Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 246
Filtrar
1.
Nature ; 626(7999): 617-625, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38081298

RESUMEN

The outer membrane in Gram-negative bacteria consists of an asymmetric phospholipid-lipopolysaccharide bilayer that is densely packed with outer-membrane ß-barrel proteins (OMPs) and lipoproteins1. The architecture and composition of this bilayer is closely monitored and is essential to cell integrity and survival2-4. Here we find that SlyB, a lipoprotein in the PhoPQ stress regulon, forms stable stress-induced complexes with the outer-membrane proteome. SlyB comprises a 10 kDa periplasmic ß-sandwich domain and a glycine zipper domain that forms a transmembrane α-helical hairpin with discrete phospholipid- and lipopolysaccharide-binding sites. After loss in lipid asymmetry, SlyB oligomerizes into ring-shaped transmembrane complexes that encapsulate ß-barrel proteins into lipid nanodomains of variable size. We find that the formation of SlyB nanodomains is essential during lipopolysaccharide destabilization by antimicrobial peptides or acute cation shortage, conditions that result in a loss of OMPs and compromised outer-membrane barrier function in the absence of a functional SlyB. Our data reveal that SlyB is a compartmentalizing transmembrane guard protein that is involved in cell-envelope proteostasis and integrity, and suggest that SlyB represents a larger family of broadly conserved lipoproteins with 2TM glycine zipper domains with the ability to form lipid nanodomains.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa , Membrana Celular , Bacterias Gramnegativas , Membrana Dobles de Lípidos , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Glicina/metabolismo , Lipopolisacáridos/metabolismo , Lipoproteínas/química , Lipoproteínas/metabolismo , Fosfolípidos/metabolismo , Sitios de Unión , Proteostasis , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Proteoma/química , Proteoma/metabolismo , Regulón , Dominios Proteicos , Péptidos Antimicrobianos/metabolismo , Bacterias Gramnegativas/química , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/metabolismo
2.
Nature ; 623(7988): 814-819, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37938784

RESUMEN

Gram-negative bacteria are surrounded by two membranes. A special feature of the outer membrane is its asymmetry. It contains lipopolysaccharide (LPS) in the outer leaflet and phospholipids in the inner leaflet1-3. The proper assembly of LPS in the outer membrane is required for cell viability and provides Gram-negative bacteria intrinsic resistance to many classes of antibiotics. LPS biosynthesis is completed in the inner membrane, so the LPS must be extracted, moved across the aqueous periplasm that separates the two membranes and translocated through the outer membrane where it assembles on the cell surface4. LPS transport and assembly requires seven conserved and essential LPS transport components5 (LptA-G). This system has been proposed to form a continuous protein bridge that provides a path for LPS to reach the cell surface6,7, but this model has not been validated in living cells. Here, using single-molecule tracking, we show that Lpt protein dynamics are consistent with the bridge model. Half of the inner membrane Lpt proteins exist in a bridge state, and bridges persist for 5-10 s, showing that their organization is highly dynamic. LPS facilitates Lpt bridge formation, suggesting a mechanism by which the production of LPS can be directly coupled to its transport. Finally, the bridge decay kinetics suggest that there may be two different types of bridges, whose stability differs according to the presence (long-lived) or absence (short-lived) of LPS. Together, our data support a model in which LPS is both a substrate and a structural component of dynamic Lpt bridges that promote outer membrane assembly.


Asunto(s)
Membrana Externa Bacteriana , Proteínas Portadoras , Bacterias Gramnegativas , Lipopolisacáridos , Proteínas de la Membrana , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/metabolismo , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Transporte Biológico , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Escherichia coli/química , Escherichia coli/citología , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Bacterias Gramnegativas/química , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/metabolismo , Lipopolisacáridos/química , Lipopolisacáridos/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo
3.
J Biol Chem ; 299(9): 105146, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37562569

RESUMEN

The Maintenance of outer membrane (OM) Lipid Asymmetry system mediates retrograde phospholipid transport from the OM to the inner membrane (IM) in Gram-negative bacteria. However, the interactions between the various subunits of the IM and OM complexes are not well understood. In a recent study in 2023 by MacRae et al. in the Journal of Biological Chemistry, the authors examine components in the Maintenance of OM Lipid Asymmetry complex, define the interaction interfaces between members of the pathway, and propose a molecular model of the lipid transfer process from the OM to the IM that will help elucidate intricacies of lipid transport.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa , Membrana Externa Bacteriana , Bacterias Gramnegativas , Metabolismo de los Lípidos , Lípidos de la Membrana , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/metabolismo , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Transporte Biológico , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/metabolismo , Lípidos de la Membrana/metabolismo , Fosfolípidos/metabolismo
4.
Nature ; 613(7945): 729-734, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36450357

RESUMEN

Peptidoglycan and almost all surface glycopolymers in bacteria are built in the cytoplasm on the lipid carrier undecaprenyl phosphate (UndP)1-4. These UndP-linked precursors are transported across the membrane and polymerized or directly transferred to surface polymers, lipids or proteins. UndP is then flipped to regenerate the pool of cytoplasmic-facing UndP. The identity of the flippase that catalyses transport has remained unknown. Here, using the antibiotic amphomycin that targets UndP5-7, we identified two broadly conserved protein families that affect UndP recycling. One (UptA) is a member of the DedA superfamily8; the other (PopT) contains the domain DUF368. Genetic, cytological and syntenic analyses indicate that these proteins are UndP transporters. Notably, homologues from Gram-positive and Gram-negative bacteria promote UndP transport in Bacillus subtilis, indicating that recycling activity is broadly conserved among family members. Inhibitors of these flippases could potentiate the activity of antibiotics targeting the cell envelope.


Asunto(s)
Proteínas Bacterianas , Proteínas Portadoras , Secuencia Conservada , Evolución Molecular , Bacterias Gramnegativas , Bacterias Grampositivas , Fosfatos de Poliisoprenilo , Antibacterianos/farmacología , Bacillus subtilis/citología , Bacillus subtilis/efectos de los fármacos , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/clasificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/clasificación , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/genética , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/citología , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/genética , Bacterias Grampositivas/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Sintenía , Peptidoglicano/metabolismo , Pared Celular/química , Pared Celular/metabolismo
5.
Nature ; 613(7945): 721-728, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36450355

RESUMEN

The microbial cell wall is essential for maintenance of cell shape and resistance to external stressors1. The primary structural component of the cell wall is peptidoglycan, a glycopolymer with peptide crosslinks located outside of the cell membrane1. Peptidoglycan biosynthesis and structure are responsive to shifting environmental conditions such as pH and salinity2-6, but the mechanisms underlying such adaptations are incompletely understood. Precursors of peptidoglycan and other cell surface glycopolymers are synthesized in the cytoplasm and then delivered across the cell membrane bound to the recyclable lipid carrier undecaprenyl phosphate7 (C55-P, also known as UndP). Here we identify the DUF368-containing and DedA transmembrane protein families as candidate C55-P translocases, filling a critical gap in knowledge of the proteins required for the biogenesis of microbial cell surface polymers. Gram-negative and Gram-positive bacteria lacking their cognate DUF368-containing protein exhibited alkaline-dependent cell wall and viability defects, along with increased cell surface C55-P levels. pH-dependent synthetic genetic interactions between DUF368-containing proteins and DedA family members suggest that C55-P transporter usage is dynamic and modulated by environmental inputs. C55-P transporter activity was required by the cholera pathogen for growth and cell shape maintenance in the intestine. We propose that conditional transporter reliance provides resilience in lipid carrier recycling, bolstering microbial fitness both inside and outside the host.


Asunto(s)
Proteínas Bacterianas , Proteínas Portadoras , Aptitud Genética , Bacterias Gramnegativas , Bacterias Grampositivas , Fosfatos de Poliisoprenilo , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Pared Celular/química , Pared Celular/metabolismo , Lípidos/análisis , Peptidoglicano/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Bacterias Gramnegativas/química , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/química , Bacterias Grampositivas/citología , Bacterias Grampositivas/metabolismo , Viabilidad Microbiana
6.
Nature ; 606(7912): 160-164, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35585231

RESUMEN

Cellular iron homeostasis is vital and maintained through tight regulation of iron import, efflux, storage and detoxification1-3. The most common modes of iron storage use proteinaceous compartments, such as ferritins and related proteins4,5. Although lipid-bounded iron compartments have also been described, the basis for their formation and function remains unknown6,7. Here we focus on one such compartment, herein named the 'ferrosome', that was previously observed in the anaerobic bacterium Desulfovibrio magneticus6. Using a proteomic approach, we identify three ferrosome-associated (Fez) proteins that are responsible for forming ferrosomes in D. magneticus. Fez proteins are encoded in a putative operon and include FezB, a P1B-6-ATPase found in phylogenetically and metabolically diverse species of bacteria and archaea. We show that two other bacterial species, Rhodopseudomonas palustris and Shewanella putrefaciens, make ferrosomes through the action of their six-gene fez operon. Additionally, we find that fez operons are sufficient for ferrosome formation in foreign hosts. Using S. putrefaciens as a model, we show that ferrosomes probably have a role in the anaerobic adaptation to iron starvation. Overall, this work establishes ferrosomes as a new class of iron storage organelles and sets the stage for studying their formation and structure in diverse microorganisms.


Asunto(s)
Compuestos Férricos , Bacterias Gramnegativas , Familia de Multigenes , Orgánulos , Proteínas Bacterianas/genética , Desulfovibrio , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/genética , Orgánulos/genética , Orgánulos/metabolismo , Filogenia , Proteómica , Rhodopseudomonas , Shewanella putrefaciens
7.
Biotechnol Bioeng ; 119(1): 34-47, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34698385

RESUMEN

Outer membrane vesicles (OMVs) are nanoscale spherical vesicles released from Gram-negative bacteria. The lipid bilayer membrane structure of OMVs consists of similar components as bacterial membrane and thus has attracted more and more attention in exploiting OMVs' bio-applications. Although the endotoxic lipopolysaccharide on natural OMVs may impose potential limits on their clinical applications, genetic modification can reduce their endotoxicity and decorate OMVs with multiple functional proteins. These genetically engineered OMVs have been employed in various fields including vaccination, drug delivery, cancer therapy, bioimaging, biosensing, and enzyme carrier. This review will first briefly introduce the background of OMVs followed by recent advances in functionalization and various applications of engineered OMVs with an emphasis on the working principles and their performance, and then discuss about the future trends of OMVs in biomedical applications.


Asunto(s)
Membrana Externa Bacteriana , Sistemas de Liberación de Medicamentos , Vesículas Extracelulares , Bacterias Gramnegativas/citología , Vacunas , Animales , Ingeniería Genética , Humanos , Inmunoensayo , Ratones
8.
Nat Microbiol ; 6(7): 910-920, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34183815

RESUMEN

Bacterial species have diverse cell shapes that enable motility, colonization and virulence. The cell wall defines bacterial shape and is primarily built by two cytoskeleton-guided synthesis machines, the elongasome and the divisome. However, the mechanisms producing complex shapes, like the curved-rod shape of Vibrio cholerae, are incompletely defined. Previous studies have reported that species-specific regulation of cytoskeleton-guided machines enables formation of complex bacterial shapes such as cell curvature and cellular appendages. In contrast, we report that CrvA and CrvB are sufficient to induce complex cell shape autonomously of the cytoskeleton in V. cholerae. The autonomy of the CrvAB module also enables it to induce curvature in the Gram-negative species Escherichia coli, Pseudomonas aeruginosa, Caulobacter crescentus and Agrobacterium tumefaciens. Using inducible gene expression, quantitative microscopy and biochemistry, we show that CrvA and CrvB circumvent the need for patterning via cytoskeletal elements by regulating each other to form an asymmetrically localized, periplasmic structure that binds directly to the cell wall. The assembly and disassembly of this periplasmic structure enables dynamic changes in cell shape. Bioinformatics indicate that CrvA and CrvB may have diverged from a single ancestral hybrid protein. Using fusion experiments in V. cholerae, we find that a synthetic CrvA/B hybrid protein is sufficient to induce curvature on its own, but that expression of two distinct proteins, CrvA and CrvB, promotes more rapid curvature induction. We conclude that morphological complexity can arise independently of cell-shape specification by the core cytoskeleton-guided synthesis machines.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacterias Gramnegativas/citología , Proteínas Bacterianas/genética , Pared Celular/metabolismo , Citoesqueleto/metabolismo , Evolución Molecular , Bacterias Gramnegativas/crecimiento & desarrollo , Bacterias Gramnegativas/metabolismo , Peptidoglicano/metabolismo , Periplasma/metabolismo , Vibrio cholerae/citología , Vibrio cholerae/crecimiento & desarrollo , Vibrio cholerae/metabolismo
9.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33836615

RESUMEN

Gram-positive bacteria assemble a multilayered cell wall that provides tensile strength to the cell. The cell wall is composed of glycan strands cross-linked by nonribosomally synthesized peptide stems. Herein, we modify the peptide stems of the Gram-positive bacterium Bacillus subtilis with noncanonical electrophilic d-amino acids, which when in proximity to adjacent stem peptides form novel covalent 5,3-cross-links. Approximately 20% of canonical cell-wall cross-links can be replaced with synthetic cross-links. While a low level of synthetic cross-link formation does not affect B. subtilis growth and phenotype, at higher levels cell growth is perturbed and bacteria elongate. A comparison of the accumulation of synthetic cross-links over time in Gram-negative and Gram-positive bacteria highlights key differences between them. The ability to perturb cell-wall architecture with synthetic building blocks provides a novel approach to studying the adaptability, elasticity, and porosity of bacterial cell walls.


Asunto(s)
Pared Celular/química , Bacilos Grampositivos/química , Peptidoglicano/química , Aminoácidos/química , Aminoácidos/metabolismo , Bacillus subtilis/química , Bacillus subtilis/citología , Bacillus subtilis/crecimiento & desarrollo , Bacillus subtilis/metabolismo , Pared Celular/metabolismo , Bacterias Gramnegativas/química , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/metabolismo , Bacilos Grampositivos/citología , Bacilos Grampositivos/crecimiento & desarrollo , Bacilos Grampositivos/metabolismo , Peptidoglicano/metabolismo , Peptidil Transferasas/genética , Peptidil Transferasas/metabolismo , Fenotipo
10.
J Oleo Sci ; 70(4): 571-580, 2021 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-33692238

RESUMEN

Polyglycerol monolaurates are generally recognized as safe food additives and are commonly used as food emulsifiers. In this study, the antimicrobial effect of four polyglycerol monolaurates on two Gram-positive bacteria (Staphylococcus aureus and Bacillus subtilis) and two Gram-negative bacteria (Escherichia. coli and Pseudomonas aeruginosa) were investigated. The minimum inhibitory concentration (MIC) of diglycerol monolaurate (PG2ML), triglycerol monolaurate (PG3ML), hexaglycerol monolaurate (PG6ML), and decaglycerol monolaurate (PG10ML) against S. aureus was 0.16, 0.32, 0.63, and 1.25 mg/mL, respectively. The MIC of PG2ML, PG3ML, PG6ML, and PG10ML against B. subtilis was 0.32, 0.63, 1.25, and 3.75 mg/mL, respectively. No apparent antimicrobial effect of these four polyglycerol monolaurates on E. coli and P. aeruginosa was observed even up to 10.00 mg/mL. The underlying mechanism was investigated by assessing cell membrane permeability, the integrity of cell membrane, and morphology. We concluded that polyglycerol monolaurates might eliminate Gram-positive bacteria by disrupting the cell membrane, thereby increasing cell membrane permeability, releasing the cellular contents, and altering the cell morphology.


Asunto(s)
Antibacterianos , Emulsionantes , Aditivos Alimentarios , Glicerol/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Lauratos/farmacología , Polímeros/farmacología , Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Farmacorresistencia Bacteriana , Microbiología de Alimentos , Glicerol/química , Bacterias Gramnegativas/citología , Bacterias Grampositivas/citología , Lauratos/química , Pruebas de Sensibilidad Microbiana , Polímeros/química , Relación Estructura-Actividad
11.
Anal Chem ; 93(2): 843-850, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33301291

RESUMEN

Droplet microfluidics disrupted analytical biology with the introduction of digital polymerase chain reaction and single-cell sequencing. The same technology may also bring important innovation in the analysis of bacteria, including antibiotic susceptibility testing at the single-cell level. Still, despite promising demonstrations, the lack of a high-throughput label-free method of detecting bacteria in nanoliter droplets prohibits analysis of the most interesting strains and widespread use of droplet technologies in analytical microbiology. We use a sensitive and fast measurement of scattered light from nanoliter droplets to demonstrate reliable detection of the proliferation of encapsulated bacteria. We verify the sensitivity of the method by simultaneous readout of fluorescent signals from bacteria expressing fluorescent proteins and demonstrate label-free readout on unlabeled Gram-negative and Gram-positive species. Our approach requires neither genetic modification of the cells nor the addition of chemical markers of metabolism. It is compatible with a wide range of bacterial species of clinical, research, and industrial interest, opening the microfluidic droplet technologies for adaptation in these fields.


Asunto(s)
Bacterias Gramnegativas/aislamiento & purificación , Bacterias Grampositivas/aislamiento & purificación , Ensayos Analíticos de Alto Rendimiento , Técnicas Analíticas Microfluídicas , Nanopartículas/química , Análisis de la Célula Individual , Bacterias Gramnegativas/citología , Bacterias Grampositivas/citología , Tamaño de la Partícula , Propiedades de Superficie
12.
PLoS Comput Biol ; 16(10): e1008355, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33112853

RESUMEN

In Gram-negative bacteria, the folding and insertion of ß-barrel outer membrane proteins (OMPs) to the outer membrane are mediated by the ß-barrel assembly machinery (BAM) complex. Two leading models of this process have been put forth: the hybrid barrel model, which claims that a lateral gate in BamA's ß-barrel can serve as a template for incoming OMPs, and the passive model, which claims that a thinned membrane near the lateral gate of BamA accelerates spontaneous OMP insertion. To examine the key elements of these two models, we have carried out 45.5 µs of equilibrium molecular dynamics simulations of BamA with and without POTRA domains from Escherichia coli, Salmonella enterica, Haemophilus ducreyi and Neisseria gonorrhoeae, together with BamA's homolog, TamA from E. coli, in their native, species-specific outer membranes. In these equilibrium simulations, we consistently observe membrane thinning near the lateral gate for all proteins. We also see occasional spontaneous lateral gate opening and sliding of the ß-strands at the gate interface for N. gonorrhoeae, indicating that the gate is dynamic. An additional 14 µs of free-energy calculations shows that the energy necessary to open the lateral gate in BamA/TamA varies by species, but is always lower than the Omp85 homolog, FhaC. Our combined results suggest OMP insertion utilizes aspects of both the hybrid barrel and passive models.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa , Membrana Celular , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Bacterias Gramnegativas/química , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/metabolismo , Simulación de Dinámica Molecular , Conformación Proteica en Lámina beta , Pliegue de Proteína
13.
Mikrochim Acta ; 187(10): 558, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32914337

RESUMEN

A single-tube method based on a dual-electrostatic interaction (EI) strategy for bacteria capture and DNA extraction was designed to enable the highly sensitive detection of nucleic acids. Specially designed magnetic nanoparticles were developed to meet the opposing requirements of a single-tube method, which exist between the strong EI required for efficient bacteria capture and the weak EI required for DNA extraction with minimal DNA adsorption. A dual-EI strategy for the single-tube (DESIGN) method was thus developed to integrate bacteria enrichment, bacteria cell lysis, and DNA recovery in a single tube, thereby minimizing precious sample loss and reducing handling time. Subsequently, we evaluated the performance with a variety of concentrations from 5 to 100 colony-forming units (CFU)/10 mL human urine and milk samples. The DESIGN method achieved the simple and sensitive detection of Salmonella enterica serovar Typhimurium in 10 mL of human urine and milk samples up to 5 CFU by quantitative PCR. Furthermore, the DESIGN method detected Brucella ovis and Escherichia coli from 10 mL of human urine with a detection limit up to 5 CFU/10 mL. Graphical abstract.


Asunto(s)
Bacterias Gramnegativas/metabolismo , Nanopartículas/química , Bacterias Gramnegativas/citología , Humanos , Electricidad Estática
14.
Adv Biosyst ; 4(9): e2000074, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32803868

RESUMEN

Nontoxic carbon nanoparticle samples prepared by both bottom-up and top-down approaches do not inhibit Gram-negative bacterial growth, indicating excellent biocompatibilities. However, cell growth inhibitory efficacies increase considerably when the carbon nanoparticles are conjugated with the antibiotic tetracycline. In tetracycline-resistant bacteria, these efficacies can approach tenfold higher activities when compared to tetracycline alone. No structural abnormality such as membrane disruptions is evident in the tested bacterial strains; this is in contrast with other nanocarbon systems such as graphene oxides, carbon nanotubes, and amine-functionalized carbon nanoparticles which do exhibit membrane disruptions. In comparison, the tetracycline-conjugated carbon nanoparticles induce membrane perturbations (but not membrane disruptions), inhibiting bacterial efflux mechanisms. It is proposed that when tetracycline is conjugated to the surface of carbon nanoparticles, it functions to direct the nanoparticles to membrane-associated tetracycline efflux pumps, thereby blocking and subsequently inhibiting their function. The conjugation between biocompatible carbon nanoparticles and subtherapeutic but well-established antibiotic molecules may provide hybrid antibiotic assembly strategies resulting in effective multidrug efflux inhibition for combating antibiotic resistance.


Asunto(s)
Antibacterianos , Farmacorresistencia Bacteriana/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Nanotubos de Carbono/química , Tetraciclina , Antibacterianos/química , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Tetraciclina/química , Tetraciclina/farmacología
15.
J Chem Theory Comput ; 16(8): 5369-5384, 2020 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-32628849

RESUMEN

The bacterial cell envelope of Gram-negative bacteria is a complex biological barrier with multiple layers consisting of the inner membrane, periplasm of peptidoglycan, and the outer membrane with lipopolysaccharides (LPS). With rising antimicrobial resistance there is increasing interest in understanding interactions of small molecules with the cell membrane to aid in the development of novel drug molecules. Hence suitable representations of the bacterial membrane are required to carry out meaningful molecular dynamics simulations. Given the complexity of the cell envelope, fully atomistic descriptions of the cell membrane with explicit solvent are computationally prohibitive, allowing limited sampling with small system sizes. However, coarse-grained (CG) models such as MARTINI allow one to study phenomena at physiologically relevant length and time scales. Although MARTINI models for lipids and the LPS are available in literature, a suitable CG model of peptidoglycan is lacking. Using an all-atom model described by Gumbart et al. [PLoS Comput. Biol. 2014, 10, e1003475], we develop a CG model of the peptidoglycan network within the MARTINI framework. The model is parametrized to reproduce the end-to-end distance of glycan strands. The structural properties such as the equilibrium angle between adjacent peptides along the strands, area per disaccharide, and cavity size distributions agree well with the atomistic simulation results. Mechanical properties such as the area compressibility and the bending modulus are accurately reproduced. While developing novel antibiotics it is important to assess barrier properties of the peptidogylcan network. We evaluate and compare the free energy of insertion for a thymol molecule using umbrella sampling on both the MARTINI and all-atom peptidoglycan models. The insertion free energy was found to be less than kBT for both the MARTINI and all-atom models. Additional restraint free simulations reveal rapid translocation of thymol across peptidogylcan. We expect that the proposed MARTINI model for peptidoglycan will be useful in understanding phenomena associated with bacterial cell walls at larger length and time scales, thereby overcoming the current limitations of all-atom models.


Asunto(s)
Pared Celular/química , Bacterias Gramnegativas/química , Lipopolisacáridos/metabolismo , Modelos Biológicos , Peptidoglicano/metabolismo , Termodinámica , Pared Celular/metabolismo , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/metabolismo , Lipopolisacáridos/química , Conformación Molecular , Simulación de Dinámica Molecular , Peptidoglicano/química
16.
Biointerphases ; 15(3): 031007, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32456440

RESUMEN

Antimicrobial peptides (AMPs) are attractive as biomaterial coatings because they have broad spectrum activity against different microbes, with a low likelihood of incurring antimicrobial resistance. Direct action against the bacterial membrane is the most common mechanism of action (MOA) of AMPs, with specific MOAs dependent on membrane composition, peptide concentration, and environmental factors that include temperature. Chrysophsin-1 (CHY1) is a broad spectrum salt-tolerant AMP that is derived from a marine fish. A cysteine modification was made to the peptide to facilitate attachment to a surface, such as a biomedical device. The authors used quartz crystal microbalance with dissipation monitoring to study how temperature (23 and 37 °C) and lipid composition influence the MOA of cysteine-modified peptide (C-CHY1) with model membranes comprised of supported lipid bilayers (SLBs). These two temperatures were used so that the authors could better understand the differences in behavior between typical lab temperatures and physiologic conditions. The authors created model membranes that mimicked properties of Gram-negative and Gram-positive bacteria in order to understand how the mechanisms might differ for different types of bacterial systems. SLB models of Gram-positive bacterial membranes were formed using combinations of phosphatidylcholine, phosphatidylglycerol (PG), and S. aureus-derived lipoteichoic acid (LTA). SLB models of Gram-negative bacterial membranes were formed using combinations of phosphatidylethanolamine (PE), PG, and E. coli-derived lipopolysaccharides (LPS). The molecules that distinguish Gram-positive and Gram-negative membranes (LTA and LPS) have the potential to alter the MOA of C-CHY1 with the SLBs. The authors' results showed that the MOA for the Gram-positive SLBs was not sensitive to temperature, but the LTA addition did have an effect. Specifically, similar trends in frequency and dissipation changes across all overtones were observed, and the same mechanistic trends were observed in the polar plots at 23 and 37 °C. However, when LTA was added, polar plots showed an association between C-CHY1 and LTA, leading to SLB saturation. This was demonstrated by significant changes in dissipation, while the frequency (mass) was not increasing after the saturation point. For the Gram-negative SLBs, the composition did not have a significant effect on MOA, but the authors saw more differences between the two temperatures studied. The authors believe this is due to the fact that the gel-liquid crystal transition temperature of PE is 25 °C, which means that the bilayer is more rigid at 23 °C, compared to temperatures above the transition point. At 23 °C, a significant energetic shift would be required to allow for additional AMP insertion. This could be seen in the polar plots, where there was a steep slope but there was very little mass addition. At 37 °C, the membrane is more fluid and there is less of an energetic requirement for insertion. Therefore, the authors observed greater mass addition and fewer changes in dissipation. A better understanding of C-CHY1 MOA using different SLB models will allow for the more rational design of future therapeutic solutions that make use of antimicrobial peptides, including those involving biomaterial coatings.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Membrana Celular/metabolismo , Bacterias Gramnegativas/citología , Bacterias Grampositivas/citología , Lipopolisacáridos/farmacología , Ácidos Teicoicos/farmacología , Membrana Celular/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Membrana Dobles de Lípidos/química , Péptidos/química , Temperatura
17.
Anal Chem ; 92(11): 7523-7531, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32330016

RESUMEN

In diagnostics of infectious diseases, matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry (MALDI-TOF MS) can be applied for the identification of pathogenic microorganisms. However, to achieve a trustworthy identification from MALDI-TOF MS data, a significant amount of biomass should be considered. The bacterial load that potentially occurs in a sample is therefore routinely amplified by culturing, which is a time-consuming procedure. In this paper, we show that culturing can be avoided by conducting MALDI-TOF MS on individual bacterial cells. This results in a more rapid identification of species with an acceptable accuracy. We propose a deep learning architecture to analyze the data and compare its performance with traditional supervised machine learning algorithms. We illustrate our workflow on a large data set that contains bacterial species related to urinary tract infections. Overall we obtain accuracies up to 85% in discriminating five different species.


Asunto(s)
Aprendizaje Profundo , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/patogenicidad , Bacterias Grampositivas/citología , Bacterias Grampositivas/patogenicidad , Análisis de la Célula Individual , Aerosoles/química , Bacterias Gramnegativas/aislamiento & purificación , Bacterias Grampositivas/aislamiento & purificación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
18.
Angew Chem Int Ed Engl ; 59(22): 8517-8521, 2020 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-32023354

RESUMEN

Multi-drug resistance in Gram-negative bacteria is often associated with low permeability of the outer membrane. To investigate the role of membrane channels in the uptake of antibiotics, we present an approach using fusion of native outer membrane vesicles (OMVs) into a planar lipid bilayer, allowing characterization of membrane protein channels in their native environment. Two major membrane channels from E. coli, OmpF and OmpC, were overexpressed from the host and the corresponding OMVs were collected. Each OMV fusion surprisingly revealed only single or few channel activities. The asymmetry of the OMVs translates after fusion into the lipid membrane with the lipopolysaccharides (LPS) dominantly present at the side of OMV addition. Compared to the conventional reconstitution method, the channels fused from OMVs containing LPS have similar conductance but a much broader distribution and significantly lower permeation. We suggest using outer membrane vesicles for functional and structural studies of membrane channels in the native membrane.


Asunto(s)
Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Fenómenos Electrofisiológicos/efectos de los fármacos , Bacterias Gramnegativas/citología , Bacterias Gramnegativas/fisiología , Lipopolisacáridos/farmacología , Transporte Biológico/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/metabolismo , Porinas/genética , Porinas/metabolismo
19.
Pol J Microbiol ; 69(4): 503-508, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33574878

RESUMEN

In this work, an exploratory study was conducted to examine Gram staining based on the capillary tube. Each Gram staining step for all bacterial strains tested was completed in capillary tubes. The results showed that different Gram staining morphologies were clearly visible in the capillary tubes. The results presented here demonstrated that the improved method could effectively distinguish between Gram-positive and Gram-negative bacteria, and only small volumes of reagents were required in this method. Collectively, this efficient method could rapidly and accurately identify the types of bacteria. Therefore, our findings could be used as a useful reference study for other staining methods.


Asunto(s)
Técnicas Bacteriológicas , Violeta de Genciana , Bacterias Gramnegativas/citología , Bacterias Grampositivas/citología , Fenazinas , Coloración y Etiquetado/métodos , Técnicas Bacteriológicas/instrumentación , Técnicas Bacteriológicas/métodos , Coloración y Etiquetado/instrumentación
20.
Protein Sci ; 29(3): 629-646, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31747090

RESUMEN

The history of modern medicine cannot be written apart from the history of the antibiotics. Antibiotics are cytotoxic secondary metabolites that are isolated from Nature. The antibacterial antibiotics disproportionately target bacterial protein structure that is distinct from eukaryotic protein structure, notably within the ribosome and within the pathways for bacterial cell-wall biosynthesis (for which there is not a eukaryotic counterpart). This review focuses on a pre-eminent class of antibiotics-the ß-lactams, exemplified by the penicillins and cephalosporins-from the perspective of the evolving mechanisms for bacterial resistance. The mechanism of action of the ß-lactams is bacterial cell-wall destruction. In the monoderm (single membrane, Gram-positive staining) pathogen Staphylococcus aureus the dominant resistance mechanism is expression of a ß-lactam-unreactive transpeptidase enzyme that functions in cell-wall construction. In the diderm (dual membrane, Gram-negative staining) pathogen Pseudomonas aeruginosa a dominant resistance mechanism (among several) is expression of a hydrolytic enzyme that destroys the critical ß-lactam ring of the antibiotic. The key sensing mechanism used by P. aeruginosa is monitoring the molecular difference between cell-wall construction and cell-wall deconstruction. In both bacteria, the resistance pathways are manifested only when the bacteria detect the presence of ß-lactams. This review summarizes how the ß-lactams are sensed and how the resistance mechanisms are manifested, with the expectation that preventing these processes will be critical to future chemotherapeutic control of multidrug resistant bacteria.


Asunto(s)
Antibacterianos/farmacología , Pared Celular/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , beta-Lactamas/farmacología , Antibacterianos/química , Farmacorresistencia Bacteriana/efectos de los fármacos , Bacterias Gramnegativas/citología , Bacterias Grampositivas/citología , Pruebas de Sensibilidad Microbiana , beta-Lactamas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA