Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Cell ; 56(5): 657-670.e4, 2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33600766

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Caulobacter crescentus/citologia , Divisão Celular , Movimento Celular , Flagelos/fisiologia , Organelas/fisiologia , Transcrição Gênica , Proteínas de Bactérias/genética , Caulobacter crescentus/genética , Caulobacter crescentus/metabolismo , Mutação , Regiões Promotoras Genéticas
2.
PLoS Genet ; 15(8): e1008284, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31437147

RESUMO

Several important human pathogens are represented in the Corynebacterineae suborder, including Mycobacterium tuberculosis and Corynebacterium diphtheriae. These bacteria are surrounded by a multilayered cell envelope composed of a cytoplasmic membrane, a peptidoglycan (PG) cell wall, a second polysaccharide layer called the arabinogalactan (AG), and finally an outer membrane-like layer made of mycolic acids. Several anti-tuberculosis drugs target the biogenesis of this complex envelope, but their efficacy is declining due to resistance. New therapies are therefore needed to treat diseases caused by these organisms, and a better understanding of the mechanisms of envelope assembly should aid in their discovery. To this end, we generated the first high-density library of transposon insertion mutants in the model organism C. glutamicum. Transposon-sequencing was then used to define its essential gene set and identify loci that, when inactivated, confer hypersensitivity to ethambutol (EMB), a drug that targets AG biogenesis. Among the EMBs loci were genes encoding RipC and the FtsEX complex, a PG cleaving enzyme required for proper cell division and its predicted regulator, respectively. Inactivation of the conserved steAB genes (cgp_1603-1604) was also found to confer EMB hypersensitivity and cell division defects. A combination of quantitative microscopy, mutational analysis, and interaction studies indicate that SteA and SteB form a complex that localizes to the cytokinetic ring to promote cell separation by RipC-FtsEX and may coordinate its PG remodeling activity with the biogenesis of other envelope layers during cell division.


Assuntos
Antituberculosos/farmacologia , Membrana Externa Bacteriana/metabolismo , Divisão Celular/genética , Corynebacterium glutamicum/fisiologia , Farmacorresistência Bacteriana/genética , Membrana Externa Bacteriana/efeitos dos fármacos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Vias Biossintéticas/efeitos dos fármacos , Corynebacterium glutamicum/efeitos dos fármacos , Elementos de DNA Transponíveis/genética , Etambutol/farmacologia , Galactanos/biossíntese , Loci Gênicos , Mutação , Ácidos Micólicos/metabolismo , Peptidoglicano/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(12): 3150-3155, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29507210

RESUMO

Penicillin-binding proteins (PBPs) are synthases required to build the essential peptidoglycan (PG) cell wall surrounding most bacterial cells. The mechanisms regulating the activity of these enzymes to control PG synthesis remain surprisingly poorly defined given their status as key antibiotic targets. Several years ago, the outer-membrane lipoprotein EcLpoB was identified as a critical activator of Escherichia coli PBP1b (EcPBP1b), one of the major PG synthases of this organism. Activation of EcPBP1b is mediated through the association of EcLpoB with a regulatory domain on EcPBP1b called UB2H. Notably, Pseudomonas aeruginosa also encodes PBP1b (PaPBP1b), which possesses a UB2H domain, but this bacterium lacks an identifiable LpoB homolog. We therefore searched for potential PaPBP1b activators and identified a lipoprotein unrelated to LpoB that is required for the in vivo activity of PaPBP1b. We named this protein LpoP and found that it interacts directly with PaPBP1b in vitro and is conserved in many Gram-negative species. Importantly, we also demonstrated that PaLpoP-PaPBP1b as well as an equivalent protein pair from Acinetobacter baylyi can fully substitute for EcLpoB-EcPBP1b in E. coli for PG synthesis. Furthermore, we show that amino acid changes in PaPBP1b that bypass the PaLpoP requirement map to similar locations in the protein as changes promoting EcLpoB bypass in EcPBP1b. Overall, our results indicate that, although different Gram-negative bacteria activate their PBP1b synthases with distinct lipoproteins, they stimulate the activity of these important drug targets using a conserved mechanism.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Ligação às Penicilinas/metabolismo , Pseudomonas aeruginosa/metabolismo , Acinetobacter/química , Proteínas de Bactérias/genética , Parede Celular/metabolismo , Elementos de DNA Transponíveis , Escherichia coli/genética , Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Lipoproteínas/química , Lipoproteínas/genética , Lipoproteínas/metabolismo , Mutação , Proteínas de Ligação às Penicilinas/química , Proteínas de Ligação às Penicilinas/genética , Filogenia , Pseudomonas aeruginosa/citologia , Pseudomonas aeruginosa/genética
4.
mBio ; 8(2)2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28351916

RESUMO

Peptidoglycan (PG) is an essential cross-linked polymer that surrounds most bacterial cells to prevent osmotic rupture of the cytoplasmic membrane. Its synthesis relies on penicillin-binding proteins, the targets of beta-lactam antibiotics. Many Gram-negative bacteria, including the opportunistic pathogen Pseudomonas aeruginosa, are resistant to beta-lactams because of a chromosomally encoded beta-lactamase called AmpC. In P. aeruginosa, expression of the ampC gene is tightly regulated and its induction is linked to cell wall stress. We reasoned that a reporter gene fusion to the ampC promoter would allow us to identify mutants defective in maintaining cell wall homeostasis and thereby uncover new factors involved in the process. A library of transposon-mutagenized P. aeruginosa was therefore screened for mutants with elevated ampC promoter activity. As an indication that the screen was working as expected, mutants with transposons disrupting the dacB gene were isolated. Defects in DacB have previously been implicated in ampC induction and clinical resistance to beta-lactam antibiotics. The screen also uncovered murU and PA3172 mutants that, upon further characterization, displayed nearly identical drug resistance and sensitivity profiles. We present genetic evidence that PA3172, renamed mupP, encodes the missing phosphatase predicted to function in the MurU PG recycling pathway that is widely distributed among Gram-negative bacteria.IMPORTANCE The cell wall biogenesis pathway is the target of many of our best antibiotics, including penicillin and related beta-lactam drugs. Resistance to these therapies is on the rise, particularly among Gram-negative species like Pseudomonas aeruginosa, a problematic opportunistic pathogen. To better understand how these organisms resist cell wall-targeting antibiotics, we screened for P. aeruginosa mutants defective in maintaining cell wall homeostasis. The screen identified a new factor, called MupP, involved in the recycling of cell wall turnover products. Characterization of MupP and other components of the pathway revealed that cell wall recycling plays important roles in both the resistance and the sensitivity of P. aeruginosa to cell wall-targeting antibiotics.


Assuntos
Parede Celular/metabolismo , Farmacorresistência Bacteriana , Peptidoglicano/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/metabolismo , Elementos de DNA Transponíveis , Mutagênese Insercional , Monoéster Fosfórico Hidrolases/genética , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética
5.
PLoS Genet ; 12(12): e1006499, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27997543

RESUMO

Heritable DNA methylation imprints are ubiquitous and underlie genetic variability from bacteria to humans. In microbial genomes, DNA methylation has been implicated in gene transcription, DNA replication and repair, nucleoid segregation, transposition and virulence of pathogenic strains. Despite the importance of local (hypo)methylation at specific loci, how and when these patterns are established during the cell cycle remains poorly characterized. Taking advantage of the small genomes and the synchronizability of α-proteobacteria, we discovered that conserved determinants of the cell cycle transcriptional circuitry establish specific hypomethylation patterns in the cell cycle model system Caulobacter crescentus. We used genome-wide methyl-N6-adenine (m6A-) analyses by restriction-enzyme-cleavage sequencing (REC-Seq) and single-molecule real-time (SMRT) sequencing to show that MucR, a transcriptional regulator that represses virulence and cell cycle genes in S-phase but no longer in G1-phase, occludes 5'-GANTC-3' sequence motifs that are methylated by the DNA adenine methyltransferase CcrM. Constitutive expression of CcrM or heterologous methylases in at least two different α-proteobacteria homogenizes m6A patterns even when MucR is present and affects promoter activity. Environmental stress (phosphate limitation) can override and reconfigure local hypomethylation patterns imposed by the cell cycle circuitry that dictate when and where local hypomethylation is instated.


Assuntos
Caulobacter crescentus/genética , Ciclo Celular/genética , Metilação de DNA/genética , Transcrição Gênica , Divisão Celular/genética , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/genética , Regulação Bacteriana da Expressão Gênica , Genoma Microbiano , Metiltransferases/genética , Fosfatos/metabolismo , Regiões Promotoras Genéticas , DNA Metiltransferases Sítio Específica (Adenina-Específica)/genética , Inanição/genética , Inanição/metabolismo
6.
Elife ; 32014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25421297

RESUMO

Despite the crucial role of bacterial capsules in pathogenesis, it is still unknown if systemic cues such as the cell cycle can control capsule biogenesis. In this study, we show that the capsule of the synchronizable model bacterium Caulobacter crescentus is cell cycle regulated and we unearth a bacterial transglutaminase homolog, HvyA, as restriction factor that prevents capsulation in G1-phase cells. This capsule protects cells from infection by a generalized transducing Caulobacter phage (φCr30), and the loss of HvyA confers insensitivity towards φCr30. Control of capsulation during the cell cycle could serve as a simple means to prevent steric hindrance of flagellar motility or to ensure that phage-mediated genetic exchange happens before the onset of DNA replication. Moreover, the multi-layered regulatory circuitry directing HvyA expression to G1-phase is conserved during evolution, and HvyA orthologues from related Sinorhizobia can prevent capsulation in Caulobacter, indicating that alpha-proteobacteria have retained HvyA activity.


Assuntos
Bacteriófagos/fisiologia , Caulobacter/citologia , Caulobacter/virologia , Ciclo Celular , Alphaproteobacteria , Cápsulas Bacterianas/metabolismo , Proteínas de Bactérias/metabolismo , Caulobacter/enzimologia , Caulobacter/ultraestrutura , Fluorescência , Fase G1 , Regulação Bacteriana da Expressão Gênica , Microscopia de Força Atômica , Modelos Biológicos , Estabilidade Proteica , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Transglutaminases/metabolismo , Compostos de Trimetilsilil/metabolismo
7.
Nat Commun ; 5: 4081, 2014 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-24939058

RESUMO

Zinc-finger domain transcriptional regulators regulate a myriad of functions in eukaryotes. Interestingly, ancestral versions (MucR) from Alpha-proteobacteria control bacterial virulence/symbiosis. Whether virulence regulators can also control cell cycle transcription is unknown. Here we report that MucR proteins implement a hitherto elusive primordial S→G1 transcriptional switch. After charting G1-specific promoters in the cell cycle model Caulobacter crescentus by comparative ChIP-seq, we use one such promoter as genetic proxy to unearth two MucR paralogs, MucR1/2, as constituents of a quadripartite and homeostatic regulatory module directing the S→G1 transcriptional switch. Surprisingly, MucR orthologues that regulate virulence and symbiosis gene transcription in Brucella, Agrobacterium or Sinorhizobium support this S→G1 switch in Caulobacter. Pan-genomic ChIP-seq analyses in Sinorhizobium and Caulobacter show that this module indeed targets orthologous genes. We propose that MucR proteins and possibly other virulence regulators primarily control bacterial cell cycle (G1-phase) transcription, rendering expression of target (virulence) genes periodic and in tune with the cell cycle.


Assuntos
Proteínas de Bactérias/metabolismo , Caulobacter/fisiologia , Fase G1/fisiologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Modelos Biológicos , Proteínas Repressoras/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular/fisiologia , Proteínas de Bactérias/genética , Sequência de Bases , Imunoprecipitação da Cromatina , Dimerização , Ensaio de Desvio de Mobilidade Eletroforética , Fase G1/genética , Regulação Bacteriana da Expressão Gênica/genética , Immunoblotting , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Repressoras/genética , Pontos de Checagem da Fase S do Ciclo Celular/genética , Análise de Sequência de DNA , Especificidade da Espécie , beta-Galactosidase
8.
PLoS Genet ; 10(1): e1004101, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24465221

RESUMO

In natural environments, bacteria often adhere to surfaces where they form complex multicellular communities. Surface adherence is determined by the biochemical composition of the cell envelope. We describe a novel regulatory mechanism by which the bacterium, Caulobacter crescentus, integrates cell cycle and nutritional signals to control development of an adhesive envelope structure known as the holdfast. Specifically, we have discovered a 68-residue protein inhibitor of holdfast development (HfiA) that directly targets a conserved glycolipid glycosyltransferase required for holdfast production (HfsJ). Multiple cell cycle regulators associate with the hfiA and hfsJ promoters and control their expression, temporally constraining holdfast development to the late stages of G1. HfiA further functions as part of a 'nutritional override' system that decouples holdfast development from the cell cycle in response to nutritional cues. This control mechanism can limit surface adhesion in nutritionally sub-optimal environments without affecting cell cycle progression. We conclude that post-translational regulation of cell envelope enzymes by small proteins like HfiA may provide a general means to modulate the surface properties of bacterial cells.


Assuntos
Aderência Bacteriana/genética , Proteínas de Bactérias/genética , Caulobacter crescentus/crescimento & desenvolvimento , Pontos de Checagem do Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Bactérias/metabolismo , Caulobacter crescentus/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Regulação Bacteriana da Expressão Gênica , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Nutrigenômica/métodos , Regiões Promotoras Genéticas , Transdução de Sinais/genética
9.
Genes Dev ; 27(18): 2049-62, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24065770

RESUMO

Eukaryotic morphogenesis is seeded with the establishment and subsequent amplification of polarity cues at key times during the cell cycle, often using (cyclic) nucleotide signals. We discovered that flagellum de- and repolarization in the model prokaryote Caulobacter crescentus is precisely orchestrated through at least three spatiotemporal mechanisms integrated at TipF. We show that TipF is a cell cycle-regulated receptor for the second messenger--bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP)--that perceives and transduces this signal through the degenerate c-di-GMP phosphodiesterase (EAL) domain to nucleate polar flagellum biogenesis. Once c-di-GMP levels rise at the G1 → S transition, TipF is activated, stabilized, and polarized, enabling the recruitment of downstream effectors, including flagellar switch proteins and the PflI positioning factor, at a preselected pole harboring the TipN landmark. These c-di-GMP-dependent events are coordinated with the onset of tipF transcription in early S phase and together enable the correct establishment and robust amplification of TipF-dependent polarization early in the cell cycle. Importantly, these mechanisms also govern the timely removal of TipF at cell division coincident with the drop in c-di-GMP levels, thereby resetting the flagellar polarization state in the next cell cycle after a preprogrammed period during which motility must be suspended.


Assuntos
Proteínas de Bactérias/metabolismo , Caulobacter crescentus/citologia , Caulobacter crescentus/metabolismo , Ciclo Celular/fisiologia , Flagelos/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Caulobacter crescentus/genética , Polaridade Celular , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Ativação Enzimática , Regulação Bacteriana da Expressão Gênica , Dados de Sequência Molecular , Ligação Proteica , Transporte Proteico , Alinhamento de Sequência , Transdução de Sinais
10.
PLoS Genet ; 9(5): e1003541, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23737758

RESUMO

Several regulators are involved in the control of cell cycle progression in the bacterial model system Caulobacter crescentus, which divides asymmetrically into a vegetative G1-phase (swarmer) cell and a replicative S-phase (stalked) cell. Here we report a novel functional interaction between the enigmatic cell cycle regulator GcrA and the N6-adenosine methyltransferase CcrM, both highly conserved proteins among Alphaproteobacteria, that are activated early and at the end of S-phase, respectively. As no direct biochemical and regulatory relationship between GcrA and CcrM were known, we used a combination of ChIP (chromatin-immunoprecipitation), biochemical and biophysical experimentation, and genetics to show that GcrA is a dimeric DNA-binding protein that preferentially targets promoters harbouring CcrM methylation sites. After tracing CcrM-dependent N6-methyl-adenosine promoter marks at a genome-wide scale, we show that these marks recruit GcrA in vitro and in vivo. Moreover, we found that, in the presence of a methylated target, GcrA recruits the RNA polymerase to the promoter, consistent with its role in transcriptional activation. Since methylation-dependent DNA binding is also observed with GcrA orthologs from other Alphaproteobacteria, we conclude that GcrA is the founding member of a new and conserved class of transcriptional regulators that function as molecular effectors of a methylation-dependent (non-heritable) epigenetic switch that regulates gene expression during the cell cycle.


Assuntos
Caulobacter crescentus/genética , Metilação de DNA/genética , Metiltransferases/genética , Transcrição Gênica , Adenosina/genética , Alphaproteobacteria/crescimento & desenvolvimento , Sequência de Aminoácidos , Caulobacter crescentus/crescimento & desenvolvimento , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Epigênese Genética , Regulação Bacteriana da Expressão Gênica , Metiltransferases/metabolismo , Regiões Promotoras Genéticas
11.
PLoS Biol ; 11(12): e1001749, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24415923

RESUMO

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.


Assuntos
Caulobacter crescentus/genética , Caulobacter crescentus/fisiologia , Ciclo Celular/genética , Ciclo Celular/fisiologia , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Biologia Computacional/métodos , Simulação por Computador , Elementos de DNA Transponíveis/genética , Elementos de DNA Transponíveis/fisiologia , Regulação Bacteriana da Expressão Gênica/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Metilação , Modelos Biológicos
12.
Microbiology (Reading) ; 157(Pt 10): 2745-2758, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21719545

RESUMO

Rhizobia are a disparate collection of soil bacteria capable of reducing atmospheric nitrogen in symbiosis with legumes (Fix phenotype). Synthesis of the nitrogenase and its accessory components is under the transcriptional control of the key regulator NifA and is generally restricted to the endosymbiotic forms of rhizobia known as bacteroids. Amongst studied rhizobia, Sinorhizobium fredii strain NGR234 has the remarkable ability to fix nitrogen in association with more than 130 species in 73 legume genera that form either determinate, indeterminate or aeschynomenoid nodules. Hence, NGR234 is a model organism to study nitrogen fixation in association with a variety of legumes. The symbiotic plasmid pSfrNGR234a carries more than 50 genes that are under the transcriptional control of NifA. To facilitate the functional analysis of NifA-regulated genes a new transposable element, TnEKm-PwA, was constructed. This transposon combines the advantages of in vitro mutagenesis of cloned DNA fragments with a conditional read-out promoter from NGR234 (PwA) that reinitiates NifA-dependent transcription downstream of transposition sites. To test the characteristics of the new transposon, the nifQdctA1y4vGHIJ operon was mutated using either the Omega interposon or TnEKm-PwA. The symbiotic phenotypes on various hosts as well as the transcriptional characteristics of these mutants were analysed in detail and compared with the ineffective (Fix(-)) phenotype of strain NGRΔnifA, which lacks a functional copy of nifA. De novo transcription from inserted copies of TnEKm-PwA inside bacteroids was confirmed by qRT-PCR. Unexpectedly, polar mutants in dctA1 and nifQ were Fix(+) on all of the hosts tested, indicating that none of the six genes of the nifQ operon of NGR234 is essential for symbiotic nitrogen fixation on plants that form nodules of either determinate or indeterminate types.


Assuntos
Proteínas de Bactérias/genética , Transportadores de Ácidos Dicarboxílicos/metabolismo , Regulação Bacteriana da Expressão Gênica , Óperon , Regiões Promotoras Genéticas , Sinorhizobium fredii/genética , Fatores de Transcrição/metabolismo , Proteínas de Bactérias/metabolismo , Sequência de Bases , Elementos de DNA Transponíveis , Transportadores de Ácidos Dicarboxílicos/genética , Fabaceae/microbiologia , Fabaceae/fisiologia , Dados de Sequência Molecular , Mutagênese Insercional , Sinorhizobium fredii/fisiologia , Simbiose , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...