unclear. In this study, we show that CdnL is down-regulated posttranslationally during starvation in a manner dependent on SpoT and the ClpXP protease. Artificial stabilization of CdnL during starvation causes misregulation of ribosomal and metabolic genes. Functionally, we demonstrate that the combined action of SR transcriptional regulators and CdnL clearance allows for rapid adaptation to nutrient repletion. Moreover, cells that are unable to clear CdnL during starvation are outcompeted by wild-type cells when subjected to nutrient fluctuations. We hypothesize that clearance of CdnL during the SR, in conjunction with direct binding of (p)ppGpp and DksA to RNAP, is critical for altering the transcriptome in order to permit cell survival during nutrient stress.

2.
mBio ; 15(4): e0315323, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38511926

ABSTRACT

The alphaproteobacterium Caulobacter crescentus thrives in oligotrophic environments and is able to optimally exploit minimal resources by entertaining an intricate network of gene expression control mechanisms. Numerous transcriptional activators and repressors have been reported to contribute to these processes, but only few studies have focused on regulation at the post-transcriptional level in C. crescentus. Small RNAs (sRNAs) are a prominent class of regulators of bacterial gene expression, and most sRNAs characterized today engage in direct base-pairing interactions to modulate the translation and/or stability of target mRNAs. In many cases, the ubiquitous RNA chaperone, Hfq, contributes to the establishment of RNA-RNA interactions. Although the deletion of the hfq gene is associated with a severe loss of fitness in C. crescentus, the RNA ligands of the chaperone have remained largely unexplored. Here we report on the identification of coding and non-coding transcripts associated with Hfq in C. crescentus and demonstrate Hfq-dependent post-transcriptional regulation in this organism. We show that the Hfq-bound sRNA RusT is transcriptionally controlled by the NtrYX two-component system and induced in response to iron starvation. By combining RusT pulse expression with whole-genome transcriptome analysis, we determine 16 candidate target transcripts that are deregulated, many of which encode outer membrane transporters. We hence suggest RusT to support remodeling of the C. crescentus cell surface when iron supplies are limited.IMPORTANCEThe conserved RNA-binding protein Hfq contributes significantly to the adaptation of bacteria to different environmental conditions. Hfq not only stabilizes associated sRNAs but also promotes inter-molecular base-pairing interactions with target transcripts. Hfq plays a pivotal role for growth and survival, controlling central metabolism and cell wall synthesis in the oligotroph Caulobacter crescentus. However, direct evidence for Hfq-dependent post-transcriptional regulation and potential oligotrophy in C. crescentus has been lacking. Here, we identified sRNAs and mRNAs associated with Hfq in vivo, and demonstrated the requirement of Hfq for sRNA-mediated regulation, particularly of outer membrane transporters in C. crescentus.


Subject(s)
Caulobacter crescentus , RNA, Small Untranslated , Caulobacter crescentus/genetics , Caulobacter crescentus/metabolism , RNA, Small Untranslated/metabolism , RNA, Bacterial/metabolism , RNA, Messenger/genetics , Membrane Transport Proteins/metabolism , Host Factor 1 Protein/genetics , Host Factor 1 Protein/metabolism , Gene Expression Regulation, Bacterial
3.
mSystems ; 9(2): e0132623, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38270456

ABSTRACT

Tuberculosis remains the most pervasive infectious disease and the recent emergence of drug-resistant strains emphasizes the need for more efficient drug treatments. A key feature of pathogenesis, conserved between the human pathogen Mycobacterium tuberculosis and the model pathogen Mycobacterium marinum, is the metabolic switch to lipid catabolism and altered expression of virulence genes at different stages of infection. This study aims to identify genes involved in sustaining viable intracellular infection. We applied transposon sequencing (Tn-Seq) to M. marinum, an unbiased genome-wide strategy combining saturation insertional mutagenesis and high-throughput sequencing. This approach allowed us to identify the localization and relative abundance of insertions in pools of transposon mutants. Gene essentiality and fitness cost of mutations were quantitatively compared between in vitro growth and different stages of infection in two evolutionary distinct phagocytes, the amoeba Dictyostelium discoideum and the murine BV2 microglial cells. In the M. marinum genome, 57% of TA sites were disrupted and 568 genes (10.2%) were essential, which is comparable to previous Tn-Seq studies on M. tuberculosis and M. bovis. Major pathways involved in the survival of M. marinum during infection of D. discoideum are related to DNA damage repair, lipid and vitamin metabolism, the type VII secretion system (T7SS) ESX-1, and the Mce1 lipid transport system. These pathways, except Mce1 and some glycolytic enzymes, were similarly affected in BV2 cells. These differences suggest subtly distinct nutrient availability or requirement in different host cells despite the known predominant use of lipids in both amoeba and microglial cells.IMPORTANCEThe emergence of biochemically and genetically tractable host model organisms for infection studies holds the promise to accelerate the pace of discoveries related to the evolution of innate immunity and the dissection of conserved mechanisms of cell-autonomous defenses. Here, we have used the genetically and biochemically tractable infection model system Dictyostelium discoideum/Mycobacterium marinum to apply a genome-wide transposon-sequencing experimental strategy to reveal comprehensively which mutations confer a fitness advantage or disadvantage during infection and compare these to a similar experiment performed using the murine microglial BV2 cells as host for M. marinum to identify conservation of virulence pathways between hosts.


Subject(s)
Amoeba , Dictyostelium , Mycobacterium marinum , Mycobacterium tuberculosis , Tuberculosis , Animals , Mice , Humans , Virulence/genetics , Microglia , Mycobacterium marinum/genetics , Dictyostelium/genetics , Lipids
4.
PLoS Biol ; 21(12): e3002040, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38051727

ABSTRACT

The acquisition of multidrug resistance (MDR) determinants jeopardizes treatment of bacterial infections with antibiotics. The tripartite efflux pump AcrAB-NodT confers adaptive MDR in the polarized α-proteobacterium Caulobacter crescentus via transcriptional induction by first-generation quinolone antibiotics. We discovered that overexpression of AcrAB-NodT by mutation or exogenous inducers confers resistance to cephalosporin and penicillin (ß-lactam) antibiotics. Combining 2-step mutagenesis-sequencing (Mut-Seq) and cephalosporin-resistant point mutants, we dissected how TipR uses a common operator of the divergent tipR and acrAB-nodT promoter for adaptive and/or potentiated AcrAB-NodT-directed efflux. Chemical screening identified diverse compounds that interfere with DNA binding by TipR or induce its dependent proteolytic turnover. We found that long-term induction of AcrAB-NodT deforms the envelope and that homeostatic control by TipR includes co-induction of the DnaJ-like co-chaperone DjlA, boosting pump assembly and/or capacity in anticipation of envelope stress. Thus, the adaptive MDR regulatory circuitry reconciles drug efflux with co-chaperone function for trans-envelope assemblies and maintenance.


Subject(s)
Bacterial Proteins , Escherichia coli Proteins , Bacterial Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Biological Transport , Cephalosporins , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , beta-Lactam Resistance , Escherichia coli Proteins/metabolism , Microbial Sensitivity Tests
5.
Mol Cell ; 81(19): 3992-4007.e10, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34562373

ABSTRACT

ParB-like CTPases mediate the segregation of bacterial chromosomes and low-copy number plasmids. They act as DNA-sliding clamps that are loaded at parS motifs in the centromere of target DNA molecules and spread laterally to form large nucleoprotein complexes serving as docking points for the DNA segregation machinery. Here, we solve crystal structures of ParB in the pre- and post-hydrolysis state and illuminate the catalytic mechanism of nucleotide hydrolysis. Moreover, we identify conformational changes that underlie the CTP- and parS-dependent closure of ParB clamps. The study of CTPase-deficient ParB variants reveals that CTP hydrolysis serves to limit the sliding time of ParB clamps and thus drives the establishment of a well-defined ParB diffusion gradient across the centromere whose dynamics are critical for DNA segregation. These findings clarify the role of the ParB CTPase cycle in partition complex assembly and function and thus advance our understanding of this prototypic CTP-dependent molecular switch.


Subject(s)
Bacterial Proteins/metabolism , Chromosome Segregation , Chromosomes, Bacterial , Cytidine Triphosphate/metabolism , DNA, Bacterial/metabolism , Myxococcus xanthus/enzymology , Bacterial Proteins/genetics , Binding Sites , Catalytic Domain , Crystallography, X-Ray , DNA, Bacterial/genetics , Gene Expression Regulation, Bacterial , Hydrolysis , Mutation , Myxococcus xanthus/genetics , Protein Conformation , Structure-Activity Relationship , Substrate Specificity , Time Factors
6.
Dev Cell ; 56(5): 657-670.e4, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33600766

ABSTRACT

In some free-living and pathogenic bacteria, problems in the synthesis and assembly of early flagellar components can cause cell-division defects. However, the mechanism that couples cell division with the flagellar biogenesis has remained elusive. Herein, we discover the regulator MadA that controls transcription of flagellar and cell-division genes in Caulobacter crescentus. We demonstrate that MadA, a small soluble protein, binds the type III export component FlhA to promote activation of FliX, which in turn is required to license the conserved σ54-dependent transcriptional activator FlbD. While in the absence of MadA, FliX and FlbD activation is crippled, bypass mutations in FlhA restore flagellar biogenesis and cell division. Furthermore, we demonstrate that MadA safeguards the divisome stoichiometry to license cell division. We propose that MadA has a sentinel-type function that senses an early flagellar biogenesis event and, through cell-division control, ensures that a flagellated offspring emerges.


Subject(s)
Bacterial Proteins/metabolism , Caulobacter crescentus/cytology , Cell Division , Cell Movement , Flagella/physiology , Organelles/physiology , Transcription, Genetic , Bacterial Proteins/genetics , Caulobacter crescentus/genetics , Caulobacter crescentus/metabolism , Mutation , Promoter Regions, Genetic
7.
Dev Cell ; 55(4): 500-513.e4, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33113346

ABSTRACT

How cellular checkpoints couple the orderly assembly of macromolecular machines with cell-cycle progression is poorly understood. The alpha-proteobacterium Caulobacter crescentus assembles a single polar flagellum during each cell cycle. We discovered that the expression of multiple flagellin transcripts is licensed by a translational checkpoint responsive to a dual input signal: a secretion-competent hook-basal-body (HBB) structure and a surge in the FlaF secretion chaperone during cytokinesis, instructed by the cell-cycle program. We find that the unorthodox FljJ flagellin, one of the six flagellin paralogs, acts as a checkpoint linchpin, binding both FlaF and the FlbT translational regulator. FljJ recruits FlbT to inhibit translation at the 5' untranslated region in other flagellin transcripts before HBB assembly. Once FlaF is synthesized and stabilized, it directs FljJ secretion through the HBB, thereby separating FlbT from its co-activator and relieving translational inhibition. The FlbT/FlaF pair is wide spread and its functional properties are conserved in alpha-proteobacteria, including pathogens.


Subject(s)
Co-Repressor Proteins/metabolism , Flagellin/metabolism , Protein Biosynthesis , 5' Untranslated Regions/genetics , Bacterial Proteins/metabolism , Base Sequence , Binding, Competitive , Caulobacter crescentus/genetics , Flagella/metabolism , Gene Expression Regulation, Bacterial , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
PLoS Genet ; 16(4): e1008724, 2020 04.
Article in English | MEDLINE | ID: mdl-32324740

ABSTRACT

The Alphaproteobacteria show a remarkable diversity of cell cycle-dependent developmental patterns, which are governed by the conserved CtrA pathway. Its central component CtrA is a DNA-binding response regulator that is controlled by a complex two-component signaling network, mediating distinct transcriptional programs in the two offspring. The CtrA pathway has been studied intensively and was shown to consist of an upstream part that reads out the developmental state of the cell and a downstream part that integrates the upstream signals and mediates CtrA phosphorylation. However, the role of this circuitry in bacterial diversification remains incompletely understood. We have therefore investigated CtrA regulation in the morphologically complex stalked budding alphaproteobacterium Hyphomonas neptunium. Compared to relatives dividing by binary fission, H. neptunium shows distinct changes in the role and regulation of various pathway components. Most notably, the response regulator DivK, which normally links the upstream and downstream parts of the CtrA pathway, is dispensable, while downstream components such as the pseudokinase DivL, the histidine kinase CckA, the phosphotransferase ChpT and CtrA are essential. Moreover, CckA is compartmentalized to the nascent bud without forming distinct polar complexes and CtrA is not regulated at the level of protein abundance. We show that the downstream pathway controls critical functions such as replication initiation, cell division and motility. Quantification of the signal flow through different nodes of the regulatory cascade revealed that the CtrA pathway is a leaky pipeline and must involve thus-far unidentified factors. Collectively, the quantitative system-level analysis of CtrA regulation in H. neptunium points to a considerable evolutionary plasticity of cell cycle regulation in alphaproteobacteria and leads to hypotheses that may also hold in well-established model organisms such as Caulobacter crescentus.


Subject(s)
Alphaproteobacteria/genetics , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Gene Regulatory Networks , Transcription Factors/genetics , Alphaproteobacteria/metabolism , Bacterial Proteins/metabolism , Cell Division , Cell Movement , DNA Replication , Evolution, Molecular , Transcription Factors/metabolism
9.
Nucleic Acids Res ; 48(9): 4769-4779, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32232335

ABSTRACT

The spatiotemporal regulation of chromosome segregation and cell division in Caulobacter crescentus is mediated by two different P-loop ATPases, ParA and MipZ. Both of these proteins form dynamic concentration gradients that control the positioning of regulatory targets within the cell. Their proper localization depends on their nucleotide-dependent cycling between a monomeric and a dimeric state and on the ability of the dimeric species to associate with the nucleoid. In this study, we use a combination of genetic screening, biochemical analysis and hydrogen/deuterium exchange mass spectrometry to comprehensively map the residues mediating the interactions of MipZ and ParA with DNA. We show that MipZ has non-specific DNA-binding activity that relies on an array of positively charged and hydrophobic residues lining both sides of the dimer interface. Extending our analysis to ParA, we find that the MipZ and ParA DNA-binding sites differ markedly in composition, although their relative positions on the dimer surface and their mode of DNA binding are conserved. In line with previous experimental work, bioinformatic analysis suggests that the same principles may apply to other members of the P-loop ATPase family. P-loop ATPases thus share common mechanistic features, although their functions have diverged considerably during the course of evolution.


Subject(s)
Adenosine Triphosphatases/chemistry , Bacterial Proteins/chemistry , Caulobacter crescentus/enzymology , DNA-Binding Proteins/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diffusion , Hydrogen Deuterium Exchange-Mass Spectrometry , Mutation , Protein Binding
10.
PLoS Genet ; 16(1): e1008591, 2020 01.
Article in English | MEDLINE | ID: mdl-31961855

ABSTRACT

Bacterial growth and division require regulated synthesis of the macromolecules used to expand and replicate components of the cell. Transcription of housekeeping genes required for metabolic homeostasis and cell proliferation is guided by the sigma factor σ70. The conserved CarD-like transcriptional regulator, CdnL, associates with promoter regions where σ70 localizes and stabilizes the open promoter complex. However, the contributions of CdnL to metabolic homeostasis and bacterial physiology are not well understood. Here, we show that Caulobacter crescentus cells lacking CdnL have severe morphological and growth defects. Specifically, ΔcdnL cells grow slowly in both rich and defined media, and are wider, more curved, and have shorter stalks than WT cells. These defects arise from transcriptional downregulation of most major classes of biosynthetic genes, leading to significant decreases in the levels of critical metabolites, including pyruvate, α-ketoglutarate, ATP, NAD+, UDP-N-acetyl-glucosamine, lipid II, and purine and pyrimidine precursors. Notably, we find that ΔcdnL cells are glutamate auxotrophs, and ΔcdnL is synthetic lethal with other genetic perturbations that limit glutamate synthesis and lipid II production. Our findings implicate CdnL as a direct and indirect regulator of genes required for metabolic homeostasis that impacts morphogenesis through availability of lipid II and other metabolites.


Subject(s)
Bacterial Proteins/metabolism , Caulobacter crescentus/genetics , Homeostasis , Transcription Factors/metabolism , Bacterial Proteins/genetics , Caulobacter crescentus/metabolism , Caulobacter crescentus/physiology , Cell Division , Conserved Sequence , Metabolome , Transcription Factors/genetics
11.
Nucleic Acids Res ; 47(20): 10628-10644, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31598724

ABSTRACT

Many bacteria acquire dissemination and virulence traits in G1-phase. CtrA, an essential and conserved cell cycle transcriptional regulator identified in the dimorphic alpha-proteobacterium Caulobacter crescentus, first activates promoters in late S-phase and then mysteriously switches to different target promoters in G1-phase. We uncovered a highly conserved determinant in the DNA-binding domain (DBD) of CtrA uncoupling this promoter switch. We also show that it reprograms CtrA occupancy in stationary cells inducing a (p)ppGpp alarmone signal perceived by the RNA polymerase beta subunit. A simple side chain modification in a critical residue within the core DBD imposes opposing developmental phenotypes and transcriptional activities of CtrA and a proximal residue can direct CtrA towards activation of the dispersal (G1-phase) program. Hence, we propose that this conserved determinant in the CtrA primary structure dictates promoter reprogramming during the growth transition in other alpha-proteobacteria that differentiate from replicative cells into dispersal cells.


Subject(s)
Bacterial Proteins/metabolism , Caulobacter crescentus/growth & development , Caulobacter crescentus/metabolism , Cell Cycle , Transcription Factors/metabolism , Amino Acid Sequence , Bacterial Capsules/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Caulobacter crescentus/cytology , DNA, Bacterial/metabolism , G1 Phase , Guanosine Tetraphosphate/metabolism , Movement , Mutation/genetics , Promoter Regions, Genetic , Protein Binding , S Phase , Suppression, Genetic , Transcription Factors/chemistry , Transcription Factors/genetics
12.
Curr Opin Microbiol ; 42: 79-86, 2018 04.
Article in English | MEDLINE | ID: mdl-29161615

ABSTRACT

The Type IV pilus (T4P) is a powerful and sophisticated bacterial nanomachine involved in numerous cellular processes, including adhesion, DNA uptake and motility. Aside from the well-described subtype T4aP of the Gram-negative genera, including Myxococcus, Pseudomonas and Neisseria, the Tad (tight adherence) pilus secretion system re-shuffles homologous parts from other secretion systems along with uncharacterized components into a new type of protein translocation apparatus. A representative of the Tad apparatus, the Caulobacter crescentus pilus assembly (Cpa) machine is built exclusively at the newborn cell pole once per cell cycle. Recent comprehensive genetic analyses unearthed a myriad of spatiotemporal determinants acting on the Tad/Cpa system, many of which are conserved in other α-proteobacteria, including obligate intracellular pathogens and symbionts.


Subject(s)
Caulobacter/genetics , Fimbriae, Bacterial/genetics , Bacterial Adhesion/genetics , Bacterial Adhesion/physiology , Bacterial Proteins/metabolism , Caulobacter/metabolism , Fimbriae, Bacterial/physiology , Gene Expression Regulation, Bacterial , Models, Molecular , Pseudomonas aeruginosa/genetics
13.
Nucleic Acids Res ; 45(15): 8916-8929, 2017 Sep 06.
Article in English | MEDLINE | ID: mdl-28911105

ABSTRACT

We searched for regulators of chromosome replication in the cell cycle model Caulobacter crescentus and found a novel DNA-binding protein (GapR) that selectively aids the initiation of chromosome replication and the initial steps of chromosome partitioning. The protein binds the chromosome origin of replication (Cori) and has higher-affinity binding to mutated Cori-DNA that increases Cori-plasmid replication in vivo. gapR gene expression is essential for normal rapid growth and sufficient GapR levels are required for the correct timing of chromosome replication. Whole genome ChIP-seq identified dynamic DNA-binding distributions for GapR, with the strongest associations at the partitioning (parABS) locus near Cori. Using molecular-genetic and fluorescence microscopy experiments, we showed that GapR also promotes the first steps of chromosome partitioning, the initial separation of the duplicated parS loci following replication from Cori. This separation occurs before the parABS-dependent partitioning phase. Therefore, this early separation, whose mechanisms is not known, coincides with the poorly defined mechanism(s) that establishes chromosome asymmetry: C. crescentus chromosomes are partitioned to distinct cell-poles which develop into replicating and non-replicating cell-types. We propose that GapR coordinates chromosome replication with asymmetry-establishing chromosome separation, noting that both roles are consistent with the phylogenetic restriction of GapR to asymmetrically dividing bacteria.


Subject(s)
Bacterial Proteins/genetics , Caulobacter crescentus/genetics , Chromosome Segregation , Chromosomes, Bacterial/metabolism , DNA Replication , DNA-Binding Proteins/genetics , Bacterial Proteins/metabolism , Caulobacter crescentus/drug effects , Caulobacter crescentus/metabolism , Cell Division/drug effects , Chromosomes, Bacterial/ultrastructure , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial , Mutation , Novobiocin/pharmacology , Plasmids/chemistry , Plasmids/metabolism , Replication Origin , Rifampin/pharmacology
14.
Elife ; 52016 12 23.
Article in English | MEDLINE | ID: mdl-28008851

ABSTRACT

Protein polarization underlies differentiation in metazoans and in bacteria. How symmetric polarization can instate functional asymmetry remains elusive. Here, we show by super-resolution photo-activated localization microscopy and edgetic mutations that the bitopic zinc-finger protein ZitP implements specialized developmental functions - pilus biogenesis and multifactorial swarming motility - while shaping distinct nanoscale (bi)polar architectures in the asymmetric model bacterium Caulobacter crescentus. Polar assemblage and accumulation of ZitP and its effector protein CpaM are orchestrated in time and space by conserved components of the cell cycle circuitry that coordinate polar morphogenesis with cell cycle progression, and also act on the master cell cycle regulator CtrA. Thus, this novel class of potentially widespread multifunctional polarity regulators is deeply embedded in the cell cycle circuitry.


Subject(s)
Bacterial Proteins/metabolism , Caulobacter crescentus/physiology , Cell Cycle , Gene Expression Regulation, Bacterial , Zinc Fingers , Caulobacter crescentus/genetics , Caulobacter crescentus/metabolism , Fimbriae, Bacterial/metabolism , Locomotion , Microscopy , Mutation , Organelle Biogenesis
15.
ISME J ; 10(9): 2129-44, 2016 09.
Article in English | MEDLINE | ID: mdl-26953603

ABSTRACT

Like other obligate intracellular bacteria, the Chlamydiae feature a compact regulatory genome that remains uncharted owing to poor genetic tractability. Exploiting the reduced number of transcription factors (TFs) encoded in the chlamydial (pan-)genome as a model for TF control supporting the intracellular lifestyle, we determined the conserved landscape of TF specificities by ChIP-Seq (chromatin immunoprecipitation-sequencing) in the chlamydial pathogen Waddlia chondrophila. Among 10 conserved TFs, Euo emerged as a master TF targeting >100 promoters through conserved residues in a DNA excisionase-like winged helix-turn-helix-like (wHTH) fold. Minimal target (Euo) boxes were found in conserved developmentally-regulated genes governing vertical genome transmission (cytokinesis and DNA replication) and genome plasticity (transposases). Our ChIP-Seq analysis with intracellular bacteria not only reveals that global TF regulation is maintained in the reduced regulatory genomes of Chlamydiae, but also predicts that master TFs interpret genomic information in the obligate intracellular α-proteobacteria, including the rickettsiae, from which modern day mitochondria evolved.


Subject(s)
Chlamydiales/genetics , Genome, Bacterial/genetics , Transcription Factors/genetics , Verrucomicrobia/genetics , Animals , Bacterial Proteins/genetics , Chlorocebus aethiops , Chromatin Immunoprecipitation , Genomics , Phylogeny , Reproducibility of Results , Vero Cells
16.
FEMS Microbiol Rev ; 39(1): 120-33, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25793963

ABSTRACT

Recent data indicate that cell cycle transcription in many alpha-Proteobacteria is executed by at least three conserved functional modules in which pairs of antagonistic regulators act jointly, rather than in isolation, to control transcription in S-, G2- or G1-phase. Inactivation of module components often results in pleiotropic defects, ranging from cell death and impaired cell division to fairly benign deficiencies in motility. Expression of module components can follow systemic (cell cycle) or external (nutritional/cell density) cues and may be implemented by auto-regulation, ancillary regulators or other (unknown) mechanisms. Here, we highlight the recent progress in understanding the molecular events and the genetic relationships of the module components in environmental, pathogenic and/or symbiotic alpha-proteobacterial genera. Additionally, we take advantage of the recent genome-wide transcriptional analyses performed in the model alpha-Proteobacterium Caulobacter crescentus to illustrate the complexity of the interactions of the global regulators at selected cell cycle-regulated promoters and we detail the consequences of (mis-)expression when the regulators are absent. This review thus provides the first detailed mechanistic framework for understanding orthologous operational principles acting on cell cycle-regulated promoters in other alpha-Proteobacteria.


Subject(s)
Alphaproteobacteria/genetics , Alphaproteobacteria/metabolism , Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Bacterial , Caulobacter crescentus/cytology , Caulobacter crescentus/genetics , Caulobacter crescentus/metabolism , Promoter Regions, Genetic/genetics
17.
PLoS Biol ; 11(12): e1001749, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24415923

ABSTRACT

What are the minimal requirements to sustain an asymmetric cell cycle? Here we use mathematical modelling and forward genetics to reduce an asymmetric cell cycle to its simplest, primordial components. In the Alphaproteobacterium Caulobacter crescentus, cell cycle progression is believed to be controlled by a cyclical genetic circuit comprising four essential master regulators. Unexpectedly, our in silico modelling predicted that one of these regulators, GcrA, is in fact dispensable. We confirmed this experimentally, finding that ΔgcrA cells are viable, but slow-growing and elongated, with the latter mostly due to an insufficiency of a key cell division protein. Furthermore, suppressor analysis showed that another cell cycle regulator, the methyltransferase CcrM, is similarly dispensable with simultaneous gcrA/ccrM disruption ameliorating the cytokinetic and growth defect of ΔgcrA cells. Within the Alphaproteobacteria, gcrA and ccrM are consistently present or absent together, rather than either gene being present alone, suggesting that gcrA/ccrM constitutes an independent, dispensable genetic module. Together our approaches unveil the essential elements of a primordial asymmetric cell cycle that should help illuminate more complex cell cycles.


Subject(s)
Caulobacter crescentus/genetics , Caulobacter crescentus/physiology , Cell Cycle/genetics , Cell Cycle/physiology , Cell Survival/genetics , Cell Survival/physiology , Computational Biology/methods , Computer Simulation , DNA Transposable Elements/genetics , DNA Transposable Elements/physiology , Gene Expression Regulation, Bacterial/genetics , Gene Expression Regulation, Bacterial/physiology , Methylation , Models, Biological
18.
Viruses ; 4(11): 2417-31, 2012 Oct 24.
Article in English | MEDLINE | ID: mdl-23202488

ABSTRACT

Recombination directionality factors (RDFs), or excisionases, are essential players of prophage excisive recombination. Despite the essentially catalytic role of the integrase in both integrative and excisive recombination, RDFs are required to direct the reaction towards excision and to prevent re-integration of the prophage genome when entering a lytic cycle. KplE1, HK620 and numerous (pro)phages that integrate at the same site in enterobacteria genomes (such as the argW tRNA gene) all share a highly conserved recombination module. This module comprises the attL and attR recombination sites and the RDF and integrase genes. The KplE1 RDF was named TorI after its initial identification as a negative regulator of the tor operon. However, it was characterized as an essential factor of excisive recombination. In this study, we designed an extensive random mutagenesis protocol of the torI gene and identified key residues involved in both functions of the TorI protein. We show that, in addition to TorI-TorR protein-protein interaction, TorI interacts in solution with the IntS integrase. Moreover, in vitro, TorR and IntS appear to compete for TorI binding. Finally, our mutagenesis results suggest that the C-terminal part of the TorI protein is dedicated to protein-protein interactions with both proteins TorR and IntS.


Subject(s)
DNA Nucleotidyltransferases/metabolism , Prophages/genetics , Prophages/metabolism , Recombination, Genetic , Viral Proteins/metabolism , Amino Acid Sequence , DNA Nucleotidyltransferases/chemistry , DNA Nucleotidyltransferases/genetics , Enzyme Activation , Models, Molecular , Molecular Sequence Data , Mutagenesis , Protein Binding , Protein Conformation , Viral Proteins/chemistry , Viral Proteins/genetics
19.
J Virol ; 86(18): 10234-5, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22923796

ABSTRACT

φCbK is a B3 morphotype bacteriophage of the Siphoviridae family that infects Caulobacter crescentus, the preeminent model system for bacterial cell cycle studies. The last 4 decades of research with φCbK as a genetic and cytological tool to study the biology of the host warrant an investigation of the phage genome composition. Herein, we report the complete genome sequence of φCbK and highlight unusual features that emerged from its annotation. The complete genome analysis of the φCbK phage provides new insight into its characteristics and potential interactions with its Caulobacter crescentus host, setting the stage for future functional studies with φCbK.


Subject(s)
Caulobacter crescentus/virology , Siphoviridae/genetics , DNA, Viral/genetics , Genome, Viral , Host-Pathogen Interactions , Molecular Sequence Data , Siphoviridae/pathogenicity
20.
PLoS Genet ; 6(10)2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20949106

ABSTRACT

Temperate phages have the ability to maintain their genome in their host, a process called lysogeny. For most, passive replication of the phage genome relies on integration into the host's chromosome and becoming a prophage. Prophages remain silent in the absence of stress and replicate passively within their host genome. However, when stressful conditions occur, a prophage excises itself and resumes the viral cycle. Integration and excision of phage genomes are mediated by regulated site-specific recombination catalyzed by tyrosine and serine recombinases. In the KplE1 prophage, site-specific recombination is mediated by the IntS integrase and the TorI recombination directionality factor (RDF). We previously described a sub-family of temperate phages that is characterized by an unusual organization of the recombination module. Consequently, the attL recombination region overlaps with the integrase promoter, and the integrase and RDF genes do not share a common activated promoter upon lytic induction as in the lambda prophage. In this study, we show that the intS gene is tightly regulated by its own product as well as by the TorI RDF protein. In silico analysis revealed that overlap of the attL region with the integrase promoter is widely encountered in prophages present in prokaryotic genomes, suggesting a general occurrence of negatively autoregulated integrase genes. The prediction that these integrase genes are negatively autoregulated was biologically assessed by studying the regulation of several integrase genes from two different Escherichia coli strains. Our results suggest that the majority of tRNA-associated integrase genes in prokaryotic genomes could be autoregulated and that this might be correlated with the recombination efficiency as in KplE1. The consequences of this unprecedented regulation for excessive recombination are discussed.


Subject(s)
Gene Expression Regulation, Viral , Integrases/genetics , Prophages/genetics , Viral Proteins/genetics , Attachment Sites, Microbiological/genetics , Base Sequence , Binding Sites/genetics , Escherichia coli/genetics , Escherichia coli/virology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Integrases/metabolism , Microscopy, Fluorescence , Models, Genetic , Molecular Sequence Data , Mutation , Promoter Regions, Genetic/genetics , Prophages/enzymology , RNA, Transfer/genetics , Recombination, Genetic , Viral Proteins/metabolism , Virus Integration
SELECTION OF CITATIONS
SEARCH DETAIL
...