Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Biol ; 20(1): e3001523, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35061668

RESUMO

Bacteria have membrane-spanning efflux pumps to secrete toxic compounds ranging from heavy metal ions to organic chemicals, including antibiotic drugs. The overall architecture of these efflux pumps is highly conserved: with an inner membrane energy-transducing subunit coupled via an adaptor protein to an outer membrane conduit subunit that enables toxic compounds to be expelled into the environment. Here, we map the distribution of efflux pumps across bacterial lineages to show these proteins are more widespread than previously recognised. Complex phylogenetics support the concept that gene cassettes encoding the subunits for these pumps are commonly acquired by horizontal gene transfer. Using TolC as a model protein, we demonstrate that assembly of conduit subunits into the outer membrane uses the chaperone TAM to physically organise the membrane-embedded staves of the conduit subunit of the efflux pump. The characteristics of this assembly pathway have impact for the acquisition of efflux pumps across bacterial species and for the development of new antimicrobial compounds that inhibit efflux pump function.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Escherichia coli/fisiologia , Chaperonas Moleculares , Membrana Externa Bacteriana/fisiologia , Transporte Biológico , Farmacorresistência Bacteriana/fisiologia , Proteínas de Escherichia coli , Proteínas de Membrana Transportadoras , Filogenia
2.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34873038

RESUMO

The hallmark of the gram-negative bacterial envelope is the presence of the outer membrane (OM). The OM is asymmetric, comprising lipopolysaccharides (LPS) in the outer leaflet and phospholipids (PLs) in the inner leaflet; this critical feature confers permeability barrier function against external insults, including antibiotics. To maintain OM lipid asymmetry, the OmpC-Mla system is believed to remove aberrantly localized PLs from the OM and transport them to the inner membrane (IM). Key to the system in driving lipid trafficking is the MlaFEDB ATP-binding cassette transporter complex in the IM, but mechanistic details, including transport directionality, remain enigmatic. Here, we develop a sensitive point-to-point in vitro lipid transfer assay that allows direct tracking of [14C]-labeled PLs between the periplasmic chaperone MlaC and MlaFEDB reconstituted into nanodiscs. We reveal that MlaC spontaneously transfers PLs to the IM transporter in an MlaD-dependent manner that can be further enhanced by coupled ATP hydrolysis. In addition, we show that MlaD is important for modulating productive coupling between ATP hydrolysis and such retrograde PL transfer. We further demonstrate that spontaneous PL transfer also occurs from MlaFEDB to MlaC, but such anterograde movement is instead abolished by ATP hydrolysis. Our work uncovers a model where PLs reversibly partition between two lipid-binding sites in MlaC and MlaFEDB, and ATP binding and/or hydrolysis shift this equilibrium to ultimately drive retrograde PL transport by the OmpC-Mla system. These mechanistic insights will inform future efforts toward discovering new antibiotics against gram-negative pathogens.


Assuntos
Trifosfato de Adenosina/metabolismo , Membrana Externa Bacteriana/fisiologia , Proteínas de Escherichia coli/metabolismo , Lipídeos/química , Transporte Biológico/fisiologia , Escherichia coli , Proteínas de Escherichia coli/genética , Imidazóis
3.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34716276

RESUMO

Gram-negative bacteria are surrounded by a protective outer membrane (OM) with phospholipids in its inner leaflet and lipopolysaccharides (LPS) in its outer leaflet. The OM is also populated with many ß-barrel outer-membrane proteins (OMPs), some of which have been shown to cluster into supramolecular assemblies. However, it remains unknown how abundant OMPs are organized across the entire bacterial surface and how this relates to the lipids in the membrane. Here, we reveal how the OM is organized from molecular to cellular length scales, using atomic force microscopy to visualize the OM of live bacteria, including engineered Escherichia coli strains and complemented by specific labeling of abundant OMPs. We find that a predominant OMP in the E. coli OM, the porin OmpF, forms a near-static network across the surface, which is interspersed with barren patches of LPS that grow and merge with other patches during cell elongation. Embedded within the porin network is OmpA, which forms noncovalent interactions to the underlying cell wall. When the OM is destabilized by mislocalization of phospholipids to the outer leaflet, a new phase appears, correlating with bacterial sensitivity to harsh environments. We conclude that the OM is a mosaic of phase-separated LPS-rich and OMP-rich regions, the maintenance of which is essential to the integrity of the membrane and hence to the lifestyle of a gram-negative bacterium.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Externa Bacteriana/metabolismo , Condensados Biomoleculares/fisiologia , Membrana Externa Bacteriana/fisiologia , Membrana Celular/metabolismo , Parede Celular/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Bicamadas Lipídicas/metabolismo , Lipopolissacarídeos/metabolismo , Simulação de Dinâmica Molecular , Fosfolipídeos/metabolismo , Porinas/metabolismo
4.
Methods Mol Biol ; 2341: 95-101, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34264465

RESUMO

Quantifying fluorescent markers in cell populations using flow cytometry has been a powerful technological advance. Fluorescent properties of cyanine dyes coupled with flow cytometry allow investigators to monitor the membrane potential (MP), an important component of the proton motive force (PMF). MP (or ΔΨ) is the electrical potential across the cell membrane. The other component of the PMF is ΔpH, or the difference in interior and exterior proton concentrations. MP plays a critical role in bacterial physiology. In Staphylococcus aureus, MP is required for generation of ATP, regulating autolytic activity, maintaining ion homeostasis, and resistance to some classes of antibiotics. This protocol exploits unique spectral and physical properties of the cyanine-based molecule diethyloxacarbocyanine iodide, or DiOC, and flow cytometry technology to quantify MP in S. aureus. This assay has been used by researchers to define the electron transport chain of S. aureus as well as determine how intrinsic and extrinsic factors affect MP.


Assuntos
Membrana Externa Bacteriana/fisiologia , Staphylococcus aureus/fisiologia , Autólise , Carbocianinas/química , Corantes/química , Citometria de Fluxo , Potenciais da Membrana , Força Próton-Motriz
5.
Res Microbiol ; 172(6): 103865, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34284091

RESUMO

An extensive morphological analysis of the Neisseria meningitidis cell envelope, including serogroup B capsule and outer membrane, based on atomic force microscopy (AFM) together with mechanical characterization by force spectroscopic measurements, has been carried out. Three meningococcal strains were used: the encapsulated serogroup B strain B1940, and the isogenic mutants B1940 siaD(+C) (lacking capsule), and B1940 cps (lacking both capsule and lipooligosaccharide outer core). AFM experiments with the encapsulated strain B1940 provided unprecedented images of the meningococcal capsule, which seems to be characterized by protrusions ("bumps") with the lateral dimensions of about 30 nm. Measurement of the Young's modulus provided quantitative assessment of the property of the capsule to confer resistance to mechanical stress. Moreover, Raman spectroscopy gave a fingerprint by which it was possible to identify the specific molecular species of the three strains analyzed, and to highlight major differences between them.


Assuntos
Cápsulas Bacterianas/ultraestrutura , Membrana Externa Bacteriana/ultraestrutura , Neisseria meningitidis Sorogrupo B/ultraestrutura , Cápsulas Bacterianas/química , Cápsulas Bacterianas/fisiologia , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/fisiologia , Módulo de Elasticidade , Microscopia de Força Atômica , Neisseria meningitidis Sorogrupo B/química , Neisseria meningitidis Sorogrupo B/genética , Polissacarídeos Bacterianos/química , Análise Espectral Raman , Estresse Mecânico , Propriedades de Superfície
6.
Elife ; 102021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33847565

RESUMO

In Proteobacteria, integral outer membrane proteins (OMPs) are crucial for the maintenance of the envelope permeability barrier to some antibiotics and detergents. In Enterobacteria, envelope stress caused by unfolded OMPs activates the sigmaE (σE) transcriptional response. σE upregulates OMP biogenesis factors, including the ß-barrel assembly machinery (BAM) that catalyses OMP folding. Here we report that DolP (formerly YraP), a σE-upregulated and poorly understood outer membrane lipoprotein, is crucial for fitness in cells that undergo envelope stress. We demonstrate that DolP interacts with the BAM complex by associating with outer membrane-assembled BamA. We provide evidence that DolP is important for proper folding of BamA that overaccumulates in the outer membrane, thus supporting OMP biogenesis and envelope integrity. Notably, mid-cell recruitment of DolP had been linked to regulation of septal peptidoglycan remodelling by an unknown mechanism. We now reveal that, during envelope stress, DolP loses its association with the mid-cell, thereby suggesting a mechanistic link between envelope stress caused by impaired OMP biogenesis and the regulation of a late step of cell division.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , Membrana Externa Bacteriana/fisiologia , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Lipoproteínas/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Aptidão Genética , Lipoproteínas/metabolismo , Dobramento de Proteína
7.
Mol Microbiol ; 116(1): 329-342, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33660879

RESUMO

The integrity of the cell envelope of E. coli relies on the concerted activity of multi-protein machineries that synthesize the peptidoglycan (PG) and the outer membrane (OM). Our previous work found that the depletion of lipopolysaccharide (LPS) export to the OM induces an essential PG remodeling process involving LD-transpeptidases (LDTs), the glycosyltransferase function of PBP1B and the carboxypeptidase PBP6a. Consequently, cells with defective OM biogenesis lyse if they lack any of these PG enzymes. Here we report that the morphological defects, and lysis associated with a ldtF mutant with impaired LPS transport, are alleviated by the loss of the predicted OM-anchored lipoprotein ActS (formerly YgeR). We show that ActS is an inactive member of LytM-type peptidoglycan endopeptidases due to a degenerated catalytic domain. ActS is capable of activating all three main periplasmic peptidoglycan amidases, AmiA, AmiB, and AmiC, which were previously reported to be activated only by EnvC and/or NlpD. Our data also suggest that in vivo ActS preferentially activates AmiC and that its function is linked to cell envelope stress.


Assuntos
Membrana Externa Bacteriana/fisiologia , Carboxipeptidases/metabolismo , Endopeptidases/metabolismo , Escherichia coli/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Carboxipeptidases/genética , Membrana Celular/fisiologia , Parede Celular/metabolismo , Endopeptidases/genética , Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Deleção de Genes , Lipopolissacarídeos/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/genética , Proteínas de Ligação às Penicilinas/metabolismo , Peptidoglicano/metabolismo , Peptidoglicano Glicosiltransferase/metabolismo , Plasmídeos/genética , D-Ala-D-Ala Carboxipeptidase Tipo Serina/metabolismo , Estresse Fisiológico/fisiologia
8.
Methods Mol Biol ; 2283: 123-130, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33765315

RESUMO

Outer membrane vesicles (OMV) shed by pathogenic bacteria have multifunctional roles in disease initiation and progression. Further, their efficacy as novel vaccines has underscored their importance as potential therapeutics. Consequently, to advance allied research related to their immunogenicity and pathogenicity it is important to separate these vesicular structures from parental cells and demonstrate them to be free from cellular debris and other non-vesicle-related constituents such as protein aggregates. To do so represents a key step in initiating OMV-related studies and the techniques and strategies adopted by the H. pylori community to achieve this will be the focus of this chapter.The key methods used typically to obtain a heterogeneous mixture of OMV (size range: ~20-300 nm in diameter) include growth of bacteria in broth culture followed by differential centrifugation, filtration, and concentration to separate OMV from the intact organisms. Additional measures may be adopted to further size-fractionate the population of OMV including gel filtration or density gradient ultra-centrifugation in order to facilitate differentiation between the activities of small versus large OMV, as recent studies have demonstrated differential modes of entry into host cells as well as size-dependent differences in the OMV proteome (Turner et al., Front Immunol 9:1466, 2018). The OMV from H. pylori harbor many of the virulence factors associated with gastric disease including the CagA oncoprotein, the cytotoxin VacA, and the HtrA protease (Olofsson et al., mBio 5:e00979-14, 2014; Mullaney et al., Proteomics Clin Appl 3:785-96, 2009) and their close association with areas of cell-cell contact and efficient endocytosis supports a role for these complexes in gastric disease (Turkina et al., FEMS Microbiol Lett 362:fnv076, 2015).


Assuntos
Membrana Externa Bacteriana/fisiologia , Helicobacter pylori/crescimento & desenvolvimento , Vesículas Transportadoras/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Técnicas Bacteriológicas , Contagem de Colônia Microbiana , Meios de Cultura , Precipitação Fracionada , Helicobacter pylori/metabolismo , Ultracentrifugação
9.
J Bacteriol ; 203(4)2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33257525

RESUMO

Many bacteria switch between a sessile and a motile mode in response to environmental and host-related signals. Porphyromonas gingivalis, an oral anaerobe implicated in the etiology of chronic periodontal disease, has long been described as a nonmotile bacterium. And yet, recent studies have shown that under certain conditions, P. gingivalis is capable of surface translocation. Considering these findings, this work aimed to increase our understanding of how P. gingivalis transitions between sessile growth and surface migration. Here, we show that the peptidylarginine deiminase secreted by P. gingivalis (PPAD), an enzyme previously shown to be upregulated during surface translocation and to constrain biofilm formation, promotes surface translocation. In the absence of PPAD, the production of outer membrane vesicles (OMVs) was drastically reduced. In turn, there was a reduction in gingipain-mediated proteolysis and a reduced zone of hydration around the site of inoculation. Transcriptome sequencing (RNA-Seq) and metabolomics analyses also showed that these changes corresponded to a shift in arginine metabolism. Overall, this report provides new evidence for the functional relevance of PPAD and proteases, as well as the importance of PPAD activity in OMV biogenesis and release. Our findings support the model that citrullination is a critical mechanism during lifestyle transition between surface-attached growth and surface translocation by modulating OMV-mediated proteolysis and arginine metabolism.IMPORTANCE Gram-negative bacteria produce nanosized OMVs that are actively released into their surroundings. The oral anaerobe P. gingivalis is prolific in OMV production, and many of the proteins packaged in these vesicles are proteolytic or protein-modifying enzymes. This includes key virulence determinants, such as the gingipains and PPAD (a unique peptidylarginine deiminase). Here, we show that PPAD activity (citrullination) is involved in OMV biogenesis. The study revealed an unusual mechanism that allows this bacterium to transform its surroundings. Since OMVs are detected in circulation and in systemic tissues, our study results also support the notion that PPAD activity may be a key factor in the correlation between periodontitis and systemic diseases, further supporting the idea of PPAD as an important therapeutic target.


Assuntos
Arginina/metabolismo , Membrana Externa Bacteriana/fisiologia , Proteínas de Bactérias/metabolismo , Porphyromonas gingivalis/fisiologia , Desiminases de Arginina em Proteínas/metabolismo , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Porphyromonas gingivalis/enzimologia , Desiminases de Arginina em Proteínas/genética
10.
Proc Natl Acad Sci U S A ; 117(45): 28026-28035, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33093201

RESUMO

The periplasmic chaperone network ensures the biogenesis of bacterial outer membrane proteins (OMPs) and has recently been identified as a promising target for antibiotics. SurA is the most important member of this network, both due to its genetic interaction with the ß-barrel assembly machinery complex as well as its ability to prevent unfolded OMP (uOMP) aggregation. Using only binding energy, the mechanism by which SurA carries out these two functions is not well-understood. Here, we use a combination of photo-crosslinking, mass spectrometry, solution scattering, and molecular modeling techniques to elucidate the key structural features that define how SurA solubilizes uOMPs. Our experimental data support a model in which SurA binds uOMPs in a groove formed between the core and P1 domains. This binding event results in a drastic expansion of the rest of the uOMP, which has many biological implications. Using these experimental data as restraints, we adopted an integrative modeling approach to create a sparse ensemble of models of a SurA•uOMP complex. We validated key structural features of the SurA•uOMP ensemble using independent scattering and chemical crosslinking data. Our data suggest that SurA utilizes three distinct binding modes to interact with uOMPs and that more than one SurA can bind a uOMP at a time. This work demonstrates that SurA operates in a distinct fashion compared to other chaperones in the OMP biogenesis network.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Escherichia coli/metabolismo , Chaperonas Moleculares/metabolismo , Peptidilprolil Isomerase/metabolismo , Membrana Externa Bacteriana/metabolismo , Membrana Externa Bacteriana/fisiologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Celular/metabolismo , Escherichia coli/enzimologia , Escherichia coli/metabolismo , Modelos Biológicos , Periplasma/metabolismo , Dobramento de Proteína
11.
Cell Biol Int ; 44(11): 2275-2283, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32749752

RESUMO

Serratia grimesii are facultative pathogenic bacteria that can penetrate a wide range of host cells and cause infection, especially in immunocompromised patients. Previously, we have found that bacterial metalloprotease grimelysin is a potential virulence determinant of S. grimesii invasion (E. S. Bozhokina et al., (2011). Cell Biology International, 35(2), 111-118). Protease is characterized as an actin-hydrolyzing enzyme with a narrow specificity toward other cell proteins. It is not known, however, whether grimelysin is transported into eukaryotic cells. Here, we show, for the first time, that S. grimesii can generate outer membrane vesicles (OMVs) displayed specific proteolytic activity against actin, characteristic of grimelysin. The presence of grimelysin was also confirmed by the Western blot analysis of S. grimesii OMVs lysate. Furthermore, confocal microscopy analysis revealed that the S. grimesii grimelysin-containing OMVs attached to the host cell membrane. Finally, pretreatment of HeLa cells with S. grimesii OMVs before the cells were infected with bacteria increased the bacterial penetration several times. These data strongly suggest that protease grimelysin promotes S. grimesii internalization by modifying bacterial and/or host molecule(s) when it is delivered as a component of OMVs.


Assuntos
Membrana Externa Bacteriana/metabolismo , Proteínas de Bactérias/metabolismo , Metaloproteases/metabolismo , Serratia/metabolismo , Actinas/metabolismo , Membrana Externa Bacteriana/fisiologia , Células Eucarióticas/metabolismo , Células Eucarióticas/microbiologia , Células HeLa , Humanos , Proteólise , Serratia/patogenicidade , Fatores de Virulência
12.
Mol Microbiol ; 114(6): 991-1005, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32808719

RESUMO

The outer membrane (OM) is an essential component of the Gram-negative bacterial envelope that protects the cells against external threats. To maintain a functional OM, cells require distinct mechanisms to ensure balance of proteins and lipids in the membrane. Mutations in OM biogenesis and/or homeostasis pathways often result in permeability defects, but how molecular changes in the OM affect barrier function is unclear. Here, we seek potential mechanism(s) that can alleviate permeability defects in Escherichia coli cells lacking the Tol-Pal complex, which accumulate excess PLs in the OM. We identify mutations in enterobacterial common antigen (ECA) biosynthesis that re-establish OM barrier function against large hydrophilic molecules, yet did not restore lipid homeostasis. Furthermore, we demonstrate that build-up of biosynthetic intermediates, but not loss of ECA itself, contributes to the rescue. This suppression of OM phenotypes is unrelated to known effects that accumulation of ECA intermediates have on the cell wall. Finally, we reveal that an unusual diacylglycerol pyrophosphoryl-linked lipid species also accumulates in ECA mutants, and might play a role in the rescue phenotype. Our work provides insights into how OM barrier function can be restored independent of lipid homeostasis, and highlights previously unappreciated effects of ECA-related species in OM biology.


Assuntos
Antígenos de Bactérias/genética , Proteínas da Membrana Bacteriana Externa/genética , Membrana Externa Bacteriana/fisiologia , Escherichia coli/genética , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Permeabilidade da Membrana Celular , Parede Celular/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Homeostase , Mutação , Proteínas Periplásmicas/genética , Proteínas Periplásmicas/metabolismo
13.
Proc Natl Acad Sci U S A ; 117(33): 20171-20179, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32747561

RESUMO

Extracellular electron transfer (EET) allows microorganisms to gain energy by linking intracellular reactions to external surfaces ranging from natural minerals to the electrodes of bioelectrochemical renewable energy technologies. In the past two decades, electrochemical techniques have been used to investigate EET in a wide range of microbes, with emphasis on dissimilatory metal-reducing bacteria, such as Shewanella oneidensis MR-1, as model organisms. However, due to the typically bulk nature of these techniques, they are unable to reveal the subpopulation variation in EET or link the observed electrochemical currents to energy gain by individual cells, thus overlooking the potentially complex spatial patterns of activity in bioelectrochemical systems. Here, to address these limitations, we use the cell membrane potential as a bioenergetic indicator of EET by S. oneidensis MR-1 cells. Using a fluorescent membrane potential indicator during in vivo single-cell-level fluorescence microscopy in a bioelectrochemical reactor, we demonstrate that membrane potential strongly correlates with EET. Increasing electrode potential and associated EET current leads to more negative membrane potential. This EET-induced membrane hyperpolarization is spatially limited to cells in contact with the electrode and within a near-electrode zone (<30 µm) where the hyperpolarization decays with increasing cell-electrode distance. The high spatial and temporal resolution of the reported technique can be used to study the single-cell-level dynamics of EET not only on electrode surfaces, but also during respiration of other solid-phase electron acceptors.


Assuntos
Membrana Externa Bacteriana/fisiologia , Transporte de Elétrons/fisiologia , Potenciais da Membrana/fisiologia , Shewanella/fisiologia , Benzotiazóis/metabolismo , Fenômenos Eletrofisiológicos , Corantes Fluorescentes , Análise de Célula Única/métodos , Gravação em Vídeo
14.
Vet Microbiol ; 247: 108778, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32768224

RESUMO

The pagC gene is ubiquitously distributed in Salmonella, but there is limited information regarding its function. Pullorum disease (PD) is a septicemic disease caused by Salmonella Pullorum, which also harbors the pagC gene. In this study, we constructed an S. Pullorum pagC gene deletion strain and its complemented strain. First, we confirmed that the pagC gene does not participate in bacterial growth regulation or environmental pH adaptation. Interestingly, the results of subsequent analyses indicated that the pagC gene defect led to increased bacterial colonization in the intestine (especially in the cecum) and increased biofilm formation, while the number of outer-membrane vesicles (OMVs) in the bacterial culture decreased. Purified OMVs were able to reduce S. Pullorum biofilm formation in vitro. In addition, the results of a mass spectrometry analysis of purified OMVs indicated that some enzymes harbored by OMVs may be involved in biofilm degradation. Based on these results, we conclude that deletion of the pagC gene leads to reduced S. Pullorum OMVs production, which subsequently promotes biofilm stability, increases bacterial colonization in the intestine, and potentially inhibits the switch from sessile to planktonic growth.


Assuntos
Membrana Externa Bacteriana/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Salmonella enterica/genética , Animais , Ceco/microbiologia , Galinhas/microbiologia , Deleção de Genes , Intestinos/microbiologia , Salmonella enterica/classificação , Salmonella enterica/fisiologia , Organismos Livres de Patógenos Específicos
15.
Mol Microbiol ; 114(5): 710-720, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32602138

RESUMO

Bacterial small proteins (below 50 amino acids) encoded by small open reading frames (sORFs) are recognized as an emerging class of functional molecules that have been largely overlooked in the past. While some were uncovered serendipitously, global approaches have recently been developed to detect these sORFs. A large portion of small proteins appears to be hydrophobic and located in the bacterial membrane. In the present review, we describe functional small hydrophobic proteins discovered in pathogenic bacteria and report recent advances in the discovery of additional ones. Small membrane proteins contribute to bacterial adaptation to changing environments and often appear to be implicated in negative feedback regulation loops by modulating the function or stability of larger membrane proteins. A subset of these proteins belongs to toxin-antitoxin modules. We highlight the features of characterized hydrophobic small proteins that may pave the way for identification of the functional small proteins among novel sORFs discovered. Besides providing new insights into bacterial pathogenesis, identification of naturally occurring small hydrophobic proteins of pathogenic bacteria can lead to new therapeutic interventions, as recently shown with the development of synthetic peptides derived from natural small proteins that display antibacterial or antivirulence properties.


Assuntos
Membrana Externa Bacteriana/metabolismo , Proteínas de Membrana/metabolismo , Bactérias/metabolismo , Bactérias/patogenicidade , Membrana Externa Bacteriana/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biologia Computacional/métodos , Interações Hidrofóbicas e Hidrofílicas , Proteínas de Membrana/fisiologia , Fases de Leitura Aberta/genética , Peptídeos/metabolismo , Proteômica/métodos , Ribossomos/metabolismo
16.
mBio ; 11(3)2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32430476

RESUMO

Activity of the NtrYX two-component system has been associated with important processes in diverse bacteria, ranging from symbiosis to nitrogen and energy metabolism. In the facultative alphaproteobacterium Rhodobacter sphaeroides, loss of the two-component system NtrYX results in increased lipid production and sensitivity to some known cell envelope-active compounds. In this study, we show that NtrYX directly controls multiple properties of the cell envelope. We find that the response regulator NtrX binds upstream of cell envelope genes, including those involved in peptidoglycan biosynthesis and modification and in cell division. We show that loss of NtrYX impacts the cellular levels of peptidoglycan precursors and lipopolysaccharide and alters cell envelope structure, increasing cell length and the thickness of the periplasm. Cell envelope function is also disrupted in the absence of NtrYX, resulting in increased outer membrane permeability. Based on the properties of R. sphaeroides cells lacking NtrYX and the target genes under direct control of this two-component system, we propose that NtrYX plays a previously undescribed, and potentially conserved, role in the assembly, structure, and function of the cell envelope in a variety of bacteria.IMPORTANCE The bacterial cell envelope provides many important functions. It protects cells from harsh environments, serves as a selective permeability barrier, houses bioenergetic functions, defines sensitivity to antibacterial agents, and plays a crucial role in biofilm formation, symbiosis, and virulence. Despite the important roles of this cellular compartment, we lack a detailed understanding of the biosynthesis and remodeling of the cell envelope. Here, we report that the R. sphaeroides two-component signaling system NtrYX is a previously undescribed regulator of cell envelope processes, providing evidence that it is directly involved in controlling transcription of genes involved in cell envelope assembly, structure, and function in this and possibly other bacteria. Thus, our data report on a newly discovered process used by bacteria to assemble and remodel the cell envelope.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , Membrana Externa Bacteriana/fisiologia , Regulação Bacteriana da Expressão Gênica , Rhodobacter sphaeroides/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Perfilação da Expressão Gênica , Rhodobacter sphaeroides/metabolismo , Transdução de Sinais
17.
mBio ; 11(3)2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32371598

RESUMO

Rod-shaped bacteria frequently localize proteins to one or both cell poles in order to regulate processes such as chromosome replication or polar organelle development. However, the roles of polar factors in responses to extracellular stimuli have been generally unexplored. We employed chemical-genetic screening to probe the interaction between one such factor from Caulobacter crescentus, TipN, and extracellular stress and found that TipN is required for normal resistance of cell envelope-directed antibiotics, including vancomycin which does not normally inhibit growth of Gram-negative bacteria. Forward genetic screening for suppressors of vancomycin sensitivity in the absence of TipN revealed the TonB-dependent receptor ChvT as the mediator of vancomycin sensitivity. Loss of ChvT improved resistance to vancomycin and cefixime in the otherwise sensitive ΔtipN strain. The activity of the two-component system regulating ChvT (ChvIG) was increased in ΔtipN cells relative to the wild type under some, but not all, cell wall stress conditions that this strain was sensitized to, in particular cefixime and detergent exposure. Together, these results indicate that TipN contributes to cell envelope stress resistance in addition to its roles in intracellular development, and its loss influences signaling through the ChvIG two-component system which has been co-opted as a sensor of cell wall stress in CaulobacterIMPORTANCE Maintenance of an intact cell envelope is essential for free-living bacteria to protect themselves against their environment. In the case of rod-shaped bacteria, the poles of the cell are potential weak points in the cell envelope due to the high curvature of the layers and the need to break and reform the cell envelope at the division plane as the cells divide. We have found that TipN, a factor required for correct division and cell pole development in Caulobacter crescentus, is also needed for maintaining normal levels of resistance to cell wall-targeting antibiotics such as vancomycin and cefixime, which interfere with peptidoglycan synthesis. Since TipN is normally located at the poles of the cell and at the division plane just before cells complete division, our results suggest that it is involved in stabilization of these weak points of the cell envelope as well as its other roles inside the cell.


Assuntos
Antibacterianos/farmacologia , Membrana Externa Bacteriana/fisiologia , Proteínas de Bactérias/metabolismo , Caulobacter crescentus/citologia , Peptidoglicano/metabolismo , Membrana Externa Bacteriana/efeitos dos fármacos , Proteínas de Bactérias/genética , Caulobacter crescentus/efeitos dos fármacos , Farmacorresistência Bacteriana/genética , Estresse Fisiológico
18.
Microb Pathog ; 144: 104180, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32240767

RESUMO

In bacterial cells, the cytoplasmic membrane forms a barrier between the environment and the cell's cytoplasm. This barrier regulates which substances (and the amount) that leave and enter the cell, to maintain homeostasis between the cytoplasm and the external environment. One of the mechanisms employed to maintain structure and functionality during exposure to environmental stress is adaptation of the membrane lipids. The aim of this study was to investigate membrane alteration as a possible survival method of non-acid adapted enteropathogenic Escherichia coli (E. coli) (EPEC) (as could be found in contaminated water or unprocessed food) through simulated gastric fluid (SGF). Enteropathogenic E. coli was grown in nutrient-rich media and then exposed to SGF of various pH (1.5, 2.5, 3.5, or 4.5) for 180 min. Flow cytometry was utilised to examine membrane integrity; and morphological changes were investigated using transmission electron microscopy (TEM). Gas chromatography-mass spectrometry (GC-MS) was used to assess the membrane lipid composition. The results of this study showed that SGF treatment caused membrane damage, as well as cell wall thickening and irregular plasma membranes. The morphological changes were accompanied by membrane lipid changes indicative of decreased membrane fluidity and increased rigidity. The findings suggest that non-acid adapted EPEC can perceive pH change in the environment and adapt accordingly.


Assuntos
Adaptação Fisiológica/fisiologia , Membrana Externa Bacteriana/fisiologia , Escherichia coli Enteropatogênica/metabolismo , Ácido Gástrico/metabolismo , Membrana Celular/metabolismo , Proteínas de Escherichia coli/metabolismo , Concentração de Íons de Hidrogênio , Lipídeos de Membrana/metabolismo , Estresse Fisiológico
19.
Microb Pathog ; 144: 104160, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32194181

RESUMO

Bacterial secretome is a comprehensive catalog of bacterial proteins that are released or secreted outside the cells. They offer a number of factors that possess several significant roles in virulence as well as cell to cell communication and hence play a core role in bacterial pathogenesis. Sometimes these proteins are bounded with membranes giving them the shape of vesicles called extracellular vesicles (EVs) or outer membrane vesicles (OMVs). Bacteria secrete these proteins via Sec and Tat pathways into the periplasm. Secreted proteins have found to be important as diagnostic markers as well as antigenic factors for the development of an effective candidate vaccine. Recently, the research in the field of secretomics is growing up and getting more interesting due to their direct involvement in the pathogenesis of the microorganisms leading to the infection. Many pathogenic bacteria have been studied for their secretome and the results illustrated novel antigens. This review highlights the secretome studies of different pathogenic bacteria in humans and animals, general secretion mechanisms, different approaches and challenges in the secretome of Mycoplasma sp.


Assuntos
Vesículas Extracelulares/fisiologia , Mycoplasma/metabolismo , Mycoplasma/patogenicidade , Percepção de Quorum/fisiologia , Fatores de Virulência/metabolismo , Membrana Externa Bacteriana/fisiologia , Transporte Proteico/fisiologia , Proteoma/metabolismo , Transdução de Sinais/fisiologia
20.
Biochim Biophys Acta Biomembr ; 1862(6): 183239, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32119861

RESUMO

Soil bacteria are decomposer organisms crucial for the biodegradation of organic pollutants, mineralization of dead organic matter and the turnover of biogenic elements. In their environment they are constantly exposed to membrane-lytic enzymes emitted to the soil by other microorganisms competing for the same niche. Therefore, the composition and structure of their membranes is of utmost importance for survival in the harsh environment. Although soil bacteria species can be Gram-negative or Gram-positive and their membranes differ significantly, they are formed by phospholipids belonging mainly to three classes: phosphatidylethanolamines (PE), phosphatidylglycerols (PG) and cardiolipins (CL). The correlation of the membrane phospholipid composition and its susceptibility to secretory membrane-lytic enzymes is widely unknown; thus, to shed light on these phenomena we applied the Langmuir monolayer technique to construct models of soil bacteria membranes differing in the mutual proportion of the main phospholipids. To characterize the systems we studied their elasticity, mesoscopic texture, 2D crystalline structure and discussed the thermodynamics of the interactions between their components. The model membranes were exposed to secretory phospholipase A2. It turned out that in spite of the structural similarities the model membranes differed significantly in their susceptibility to s-PLA2 attack. The membranes devoid of cardiolipin were completely degraded, whereas, these containing cardiolipin were much more resistant to the enzymatic hydrolysis. It also turned out that the sole presence of cardiolipin in the model membrane did not guarantee the membrane durability and that the interplay between cardiolipin and the zwitterionic phosphatidylethanolamine was here of crucial importance.


Assuntos
Membrana Externa Bacteriana/química , Cardiolipinas/fisiologia , Membranas Artificiais , Fosfolipases A2 Secretórias/metabolismo , Fosfolipídeos/química , Membrana Externa Bacteriana/fisiologia , Cardiolipinas/análise , Modelos Biológicos , Fosfatidiletanolaminas/análise , Fosfatidilgliceróis/análise , Fosfolipídeos/análise , Microbiologia do Solo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...