Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
Nature ; 613(7945): 729-734, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36450357

RESUMO

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.


Assuntos
Proteínas de Bactérias , Proteínas de Transporte , Sequência Conservada , Evolução Molecular , Bactérias Gram-Negativas , Bactérias Gram-Positivas , Fosfatos de Poli-Isoprenil , Antibacterianos/farmacologia , Bacillus subtilis/citologia , Bacillus subtilis/efeitos dos fármacos , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/classificação , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/classificação , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Bactérias Gram-Negativas/citologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/genética , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Positivas/citologia , Bactérias Gram-Positivas/efeitos dos fármacos , Bactérias Gram-Positivas/genética , Bactérias Gram-Positivas/metabolismo , Fosfatos de Poli-Isoprenil/metabolismo , Sintenia , Peptidoglicano/metabolismo , Parede Celular/química , Parede Celular/metabolismo
2.
Nature ; 613(7945): 721-728, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36450355

RESUMO

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.


Assuntos
Proteínas de Bactérias , Proteínas de Transporte , Aptidão Genética , Bactérias Gram-Negativas , Bactérias Gram-Positivas , Fosfatos de Poli-Isoprenil , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Parede Celular/química , Parede Celular/metabolismo , Lipídeos/análise , Peptidoglicano/metabolismo , Fosfatos de Poli-Isoprenil/metabolismo , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/citologia , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Positivas/química , Bactérias Gram-Positivas/citologia , Bactérias Gram-Positivas/metabolismo , Viabilidade Microbiana
3.
J Biol Chem ; 298(10): 102488, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36113580

RESUMO

Rhamnose-rich cell wall polysaccharides (Rha-CWPSs) have emerged as crucial cell wall components of numerous Gram-positive, ovoid-shaped bacteria-including streptococci, enterococci, and lactococci-of which many are of clinical or biotechnological importance. Rha-CWPS are composed of a conserved polyrhamnose backbone with side-chain substituents of variable size and structure. Because these substituents contain phosphate groups, Rha-CWPS can also be classified as polyanionic glycopolymers, similar to wall teichoic acids, of which they appear to be functional homologs. Recent advances have highlighted the critical role of these side-chain substituents in bacterial cell growth and division, as well as in specific interactions between bacteria and infecting bacteriophages or eukaryotic hosts. Here, we review the current state of knowledge on the structure and biosynthesis of Rha-CWPS in several ovoid-shaped bacterial species. We emphasize the role played by multicomponent transmembrane glycosylation systems in the addition of side-chain substituents of various sizes as extracytoplasmic modifications of the polyrhamnose backbone. We provide an overview of the contribution of Rha-CWPS to cell wall architecture and biogenesis and discuss current hypotheses regarding their importance in the cell division process. Finally, we sum up the critical roles that Rha-CWPS can play as bacteriophage receptors or in escaping host defenses, roles that are mediated mainly through their side-chain substituents. From an applied perspective, increased knowledge of Rha-CWPS can lead to advancements in strategies for preventing phage infection of lactococci and streptococci in food fermentation and for combating pathogenic streptococci and enterococci.


Assuntos
Bacteriófagos , Parede Celular , Bactérias Gram-Positivas , Parede Celular/química , Bactérias Gram-Positivas/química , Bactérias Gram-Positivas/citologia , Polissacarídeos/química , Ramnose , Ácidos Teicoicos/química , Divisão Celular/fisiologia
4.
J Mol Biol ; 433(9): 166911, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33676927

RESUMO

Fluoroquinolones (FQ) are antibiotics widely used in clinical practise, but the development of bacterial resistance to these drugs is currently a critical public health problem. In this context, ternary copper complexes of FQ (CuFQPhen) have been studied as a potential alternative. In this study, we compared the passive diffusion across the lipid bilayer of one of the most used FQ, ciprofloxacin (Cpx), and its ternary copper complex, CuCpxPhen, that has shown previous promising results regarding antibacterial activity and membrane partition. A combination of spectroscopic studies and molecular dynamics simulations were used and two different model membranes tested: one composed of anionic phospholipids, and the other composed of zwitterionic phospholipids. The obtained results showed a significantly higher membrane permeabilization activity, larger partition, and a more favourable free energy landscape for the permeation of CuCpxPhen across the membrane, when compared to Cpx. Furthermore, the computational results indicated a more favourable translocation of CuCpxPhen across the anionic membrane, when compared to the zwitterionic one, suggesting a higher specificity towards the former. These findings are important to decipher the influx mechanism of CuFQPhen in bacterial cells, which is crucial for the ultimate use of CuFQPhen complexes as an alternative to FQ to tackle multidrug-resistant bacteria.


Assuntos
Antibacterianos/química , Antibacterianos/metabolismo , Membrana Celular/metabolismo , Ciprofloxacina/química , Ciprofloxacina/metabolismo , Cobre/metabolismo , Difusão , Bactérias Gram-Positivas , Cardiolipinas/metabolismo , Membrana Celular/química , Cobre/química , Farmacorresistência Bacteriana Múltipla , Bactérias Gram-Positivas/química , Bactérias Gram-Positivas/citologia , Bactérias Gram-Positivas/metabolismo , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Simulação de Dinâmica Molecular , Fosfatidilgliceróis/metabolismo , Prótons , Termodinâmica
5.
Nat Commun ; 12(1): 1709, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33731718

RESUMO

Long-distance extracellular electron transfer has been observed in Gram-negative bacteria and plays roles in both natural and engineering processes. The electron transfer can be mediated by conductive protein appendages (in short unicellular bacteria such as Geobacter species) or by conductive cell envelopes (in filamentous multicellular cable bacteria). Here we show that Lysinibacillus varians GY32, a filamentous unicellular Gram-positive bacterium, is capable of bidirectional extracellular electron transfer. In microbial fuel cells, L. varians can form centimetre-range conductive cellular networks and, when grown on graphite electrodes, the cells can reach a remarkable length of 1.08 mm. Atomic force microscopy and microelectrode analyses suggest that the conductivity is linked to pili-like protein appendages. Our results show that long-distance electron transfer is not limited to Gram-negative bacteria.


Assuntos
Transporte de Elétrons/fisiologia , Bactérias Gram-Positivas/metabolismo , Bacillaceae/citologia , Bacillaceae/crescimento & desenvolvimento , Bacillaceae/metabolismo , Fontes de Energia Bioelétrica/microbiologia , Condutividade Elétrica , Eletrodos/microbiologia , Fímbrias Bacterianas/metabolismo , Fímbrias Bacterianas/ultraestrutura , Bactérias Gram-Positivas/citologia , Bactérias Gram-Positivas/crescimento & desenvolvimento , Grafite , Microscopia de Força Atômica , Nanofios
6.
J Oleo Sci ; 70(4): 571-580, 2021 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-33692238

RESUMO

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.


Assuntos
Antibacterianos , Emulsificantes , Aditivos Alimentares , Glicerol/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Lauratos/farmacologia , Polímeros/farmacologia , Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Farmacorresistência Bacteriana , Microbiologia de Alimentos , Glicerol/química , Bactérias Gram-Negativas/citologia , Bactérias Gram-Positivas/citologia , Lauratos/química , Testes de Sensibilidade Microbiana , Polímeros/química , Relação Estrutura-Atividade
7.
ACS Appl Bio Mater ; 4(3): 2104-2112, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35014338

RESUMO

Bacterial infections are a global healthcare problem, resulting in serious clinical morbidities and mortality. Real-time monitoring of live bacteria by fluorescent imaging technology has potential in diagnosis of bacterial infections, elucidating antimicrobial agents' mode of action, assessing drug toxicity, and examining bacterial antimicrobial resistance. In this work, a naphthalimide-derived fluorescent probe ZTRS-BP was developed for wash-free Gram-positive bacteria imaging. The probe aggregated in aqueous solutions and exhibited aggregation-caused fluorescence quenching (ACQ). The interaction with Gram-positive bacteria cell walls would selectively disaggregate the probe and the liberated probes were dispersed on the outside of the bacteria cell walls to achieve surface fluorescence imaging. There were no such interactions with Gram-negative bacteria, which indicates that selective binding and imaging of Gram-positive bacteria was achieved. The binding of zinc ions by ZTRS-BP can enhance the fluorescent signals on the bacterial surface by inhibiting the process of photoinduced electron transfer. ZTRS-BP-Zn(II) complex was an excellent dye to discriminate mixed Gram-positive and Gram-negative bacteria. Also, live and dead bacteria can be differentially imaged by ZTRS-BP-Zn(II). Furthermore, ZTRS-BP-Zn(II) was used for real-time monitoring bacteria viability such as B. cereus treated with antibiotic vancomycin.


Assuntos
Materiais Biocompatíveis/química , Membrana Celular/química , Corantes Fluorescentes/química , Bactérias Gram-Positivas/isolamento & purificação , Bactérias Gram-Positivas/citologia , Teste de Materiais , Viabilidade Microbiana , Estrutura Molecular , Imagem Óptica , Tamanho da Partícula , Fatores de Tempo
8.
Anal Chem ; 93(2): 843-850, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33301291

RESUMO

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.


Assuntos
Bactérias Gram-Negativas/isolamento & purificação , Bactérias Gram-Positivas/isolamento & purificação , Ensaios de Triagem em Larga Escala , Técnicas Analíticas Microfluídicas , Nanopartículas/química , Análise de Célula Única , Bactérias Gram-Negativas/citologia , Bactérias Gram-Positivas/citologia , Tamanho da Partícula , Propriedades de Superfície
9.
Biointerphases ; 15(3): 031007, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32456440

RESUMO

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.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Membrana Celular/metabolismo , Bactérias Gram-Negativas/citologia , Bactérias Gram-Positivas/citologia , Lipopolissacarídeos/farmacologia , Ácidos Teicoicos/farmacologia , Membrana Celular/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Bicamadas Lipídicas/química , Peptídeos/química , Temperatura
10.
Anal Chem ; 92(11): 7523-7531, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32330016

RESUMO

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.


Assuntos
Aprendizado Profundo , Bactérias Gram-Negativas/citologia , Bactérias Gram-Negativas/patogenicidade , Bactérias Gram-Positivas/citologia , Bactérias Gram-Positivas/patogenicidade , Análise de Célula Única , Aerossóis/química , Bactérias Gram-Negativas/isolamento & purificação , Bactérias Gram-Positivas/isolamento & purificação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
11.
Pol J Microbiol ; 69(4): 503-508, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33574878

RESUMO

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.


Assuntos
Técnicas Bacteriológicas , Violeta Genciana , Bactérias Gram-Negativas/citologia , Bactérias Gram-Positivas/citologia , Fenazinas , Coloração e Rotulagem/métodos , Técnicas Bacteriológicas/instrumentação , Técnicas Bacteriológicas/métodos , Coloração e Rotulagem/instrumentação
12.
Protein Sci ; 29(3): 629-646, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31747090

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Parede Celular/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , beta-Lactamas/farmacologia , Antibacterianos/química , Farmacorresistência Bacteriana/efeitos dos fármacos , Bactérias Gram-Negativas/citologia , Bactérias Gram-Positivas/citologia , Testes de Sensibilidade Microbiana , beta-Lactamas/química
13.
Phys Chem Chem Phys ; 21(23): 12530-12539, 2019 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-31147666

RESUMO

The emergence of antibiotic-resistance is a major concern to global human health and identification of novel antibiotics is critical to mitigate the threat. Mutacin 1140 (MU1140) is a promising antimicrobial lanthipeptide and is effective against Gram-positive bacteria. Like nisin, MU1140 targets and sequesters lipid II and interferes with its function, which results in the inhibition of bacterial cell wall synthesis, and leads to bacteria cell lysis. MU1140 contains a structurally similar thioether cage for binding the lipid II pyrophosphate as for nisin. In addition to lipid II binding, nisin is known to form membrane pores. Membrane pore formation and membrane disruption is a common mode of action for many antimicrobial peptides, including gallidermin, a lantibiotic peptide with similar structural features as MU1140. However, whether and how MU1140 and its variants can form permeable membrane pores remains to be demonstrated. In this work, we explored the potential mechanisms of membrane pore formation by performing molecular simulations of the MU1140-lipid II complex in the bacterial membrane. Our results suggest that MU1140-lipid II complexes are able to form water permeating membrane pores. We find that a single chain of MU1140 complexed with lipid II in the transmembrane region can permeate water molecules across the membrane via a single-file water transport mechanism. The ordering of the water molecules in the single-file chain region as well as the diffusion behavior is similar to those observed in other biological water channels. Multiple complexes of MU1140-lipid II in the membrane showed enhanced permeability for the water molecules, as well as a noticeable membrane distortion and lipid relocation, suggesting that a higher concentration of MU1140 assembly in the membrane can cause significant disruption of the bacterial membrane. These investigations provide an atomistic level insight into a novel mode of action for MU1140 that can be exploited to develop optimized peptide variants with improved antimicrobial properties.


Assuntos
Bacteriocinas/farmacologia , Bactérias Gram-Positivas/efeitos dos fármacos , Simulação de Dinâmica Molecular , Peptídeos/farmacologia , Bacteriocinas/química , Membrana Celular/efeitos dos fármacos , Bactérias Gram-Positivas/citologia , Lipídeos/química , Lipídeos/farmacologia , Testes de Sensibilidade Microbiana , Peptídeos/química , Água/química
14.
Colloids Surf B Biointerfaces ; 181: 6-15, 2019 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31103799

RESUMO

The antibacterial nature of graphene oxide (GO) has stimulated wide interest in the medical field. Although the antibacterial activity of GO towards bacteria has been well studied, a deeper understanding of the mechanism of action of GO is still lacking. The objective of the study was to elucidate the difference in the interactions of GO towards Gram-positive and Gram-negative bacteria. The synthesized GO was characterized by Ultraviolet-visible spectroscopy (UV-vis), Raman and Attenuated Total Reflectance-Fourier-transform infrared spectroscopy (ATR-FTIR). Viability, time-kill and Lactose Dehydrogenase (LDH) release assays were carried out along with FESEM, TEM and ATR-FTIR analysis of GO treated bacterial cells. Characterizations of synthesized GO confirmed the transition of graphene to GO and the antibacterial activity of GO was concentration and time-dependent. Loss of membrane integrity in bacteria was enhanced with increasing GO concentrations and this corresponded to the elevated release of LDH in the reaction medium. Surface morphology of GO treated bacterial culture showed apparent differences in the mechanism of action of GO towards Gram-positive and Gram-negative bacteria where cell entrapment was mainly observed for Gram-positive Staphylococcus aureus and Enterococcus faecalis whereas membrane disruption due to physical contact was noted for Gram-negative Escherichia coli and Pseudomonas aeruginosa. ATR-FTIR characterizations of the GO treated bacterial cells showed changes in the fatty acids, amide I and amide II of proteins, peptides and amino acid regions compared to untreated bacterial cells. Therefore, the data generated further enhance our understanding of the antibacterial activity of GO towards bacteria.


Assuntos
Antibacterianos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Grafite/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Bactérias Gram-Negativas/citologia , Bactérias Gram-Positivas/citologia , Grafite/síntese química , Grafite/química , Testes de Sensibilidade Microbiana , Tamanho da Partícula , Relação Estrutura-Atividade , Propriedades de Superfície
15.
Biochem Biophys Res Commun ; 514(2): 497-502, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31056261

RESUMO

Cationic antimicrobial peptides (CAMPs) are important antibiotics because they possess a broad spectrum of activity against both Gram-positive and Gram-negative bacteria, including those resistant to traditional antibiotics. The cyclic peptide bactenecin is a 12-amino acid CAMP that contains one intramolecular disulfide bond. To improve the antibacterial activity of bactenecin, we designed and synthesized several bactenecin analogs by applying multiple approaches, including amino acid substitution, use of the d-enantiomeric form, and lipidation. Among the synthetic analogs, d-enantiomeric bactenecin conjugated to capric acid, which we named dBacK-(cap), exhibited a significantly enhanced antibacterial spectrum with MIC values ranging from 1 to 8 µM against both Gram-positive and Gram-negative bacteria, including some drug-resistant bacteria. Upon exposure to dBacK-(cap), S. aureus cells were killed within 1 h at the MIC value, but full inactivation of E. coli required over 2 h. These results indicate that covalent addition of a d-amino acid and a fatty acid to bactenecin is the most effective approach for enhancing its antibacterial activity.


Assuntos
Antibacterianos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Antibacterianos/síntese química , Antibacterianos/química , Permeabilidade da Membrana Celular , Desenho de Fármacos , Bactérias Gram-Negativas/citologia , Bactérias Gram-Negativas/ultraestrutura , Bactérias Gram-Positivas/citologia , Bactérias Gram-Positivas/ultraestrutura , Cinética , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/química
16.
Biosens Bioelectron ; 136: 23-30, 2019 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-31029006

RESUMO

In this study, we report a facile, reusable, and highly sensitive label-free impedance sensor for discriminating Gram-positive and Gram-negative bacteria. The impedance sensor was fabricated using gold interdigitated electrodes onto a tungsten oxide thin film. X-Ray diffraction confirmed the formation of polycrystalline tungsten oxide. Field emission scanning electron microscopy and atomic force microscopy revealed that tungsten oxide has a porous structure. Tungsten oxide was functionalized with vancomycin, a glycopeptide antibiotic known to have a specific interaction with the peptidoglycan layer of Gram-positive bacteria. Fourier transform infrared microscopy and scanning electron microscopy were employed to test the morphological coating of vancomycin on interdigitated electrodes/tungsten oxide sensor. The functionalized tungsten oxide sensor was highly efficient in the capture of Gram-positive bacteria. The impedance measurement was also sensitive to differentiate between viable and non-viable Gram-positive bacteria. Limit of detection 102 colony forming unit/ml, linear dynamic range 102-107 colony forming unit/ml under physiological conditions and reusable nature of this vancomycin coated impedance sensor provide a label-free strategy for quick, sensitive and highly selective detection of Gram-positive bacteria.


Assuntos
Antibacterianos/farmacologia , Bactérias Gram-Positivas/isolamento & purificação , Vancomicina/farmacologia , Impedância Elétrica , Eletrodos , Bactérias Gram-Negativas/classificação , Bactérias Gram-Positivas/classificação , Bactérias Gram-Positivas/citologia , Bactérias Gram-Positivas/efeitos dos fármacos , Viabilidade Microbiana , Microscopia de Força Atômica , Óxidos , Sensibilidade e Especificidade , Espectroscopia de Infravermelho com Transformada de Fourier , Tungstênio
17.
J Colloid Interface Sci ; 546: 192-210, 2019 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-30921674

RESUMO

The attachment of single-celled organisms, namely bacteria and fungi, to abiotic surfaces is of great interest to both the scientific and medical communities. This is because the interaction of such cells has important implications in a range of areas, including biofilm formation, biofouling, antimicrobial surface technologies, and bio-nanotechnologies, as well as infection development, control, and mitigation. While central to many biological phenomena, the factors which govern microbial surface attachment are still not fully understood. This lack of understanding is a direct consequence of the complex nature of cell-surface interactions, which can involve both specific and non-specific interactions. For applications involving micro- and nano-structured surfaces, developing an understanding of such phenomenon is further complicated by the diverse nature of surface architectures, surface chemistry, variation in cellular physiology, and the intended technological output. These factors are extremely important to understand in the emerging field of antibacterial nanostructured surfaces. The aim of this perspective is to re-frame the discussion surrounding the mechanism of nanostructured-microbial surface interactions. Broadly, the article reviews our current understanding of these phenomena, while highlighting the knowledge gaps surrounding the adhesive forces which govern bacterial-nanostructure interactions and the role of cell membrane rigidity in modulating surface activity. The roles of surface charge, cell rigidity, and cell-surface adhesion force in bacterial-surface adsorption are discussed in detail. Presently, most studies have overlooked these areas, which has left many questions unanswered. Further, this perspective article highlights the numerous experimental issues and misinterpretations which surround current studies of antibacterial nanostructured surfaces.


Assuntos
Antibacterianos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Nanoestruturas/química , Antibacterianos/química , Aderência Bacteriana/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Elasticidade/efeitos dos fármacos , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/citologia , Bactérias Gram-Positivas/química , Bactérias Gram-Positivas/citologia , Tamanho da Partícula , Propriedades de Superfície
18.
Langmuir ; 35(16): 5557-5567, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30888181

RESUMO

Small-molecule cationic amphiphiles (CAms) were designed to combat the rapid rise in drug-resistant bacteria. CAms were designed to target and compromise the structural integrity of bacteria membranes, leading to cell rupture and death. Discrete structural features of CAms were varied, and structure-activity relationship studies were performed to guide the rational design of potent antimicrobials with desirable selectivity and cytocompatibility profiles. In particular, the effects of cationic conformational flexibility, hydrophobic domain flexibility, and hydrophobic domain architecture were evaluated. Their influence on antimicrobial efficacy in Gram-positive and Gram-negative bacteria was determined, and their safety profiles were established by assessing their impact on mammalian cells. All CAms have a potent activity against bacteria, and hydrophobic domain rigidity and branched architecture contribute to specificity. The insights gained from this project will aid in the optimization of CAm structures.


Assuntos
Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Tensoativos/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Cátions/síntese química , Cátions/química , Cátions/farmacologia , Células Cultivadas , Bactérias Gram-Negativas/citologia , Bactérias Gram-Positivas/citologia , Humanos , Interações Hidrofóbicas e Hidrofílicas , Testes de Sensibilidade Microbiana , Tamanho da Partícula , Propriedades de Superfície , Tensoativos/síntese química , Tensoativos/química
19.
ACS Nano ; 13(2): 2262-2273, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30758938

RESUMO

Researchers have demonstrated great promise for inorganic nanowire use in analyzing cells or intracellular components. Although a stealth effect of nanowires toward cell surfaces allows preservation of the living intact cells when analyzing cells, as a completely opposite approach, the applicability to analyze intracellular components through disrupting cells is also central to understanding cellular information. However, the reported lysis strategy is insufficient for microbial cell lysis due to the cell robustness and wrong approach taken so far ( i. e., nanowire penetration into a cell membrane). Here we propose a nanowire-mediated lysis method for microbial cells by introducing the rupture approach initiated by cell membrane stretching; in other words, the nanowires do not penetrate the membrane, but rather they break the membrane between the nanowires. Entangling cells with the bacteria-compatible and flexible nanowires and membrane stretching of the entangled cells, induced by the shear force, play important roles for the nanowire-mediated lysis to Gram-positive and Gram-negative bacteria and yeast cells. Additionally, the nanowire-mediated lysis is readily compatible with the loop-mediated isothermal amplification (LAMP) method because the lysis is triggered by simply introducing the microbial cells. We show that an integration of the nanowire-mediated lysis with LAMP provides a means for a simple, rapid, one-step identification assay (just introducing a premixed solution into a device), resulting in visual chromatic identification of microbial cells. This approach allows researchers to develop a microfluidic analytical platform not only for microbial cell identification including drug- and heat-resistance cells but also for on-site detection without any contamination.


Assuntos
Bactérias Gram-Negativas/citologia , Bactérias Gram-Positivas/citologia , Nanofios/química , Saccharomyces cerevisiae/citologia , Óxido de Zinco/química
20.
Microb Pathog ; 126: 323-331, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30458252

RESUMO

Many essential oils (EOs) are screened as potential sources of antimicrobial compounds. EOs from the genus Satureja have recognized biological properties, including analgesic, anti-inflammatory, immunomodulatory, anticancer, and antimicrobial activity. This study aimed to obtain a metabolite profile of commercial essential oil of S. montana L. (SEO) and to evaluate its antimicrobial properties, both alone and combined with gentamicin towards Gram-negative and Gram-positive bacterial strains. Untargeted analyses based on direct infusion Fourier-transform ion cyclotron resonance mass spectrometry (FT-ICR MS) and on GC-MS have provided a high metabolome coverage, allowing to identify carvacrol, cymene and thymol as the major components of commercial SEO. SEO exerted an antimicrobial activity and induced a synergistic interaction with gentamicin against both reference and clinical bacterial strains. A significant reduction of Escherichia coli, Staphylococcus aureus and Listeria monocytogenes biofilm formation was induced by SEO. As a result of SEO treatment, clear morphological bacterial alterations were visualized by scanning electron microscopy: L. monocytogenes and S. aureus showed malformed cell surface or broken cells with pores formation, whereas E. coli displayed collapsed cell surface. These results encourage further studies about bactericidal and antibiotic synergistic effect of SEO for combined therapy in clinical setting as well as in agricultural systems.


Assuntos
Anti-Infecciosos/farmacologia , Gentamicinas/farmacologia , Óleos Voláteis/farmacologia , Extratos Vegetais/farmacologia , Óleos de Plantas/farmacologia , Satureja/química , Biofilmes/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cimenos , Combinação de Medicamentos , Sinergismo Farmacológico , Cromatografia Gasosa-Espectrometria de Massas , Bactérias Gram-Negativas/citologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/citologia , Bactérias Gram-Positivas/efeitos dos fármacos , Humanos , Testes de Sensibilidade Microbiana , Monoterpenos/isolamento & purificação , Monoterpenos/farmacologia , Óleos Voláteis/química , Óleos de Plantas/química , Timol/isolamento & purificação , Timol/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...