Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Langmuir ; 40(14): 7353-7363, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38536768

RESUMEN

Nanomaterials of zinc oxide (ZnO) exhibit antibacterial activities under ambient illumination that result in cell membrane permeability and disorganization, representing an important opportunity for health-related applications. However, the development of antibiofouling surfaces incorporating ZnO nanomaterials has remained limited. In this work, we fabricate superhydrophobic surfaces based on ZnO nanopillars. Water droplets on these superhydrophobic surfaces exhibit small contact angle hysteresis (within 2-3°) and a minimal tilting angle of 1°. Further, falling droplets bounce off when impacting the superhydrophobic ZnO surfaces with a range of Weber numbers (8-46), demonstrating that the surface facilitates a robust Cassie-Baxter wetting state. In addition, the antibiofouling efficacy of the surfaces has been established against model pathogenic Gram-positive bacteria Staphylococcus aureus (S. aureus) and Gram-negative bacteria Escherichia coli (E. coli). No viable colonies of E. coli were recoverable on the superhydrophobic surfaces of ZnO nanopillars incubated with cultured bacterial solutions for 18 h. Further, our tests demonstrate a substantial reduction in the quantity of S. aureus that attached to the superhydrophobic ZnO nanopillars. Thus, the superhydrophobic ZnO surfaces offer a viable design of antibiofouling materials that do not require additional UV illumination or antimicrobial agents.


Asunto(s)
Óxido de Zinc , Humectabilidad , Óxido de Zinc/farmacología , Óxido de Zinc/química , Propiedades de Superficie , Escherichia coli , Staphylococcus aureus , Antibacterianos/farmacología , Antibacterianos/química
2.
mBio ; 15(4): e0032524, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38426748

RESUMEN

Gram-negative bacteria have a thin peptidoglycan layer between the cytoplasmic and outer membranes protecting the cell from osmotic challenges. Hydrolases of this structure are needed to cleave bonds to allow the newly synthesized peptidoglycan strands to be inserted by synthases. These enzymes need to be tightly regulated and their activities coordinated to prevent cell lysis. To better understand this process in Escherichia coli, we probed the genetic interactions of mrcA (encodes PBP1A) and mrcB (encodes PBP1B) with genes encoding peptidoglycan amidases and endopeptidases in envelope stress conditions. Our extensive genetic interaction network analysis revealed relatively few combinations of hydrolase gene deletions with reduced fitness in the absence of PBP1A or PBP1B, showing that none of the amidases or endopeptidases is strictly required for the functioning of one of the class A PBPs. This illustrates the robustness of the peptidoglycan growth mechanism. However, we discovered that the fitness of ∆mrcB cells is significantly reduced under high salt stress and in vitro activity assays suggest that this phenotype is caused by a reduced peptidoglycan synthesis activity of PBP1A at high salt concentration.IMPORTANCEEscherichia coli and many other bacteria have a surprisingly high number of peptidoglycan hydrolases. These enzymes function in concert with synthases to facilitate the expansion of the peptidoglycan sacculus under a range of growth and stress conditions. The synthases PBP1A and PBP1B both contribute to peptidoglycan expansion during cell division and growth. Our genetic interaction analysis revealed that these two penicillin-binding proteins (PBPs) do not need specific amidases, endopeptidases, or lytic transglycosylases for function. We show that PBP1A and PBP1B do not work equally well when cells encounter high salt stress and demonstrate that PBP1A alone cannot provide sufficient PG synthesis activity under this condition. These results show how the two class A PBPs and peptidoglycan hydrolases govern cell envelope integrity in E. coli in response to environmental challenges and particularly highlight the importance of PBP1B in maintaining cell fitness under high salt conditions.


Asunto(s)
Proteínas de Escherichia coli , Peptidoglicano Glicosiltransferasa , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Peptidoglicano/metabolismo , Peptidoglicano Glicosiltransferasa/metabolismo , Proteínas de Unión a las Penicilinas/metabolismo , Pared Celular/metabolismo , Endopeptidasas/genética , Endopeptidasas/metabolismo , Amidohidrolasas/genética , Amidohidrolasas/metabolismo
3.
bioRxiv ; 2023 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-37961452

RESUMEN

Mycobacterial glycolipids are important cell envelope structures that drive host-pathogen interactions. Arguably, the most important amongst these are lipoarabinomannan (LAM) and its precursor, lipomannan (LM), which are both trafficked out of the bacterium to the host via unknown mechanisms. An important class of exported LM/LAM is the capsular derivative of these molecules which is devoid of its lipid anchor. Here, we describe the identification of a glycoside hydrolase family 76 enzyme that we term LamH which specifically cleaves α-1,6-mannoside linkages within LM and LAM, driving its export to the capsule releasing its phosphatidyl-myo-inositol mannoside lipid anchor. Unexpectedly, we found that the catalytic activity of this enzyme is important for efficient exit from stationary phase cultures where arabinomannan acts as a signal for growth phase transition. Finally, we demonstrate that LamH is important for Mycobacterium tuberculosis survival in macrophages. These data provide a new framework for understanding the biological role of LAM in mycobacteria.

4.
Bioinformatics ; 39(7)2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37348862

RESUMEN

MOTIVATION: Wastewater treatment plants (WWTPs) harbor a dense and diverse microbial community. They constantly receive antimicrobial residues and resistant strains, and therefore provide conditions for horizontal gene transfer (HGT) of antimicrobial resistance (AMR) determinants. This facilitates the transmission of clinically important genes between, e.g. enteric and environmental bacteria, and vice versa. Despite the clinical importance, tools for predicting HGT remain underdeveloped. RESULTS: In this study, we examined to which extent water cycle microbial community composition, as inferred by partial 16S rRNA gene sequences, can predict plasmid permissiveness, i.e. the ability of cells to receive a plasmid through conjugation, based on data from standardized filter mating assays using fluorescent bio-reporter plasmids. We leveraged a range of machine learning models for predicting the permissiveness for each taxon in the community, representing the range of hosts a plasmid is able to transfer to, for three broad host-range resistance IncP plasmids (pKJK5, pB10, and RP4). Our results indicate that the predicted permissiveness from the best performing model (random forest) showed a moderate-to-strong average correlation of 0.49 for pB10 [95% confidence interval (CI): 0.44-0.55], 0.43 for pKJK5 (0.95% CI: 0.41-0.49), and 0.53 for RP4 (0.95% CI: 0.48-0.57) with the experimental permissiveness in the unseen test dataset. Predictive phylogenetic signals occurred despite the broad host-range nature of these plasmids. Our results provide a framework that contributes to the assessment of the risk of AMR pollution in wastewater systems. AVAILABILITY AND IMPLEMENTATION: The predictive tool is available as an application at https://github.com/DaneshMoradigaravand/PlasmidPerm.


Asunto(s)
Microbiota , Aguas Residuales , ARN Ribosómico 16S/genética , Filogenia , Tolerancia , Plásmidos/genética , Transferencia de Gen Horizontal
5.
Bioinformatics ; 39(4)2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-37014365

RESUMEN

MOTIVATION: High-throughput chemical genomic screens produce informative datasets, providing valuable insights into unknown gene function on a genome-wide level. However, there is currently no comprehensive analytic package publicly available. We developed ChemGAPP to bridge this gap. ChemGAPP integrates various steps in a streamlined and user-friendly format, including rigorous quality control measures to curate screening data. RESULTS: ChemGAPP provides three sub-packages for different chemical-genomic screens: ChemGAPP Big for large-scale screens; ChemGAPP Small for small-scale screens; and ChemGAPP GI for genetic interaction screens. ChemGAPP Big, tested against the Escherichiacoli KEIO collection, revealed reliable fitness scores which displayed biologically relevant phenotypes. ChemGAPP Small demonstrated significant changes in phenotype in a small-scale screen. ChemGAPP GI was benchmarked against three sets of genes with known epistasis types and successfully reproduced each interaction type. AVAILABILITY AND IMPLEMENTATION: ChemGAPP is available at https://github.com/HannahMDoherty/ChemGAPP, as a standalone Python package as well as Streamlit applications.


Asunto(s)
Genómica , Programas Informáticos , Genoma , Fenotipo , Pruebas Genéticas
6.
J Biomed Mater Res B Appl Biomater ; 111(2): 241-260, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36054531

RESUMEN

In vitro cytotoxicity assessment is indispensable in developing new biodegradable implant materials. Zn, which demonstrates an ideal corrosion rate between Mg- and Fe-based alloys, has been reported to have excellent in vivo biocompatibility. Therefore, modifications aimed at improving Zn's mechanical properties should not degrade its biological response. As sufficient strength, ductility and corrosion behavior required of load-bearing implants has been obtained in plastically deformed Zn-3Ag-0.5Mg, the effect of simultaneous Ag and Mg additions on in vitro cytocompatibility and antibacterial properties was studied, in relation to Zn and Zn-3Ag. Direct cell culture on samples and indirect extract-based tests showed almost no significant differences between the tested Zn-based materials. The diluted extracts of Zn, Zn-3Ag, and Zn-3Ag-0.5Mg showed no cytotoxicity toward MG-63 cells at a concentration of ≤12.5%. The cytotoxic effect was observed only at high Zn2+ ion concentrations and when in direct contact with metallic samples. The highest LD50 (lethal dose killing 50% of cells) of 13.4 mg/L of Zn2+ ions were determined for the Zn-3Ag-0.5Mg. Similar antibacterial activity against Escherichia coli and Staphylococcus aureus was observed for Zn and Zn alloys, so the effect is attributed mainly to the released Zn2+ ions exhibiting bactericidal properties. Most importantly, our experiments indicated the limitations of water-soluble tetrazolium salt-based cytotoxicity assays for direct tests on Zn-based materials. The discrepancies between the WST-8 assay and SEM observations are attributed to the interference of Zn2+ ions with tetrazolium salt, therefore favoring its transformation into formazan, giving false cell viability quantitative results.


Asunto(s)
Implantes Absorbibles , Aleaciones , Aleaciones/farmacología , Ensayo de Materiales , Línea Celular , Corrosión , Antibacterianos/farmacología , Escherichia coli , Iones , Zinc/farmacología , Sales de Tetrazolio/farmacología , Materiales Biocompatibles/farmacología
7.
Microbiol Spectr ; 10(4): e0083322, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35856675

RESUMEN

In recent years the availability of genome sequence information has grown logarithmically resulting in the identification of a plethora of uncharacterized genes. To address this gap in functional annotation, many high-throughput screens have been devised to uncover novel gene functions. Gene-replacement libraries are one such tool that can be screened in a high-throughput way to link genotype and phenotype and are key community resources. However, for a phenotype to be attributed to a specific gene, there needs to be confidence in the genotype. Construction of large libraries can be laborious and occasionally errors will arise. Here, we present a rapid and accurate method for the validation of any ordered library where a gene has been replaced or disrupted by a uniform linear insertion (LI). We applied our method (LI-detector) to the well-known Keio library of Escherichia coli gene-deletion mutants. Our method identified 3,718 constructed mutants out of a total of 3,728 confirmed isolates, with a success rate of 99.7% for identifying the correct kanamycin cassette position. This data set provides a benchmark for the purity of the Keio mutants and a screening method for mapping the position of any linear insertion, such as an antibiotic resistance cassette in any ordered library. IMPORTANCE The construction of ordered gene replacement libraries requires significant investment of time and resources to create a valuable community resource. During construction, technical errors may result in a limited number of incorrect mutants being made. Such mutants may confound the output of subsequent experiments. Here, using the remarkable E. coli Keio knockout library, we describe a method to rapidly validate the construction of every mutant.


Asunto(s)
Elementos Transponibles de ADN , Infecciones por Escherichia coli , Escherichia coli/genética , Biblioteca de Genes , Humanos , Mutagénesis Insercional
8.
Microbiology (Reading) ; 168(5)2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35604759

RESUMEN

Bacterial amidases are essential to split the shared envelope of adjunct daughter cells to allow cell separation. Their activity needs to be precisely controlled to prevent cell lysis. In Escherichia coli, amidase activity is controlled by three regulatory proteins NlpD, EnvC and ActS. However, recent studies linked the outer membrane lipoprotein DolP (formerly YraP) as a potential upstream regulator of NlpD. In this study we explored this link in further detail. To our surprise DolP did not modulate amidase activity in vitro and was unable to interact with NlpD in pull-down and MST (MicroScale Thermophoresis) assays. Next, we excluded the hypothesis that ΔdolP phenocopied ΔnlpD in a range of envelope stresses. However, morphological analysis of double deletion mutants of amidases (AmiA, AmiB AmiC) and amidase regulators with dolP revealed that ΔamiAΔdolP and ΔenvCΔdolP mutants display longer chain length compared to their parental strains indicating a role for DolP in cell division. Overall, we present evidence that DolP does not affect NlpD function in vitro, implying that DolP is not an upstream regulator of NlpD. However, DolP may impact daughter cell separation by interacting directly with AmiA or AmiC, or by a yet undiscovered mechanism.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Separación Celular , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Lipoproteínas/genética , Lipoproteínas/metabolismo , Peptidoglicano/metabolismo
9.
PLoS Genet ; 18(5): e1010222, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35604931

RESUMEN

Insertion of new material into the Escherichia coli peptidoglycan (PG) sacculus between the cytoplasmic membrane and the outer membrane requires a well-organized balance between synthetic and hydrolytic activities to maintain cell shape and avoid lysis. Since most bacteria carry multiple enzymes carrying the same type of PG hydrolytic activity, we know little about the specific function of given enzymes. Here we show that the DD-carboxy/endopeptidase PBP4 localizes in a PBP1A/LpoA and FtsEX dependent fashion at midcell during septal PG synthesis. Midcell localization of PBP4 requires its non-catalytic domain 3 of unknown function, but not the activity of PBP4 or FtsE. Microscale thermophoresis with isolated proteins shows that PBP4 interacts with NlpI and the FtsEX-interacting protein EnvC, an activator of amidases AmiA and AmiB, which are needed to generate denuded glycan strands to recruit the initiator of septal PG synthesis, FtsN. The domain 3 of PBP4 is needed for the interaction with NlpI and EnvC, but not PBP1A or LpoA. In vivo crosslinking experiments confirm the interaction of PBP4 with PBP1A and LpoA. We propose that the interaction of PBP4 with EnvC, whilst not absolutely necessary for mid-cell recruitment of either protein, coordinates the activities of PBP4 and the amidases, which affects the formation of denuded glycan strands that attract FtsN. Consistent with this model, we found that the divisome assembly at midcell was premature in cells lacking PBP4, illustrating how the complexity of interactions affect the timing of cell division initiation.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Transportadoras de Casetes de Unión a ATP/metabolismo , Amidohidrolasas/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Endopeptidasas , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Lipoproteínas/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Peptidoglicano/metabolismo
10.
PLoS Genet ; 17(12): e1009586, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34941903

RESUMEN

The cell envelope is essential for viability in all domains of life. It retains enzymes and substrates within a confined space while providing a protective barrier to the external environment. Destabilising the envelope of bacterial pathogens is a common strategy employed by antimicrobial treatment. However, even in one of the best studied organisms, Escherichia coli, there remain gaps in our understanding of how the synthesis of the successive layers of the cell envelope are coordinated during growth and cell division. Here, we used a whole-genome phenotypic screen to identify mutants with a defective cell envelope. We report that loss of yhcB, a conserved gene of unknown function, results in loss of envelope stability, increased cell permeability and dysregulated control of cell size. Using whole genome transposon mutagenesis strategies, we report the comprehensive genetic interaction network of yhcB, revealing all genes with a synthetic negative and a synthetic positive relationship. These genes include those previously reported to have a role in cell envelope biogenesis. Surprisingly, we identified genes previously annotated as essential that became non-essential in a ΔyhcB background. Subsequent analyses suggest that YhcB functions at the junction of several envelope biosynthetic pathways coordinating the spatiotemporal growth of the cell, highlighting YhcB as an as yet unexplored antimicrobial target.


Asunto(s)
Pared Celular/genética , Proteínas de Escherichia coli/genética , Lipopolisacáridos/genética , Oxidorreductasas/genética , Peptidoglicano/genética , División Celular/genética , Membrana Celular/genética , Membrana Celular/microbiología , Pared Celular/microbiología , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica/genética , Lipopolisacáridos/biosíntesis , Mutagénesis , Fosfolípidos/biosíntesis , Fosfolípidos/genética
11.
Int J Mol Sci ; 22(23)2021 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-34884635

RESUMEN

Bacteria must maintain the ability to modify and repair the peptidoglycan layer without jeopardising its essential functions in cell shape, cellular integrity and intermolecular interactions. A range of new experimental techniques is bringing an advanced understanding of how bacteria regulate and achieve peptidoglycan synthesis, particularly in respect of the central role played by complexes of Sporulation, Elongation or Division (SEDs) and class B penicillin-binding proteins required for cell division, growth and shape. In this review we highlight relationships implicated by a bioinformatic approach between the outer membrane, cytoskeletal components, periplasmic control proteins, and cell elongation/division proteins to provide further perspective on the interactions of these cell division, growth and shape complexes. We detail the network of protein interactions that assist in the formation of peptidoglycan and highlight the increasingly dynamic and connected set of protein machinery and macrostructures that assist in creating the cell envelope layers in Gram-negative bacteria.


Asunto(s)
Membrana Celular/metabolismo , Bacterias Gramnegativas/metabolismo , Proteínas de Unión a las Penicilinas/metabolismo , Peptidoglicano/metabolismo , Proteínas Periplasmáticas/metabolismo , Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo
12.
Nature ; 597(7877): 533-538, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34497420

RESUMEN

Bacteria in the gut can modulate the availability and efficacy of therapeutic drugs. However, the systematic mapping of the interactions between drugs and bacteria has only started recently1 and the main underlying mechanism proposed is the chemical transformation of drugs by microorganisms (biotransformation). Here we investigated the depletion of 15 structurally diverse drugs by 25 representative strains of gut bacteria. This revealed 70 bacteria-drug interactions, 29 of which had not to our knowledge been reported before. Over half of the new interactions can be ascribed to bioaccumulation; that is, bacteria storing the drug intracellularly without chemically modifying it, and in most cases without the growth of the bacteria being affected. As a case in point, we studied the molecular basis of bioaccumulation of the widely used antidepressant duloxetine by using click chemistry, thermal proteome profiling and metabolomics. We find that duloxetine binds to several metabolic enzymes and changes the metabolite secretion of the respective bacteria. When tested in a defined microbial community of accumulators and non-accumulators, duloxetine markedly altered the composition of the community through metabolic cross-feeding. We further validated our findings in an animal model, showing that bioaccumulating bacteria attenuate the behavioural response of Caenorhabditis elegans to duloxetine. Together, our results show that bioaccumulation by gut bacteria may be a common mechanism that alters drug availability and bacterial metabolism, with implications for microbiota composition, pharmacokinetics, side effects and drug responses, probably in an individual manner.


Asunto(s)
Bacterias/metabolismo , Bioacumulación , Clorhidrato de Duloxetina/metabolismo , Microbioma Gastrointestinal/fisiología , Animales , Antidepresivos/metabolismo , Antidepresivos/farmacocinética , Caenorhabditis elegans/metabolismo , Células/metabolismo , Química Clic , Clorhidrato de Duloxetina/efectos adversos , Clorhidrato de Duloxetina/farmacocinética , Humanos , Metabolómica , Modelos Animales , Proteómica , Reproducibilidad de los Resultados
13.
Mol Microbiol ; 116(1): 329-342, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33660879

RESUMEN

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.


Asunto(s)
Membrana Externa Bacteriana/fisiología , Carboxipeptidasas/metabolismo , Endopeptidasas/metabolismo , Escherichia coli/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Carboxipeptidasas/genética , Membrana Celular/fisiología , Pared Celular/metabolismo , Endopeptidasas/genética , Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Eliminación de Gen , Lipopolisacáridos/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/genética , Proteínas de Unión a las Penicilinas/metabolismo , Peptidoglicano/metabolismo , Peptidoglicano Glicosiltransferasa/metabolismo , Plásmidos/genética , D-Ala-D-Ala Carboxipeptidasa de Tipo Serina/metabolismo , Estrés Fisiológico/fisiología
14.
Elife ; 92020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-33315009

RESUMEN

The Gram-negative outer-membrane envelops the bacterium and functions as a permeability barrier against antibiotics, detergents, and environmental stresses. Some virulence factors serve to maintain the integrity of the outer membrane, including DolP (formerly YraP) a protein of unresolved structure and function. Here, we reveal DolP is a lipoprotein functionally conserved amongst Gram-negative bacteria and that loss of DolP increases membrane fluidity. We present the NMR solution structure for Escherichia coli DolP, which is composed of two BON domains that form an interconnected opposing pair. The C-terminal BON domain binds anionic phospholipids through an extensive membrane:protein interface. This interaction is essential for DolP function and is required for sub-cellular localisation of the protein to the cell division site, providing evidence of subcellular localisation of these phospholipids within the outer membrane. The structure of DolP provides a new target for developing therapies that disrupt the integrity of the bacterial cell envelope.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Membrana Celular/metabolismo , Proteínas de Escherichia coli/metabolismo , Transporte de Proteínas/fisiología , Antibacterianos/metabolismo , Pared Celular/metabolismo , Escherichia coli/metabolismo , Bacterias Gramnegativas/metabolismo , Lipoproteínas/metabolismo , Factores de Virulencia/metabolismo
15.
J Bacteriol ; 203(2)2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33106348

RESUMEN

The asymmetric Gram-negative outer membrane (OM) is the first line of defense for bacteria against environmental insults and attack by antimicrobials. The key component of the OM is lipopolysaccharide, which is transported to the surface by the essential lipopolysaccharide transport (Lpt) system. Correct folding of the Lpt system component LptD is regulated by a periplasmic metalloprotease, BepA. Here, we present the crystal structure of BepA from Escherichia coli, solved to a resolution of 2.18 Å, in which the M48 protease active site is occluded by an active-site plug. Informed by our structure, we demonstrate that free movement of the active-site plug is essential for BepA function, suggesting that the protein is autoregulated by the active-site plug, which is conserved throughout the M48 metalloprotease family. Targeted mutagenesis of conserved residues reveals that the negative pocket and the tetratricopeptide repeat (TPR) cavity are required for function and degradation of the BAM complex component BamA under conditions of stress. Last, we show that loss of BepA causes disruption of OM lipid asymmetry, leading to surface exposed phospholipid.IMPORTANCE M48 metalloproteases are widely distributed in all domains of life. E. coli possesses four members of this family located in multiple cellular compartments. The functions of these proteases are not well understood. Recent investigations revealed that one family member, BepA, has an important role in the maturation of a central component of the lipopolysaccharide (LPS) biogenesis machinery. Here, we present the structure of BepA and the results of a structure-guided mutagenesis strategy, which reveal the key residues required for activity that inform how all M48 metalloproteases function.


Asunto(s)
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Metaloproteasas/química , Metaloproteasas/metabolismo , Secuencia de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Cristalografía por Rayos X , Proteínas de Escherichia coli/aislamiento & purificación , Metaloproteasas/aislamiento & purificación , Permeabilidad , Sensibilidad y Especificidad , Relación Estructura-Actividad
16.
FASEB J ; 34(5): 6284-6301, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32201986

RESUMEN

Mitophagy is a key process regulating mitochondrial quality control. Several mechanisms have been proposed to regulate mitophagy, but these have mostly been studied using stably expressed non-native proteins in immortalized cell lines. In skeletal muscle, mitophagy and its molecular mechanisms require more thorough investigation. To measure mitophagy directly, we generated a stable skeletal muscle C2C12 cell line, expressing a mitophagy reporter construct (mCherry-green fluorescence protein-mtFIS1101-152 ). Here, we report that both carbonyl cyanide m-chlorophenyl hydrazone (CCCP) treatment and adenosine monophosphate activated protein kinase (AMPK) activation by 991 promote mitochondrial fission via phosphorylation of MFF and induce mitophagy by ~20%. Upon CCCP treatment, but not 991, ubiquitin phosphorylation, a read-out of PTEN-induced kinase 1 (PINK1) activity, and Parkin E3 ligase activity toward CDGSH iron sulfur domain 1 (CISD1) were increased. Although the PINK1-Parkin signaling pathway is active in response to CCCP treatment, we observed no change in markers of mitochondrial protein content. Interestingly, our data shows that TANK-binding kinase 1 (TBK1) phosphorylation is increased after both CCCP and 991 treatments, suggesting TBK1 activation to be independent of both PINK1 and Parkin. Finally, we confirmed in non-muscle cell lines that TBK1 phosphorylation occurs in the absence of PINK1 and is regulated by AMPK-dependent signaling. Thus, AMPK activation promotes mitophagy by enhancing mitochondrial fission (via MFF phosphorylation) and autophagosomal engulfment (via TBK1 activation) in a PINK1-Parkin independent manner.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Dinámicas Mitocondriales , Mitofagia , Músculo Esquelético/patología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Activación Enzimática , Células HeLa , Humanos , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Ionóforos de Protónes/farmacología , Transducción de Señal , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
17.
IEEE Trans Nanobioscience ; 19(3): 394-402, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32142451

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen with a large repertoire of virulence factors that allow it to cause acute and chronic infections. Treatment of P. aeruginosa infections often fail due to its antibiotic resistance mechanisms, thus novel strategies aim at targeting virulence factors instead of growth-related features. Although the elements of the virulence networks of P. aeruginosa have been identified, how they interact and influence the overall virulence regulation is unclear. In this study, we reconstructed the signaling and transcriptional regulatory networks of 12 acute and 8 chronic virulence factors, and the 4 quorum sensing systems of P. aeruginosa. Using Boolean modelling, we showed that the static interactions and the time when they take place are important features in the quorum sensing network. We also found that the virulence factors of the acute networks are under strict repression or non-strict activation, while those of most of the chronic networks are under repression. In conclusion, Boolean modelling provides a system-level view of the P. aeruginosa virulence and quorum sensing networks to gain new insights into the various mechanisms that support its pathogenicity. Thus, we suggest that Boolean modelling could be used to guide the design of new treatments against P. aeruginosa.


Asunto(s)
Farmacorresistencia Bacteriana/fisiología , Modelos Biológicos , Pseudomonas aeruginosa , Percepción de Quorum/fisiología , Antibacterianos/farmacología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Pseudomonas aeruginosa/fisiología , Factores de Virulencia/química , Factores de Virulencia/metabolismo
18.
EMBO J ; 39(5): e102246, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-32009249

RESUMEN

The peptidoglycan (PG) sacculus provides bacteria with the mechanical strength to maintain cell shape and resist osmotic stress. Enlargement of the mesh-like sacculus requires the combined activity of peptidoglycan synthases and hydrolases. In Escherichia coli, the activity of two PG synthases is driven by lipoproteins anchored in the outer membrane (OM). However, the regulation of PG hydrolases is less well understood, with only regulators for PG amidases having been described. Here, we identify the OM lipoprotein NlpI as a general adaptor protein for PG hydrolases. NlpI binds to different classes of hydrolases and can specifically form complexes with various PG endopeptidases. In addition, NlpI seems to contribute both to PG elongation and division biosynthetic complexes based on its localization and genetic interactions. Consistent with such a role, we reconstitute PG multi-enzyme complexes containing NlpI, the PG synthesis regulator LpoA, its cognate bifunctional synthase, PBP1A, and different endopeptidases. Our results indicate that peptidoglycan regulators and adaptors are part of PG biosynthetic multi-enzyme complexes, regulating and potentially coordinating the spatiotemporal action of PG synthases and hydrolases.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Lipoproteínas/metabolismo , Complejos Multienzimáticos , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Pared Celular/enzimología , Endopeptidasas/genética , Endopeptidasas/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Lipoproteínas/genética , N-Acetil Muramoil-L-Alanina Amidasa/genética , Peptidoglicano/metabolismo
19.
ACS Infect Dis ; 6(4): 603-612, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-31851822

RESUMEN

Biofilm inhibition by exogenous molecules has been an attractive strategy for the development of novel therapeutics. We investigated the biofilm inhibitor taurolithocholic acid (TLCA) and its effects on the specialized metabolism, virulence, and biofilm formation of the clinically relevant bacterium Pseudomonas aeruginosa strain PA14. Our study shows that TLCA alters the specialized metabolism, thereby affecting P. aeruginosa colony biofilm physiology. We observed an upregulation of metabolites correlated to virulence such as the siderophore pyochelin. A wax moth virulence assay confirmed that treatment with TLCA increases the virulence of P. aeruginosa. On the basis of our results, we believe that future endeavors to identify biofilm inhibitors must consider how a putative lead alters the specialized metabolism of a bacterial community to prevent pathogens from entering a highly virulent state.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Ácido Taurolitocólico/farmacología , Biopelículas/crecimiento & desarrollo , Redes y Vías Metabólicas/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Virulencia/efectos de los fármacos
20.
Structure ; 27(12): 1842-1854.e4, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31653338

RESUMEN

The only enzyme responsible for cadaverine production in the major multidrug-resistant human pathogen Pseudomonas aeruginosa is the lysine decarboxylase LdcA. This enzyme modulates the general polyamine homeostasis, promotes growth, and reduces bacterial persistence during carbenicillin treatment. Here we present a 3.7-Å resolution cryoelectron microscopy structure of LdcA. We introduce an original approach correlating phylogenetic signal with structural information and reveal possible recombination among LdcA and arginine decarboxylase subfamilies within structural domain boundaries. We show that LdcA is involved in full virulence in an insect pathogenesis model. Furthermore, unlike its enterobacterial counterparts, LdcA is regulated neither by the stringent response alarmone ppGpp nor by the AAA+ ATPase RavA. Instead, the P. aeruginosa ravA gene seems to play a defensive role. Altogether, our study identifies LdcA as an important player in P. aeruginosa physiology and virulence and as a potential drug target.


Asunto(s)
Proteínas Bacterianas/química , Carboxiliasas/química , Evolución Molecular , Pseudomonas aeruginosa/enzimología , Factores de Virulencia/química , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Carboxiliasas/genética , Carboxiliasas/metabolismo , Microscopía por Crioelectrón , Expresión Génica , Humanos , Cinética , Modelos Moleculares , Filogenia , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Estructura Terciaria de Proteína , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/clasificación , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Recombinación Genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...