Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Nanomedicine ; 19: 4007-4019, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38715701

RESUMEN

Introduction: Nanosized outer membrane vesicles (OMVs) from Gram-negative bacteria have attracted increasing interest because of their antitumor activity. However, the antitumor effects of MVs isolated from Gram-positive bacteria have rarely been investigated. Methods: MVs of Staphylococcus aureus USA300 were prepared and their antitumor efficacy was evaluated using tumor-bearing mouse models. A gene knock-in assay was performed to generate luciferase Antares2-MVs for bioluminescent detection. Cell counting kit-8 and lactic dehydrogenase release assays were used to detect the toxicity of the MVs against tumor cells in vitro. Active caspase-1 and gasdermin D (GSDMD) levels were determined using Western blot, and the tumor inhibition ability of MVs was determined in B16F10 cells treated with a caspase-1 inhibitor. Results: The vesicular particles of S. aureus USA300 MVs were 55.23 ± 8.17 nm in diameter, and 5 µg of MVs remarkably inhibited the growth of B16F10 melanoma in C57BL/6 mice and CT26 colon adenocarcinoma in BALB/c mice. The bioluminescent signals correlated well with the concentrations of the engineered Antares2-MVs (R2 = 0.999), and the sensitivity for bioluminescence imaging was 4 × 10-3 µg. Antares2-MVs can directly target tumor tissues in vivo, and 20 µg/mL Antares2-MVs considerably reduced the growth of B16F10 and CT26 tumor cells, but not non-carcinomatous bEnd.3 cells. MV treatment substantially increased the level of active caspase-1, which processes GSDMD to trigger pyroptosis in tumor cells. Blocking caspase-1 activation with VX-765 significantly protected tumor cells from MV killing in vitro and in vivo. Conclusion: S. aureus MVs can kill tumor cells by activating the pyroptosis pathway, and the induction of pyroptosis in tumor cells is a promising strategy for cancer treatment.


Asunto(s)
Caspasa 1 , Ratones Endogámicos BALB C , Piroptosis , Staphylococcus aureus , Animales , Piroptosis/efectos de los fármacos , Caspasa 1/metabolismo , Línea Celular Tumoral , Staphylococcus aureus/fisiología , Staphylococcus aureus/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión a Fosfato/metabolismo , Melanoma Experimental/patología , Neoplasias del Colon , Antineoplásicos/farmacología , Antineoplásicos/química , Membrana Externa Bacteriana/efectos de los fármacos , Femenino
2.
FEMS Microbiol Lett ; 368(21-24)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34910142

RESUMEN

Almost all major classes of bacteria are surrounded by a peptidoglycan cell wall, which is a crucial target for antibiotics. It is now understood that many bacteria can tolerate loss of the cell wall provided that they are in an isotonic environment. Furthermore, in some cases the cells can continue to proliferate in a state known as the L-form. L-form proliferation occurs by an unusual blebbing or tubulation mechanism that is completely independent of the normally essential division machine or cell wall synthetic enzymes, and is resistant to cell wall-active antibiotics. However, the growth is limited by reactive oxygen species generated by the respiratory chain pathway. In this work, we examined the walled to L-form transition in a pathogenic Gram-negative bacterium, Streptobacillus moniliformis, which naturally lacks the respiratory chain pathway, under aerobic conditions. L-form-like cells often emerged spontaneously, but proliferation was not observed unless the cells were treated with cell wall-active antibiotics. Time-lapse imaging revealed that cell division of S. moniliformis L-forms involves unusual membrane dynamics with an apparent imbalance between outer membrane and cytoplasmic volume growth. The results suggest that outer membrane expansion may be an important general factor for L-form proliferation of diderm bacteria.


Asunto(s)
Formas L , Antibacterianos/farmacología , Membrana Externa Bacteriana/efectos de los fármacos , Membrana Externa Bacteriana/metabolismo , Proliferación Celular/fisiología , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Citoplasma/metabolismo , Formas L/fisiología , Streptobacillus/efectos de los fármacos , Streptobacillus/crecimiento & desarrollo
3.
Small Methods ; 5(12): e2101304, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34928043

RESUMEN

Treatment of microbial-associated infections continues to be hampered by impaired antibacterial efficiency and the variability in nanomedicines. Herein, an octapeptide library with a double-layered zipper, constructed via a systematic arrangement, simplifying the sequence and optimizing the structure (diverse motifs including surfactant-like, central-bola, and end-bola), is assessed in terms of biological efficiency and self-assembly properties. The results indicate that peptides with double-layered Trp zipper exhibit significant antimicrobial activity. Extracellularly, affinity interactions between micelles and bacteria induce the lateral flow of the membrane and electric potential perturbation. Intracellularly, lead molecules cause apoptosis-like death, as indicated by excessive accumulation of reactive oxygen species, generation of a DNA ladder, and upregulation of mazEF expression. Among them, RW-1 performs the best in vivo and in vitro. The intersecting combination of Trp zipper and surfactants possesses overwhelming superiority with respect to bacterial sweepers (therapeutic index [TI] = 52.89), nanostructures (micelles), and bacterial damage compared to RW-2 (central-bola) and RW-3 (end-bola). These findings confirm that the combination of double-layered Trp zipper and surfactants has potential for application as a combined motif for combating microbial infection and connects the vast gap between antimicrobial peptides and self-assembly, such as Jacob's ladder.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Escherichia coli/crecimiento & desarrollo , Tensoactivos/química , Triptófano/química , Secuencia de Aminoácidos , Péptidos Catiónicos Antimicrobianos/química , Membrana Externa Bacteriana/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Plomo/química , Micelas , Viabilidad Microbiana/efectos de los fármacos , Triptófano/genética
4.
Microbiol Spectr ; 9(2): e0029921, 2021 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-34643411

RESUMEN

Bovine mastitis infection in dairy cattle is a significant economic burden for the dairy industry globally. To reduce the use of antibiotics in treatment of clinical mastitis, new alternative treatment options are needed. Antimicrobial peptides from bacteria, also known as bacteriocins, are potential alternatives for combating mastitis pathogens. In search of novel bacteriocins against mastitis pathogens, we screened samples of Norwegian bovine raw milk and found a Streptococcus uberis strain with potent antimicrobial activity toward Enterococcus, Streptococcus, Listeria, and Lactococcus. Whole-genome sequencing of the strain revealed a multibacteriocin gene cluster encoding one class IIb bacteriocin, two class IId bacteriocins, in addition to a three-component regulatory system and a dedicated ABC transporter. Isolation and purification of the antimicrobial activity from culture supernatants resulted in the detection of a 6.3-kDa mass peak by matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry, a mass corresponding to the predicted size of one of the class IId bacteriocins. The identification of this bacteriocin, called ubericin K, was further confirmed by in vitro protein synthesis, which showed the same inhibitory spectrum as the purified antimicrobial compound. Ubericin K shows highest sequence similarity to the class IId bacteriocins bovicin 255, lactococcin A, and garvieacin Q. We found that ubericin K uses the sugar transporter mannose phosphotransferase (PTS) as a target receptor. Further, by using the pHlourin sensor system to detect intracellular pH changes due to leakage across the membrane, ubericin K was shown to be a pore former, killing target cells by membrane disruption. IMPORTANCE Bacterial infections in dairy cows are a major burden to farmers worldwide because infected cows require expensive treatments and produce less milk. Today, infected cows are treated with antibiotics, a practice that is becoming less effective due to antibiotic resistance. Compounds other than antibiotics also exist that kill bacteria causing infections in cows; these compounds, known as bacteriocins, are natural products produced by other bacteria in the environment. In this work, we discover a new bacteriocin that we call ubericin K, which kills several species of bacteria known to cause infections in dairy cows. We also use in vitro synthesis as a novel method for rapidly characterizing bacteriocins directly from genomic data, which could be useful for other researchers. We believe that ubericin K and the methods described in this work will aid in the transition away from antibiotics in the dairy industry.


Asunto(s)
Antibacterianos/uso terapéutico , Bacteriocinas/uso terapéutico , Enfermedades de los Bovinos/tratamiento farmacológico , Mastitis Bovina/tratamiento farmacológico , Streptococcus/metabolismo , Animales , Membrana Externa Bacteriana/efectos de los fármacos , Membrana Externa Bacteriana/patología , Bacteriocinas/genética , Bovinos , Enfermedades de los Bovinos/microbiología , Enterococcus/efectos de los fármacos , Enterococcus/crecimiento & desarrollo , Femenino , Lactococcus/efectos de los fármacos , Lactococcus/crecimiento & desarrollo , Listeria/efectos de los fármacos , Listeria/crecimiento & desarrollo , Mastitis Bovina/microbiología , Pruebas de Sensibilidad Microbiana , Fosfotransferasas/metabolismo , Percepción de Quorum , Streptococcus/genética
5.
Int J Antimicrob Agents ; 58(5): 106434, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34525402

RESUMEN

The outer membrane of Gram-negative bacteria constitutes a permeability barrier that prevents certain antibiotics reaching their target, thus conferring a high tolerance to a wide range of antibiotics. Combined therapies of antibiotics and outer membrane-perturbing drugs have been proposed as an alternative treatment to extend the use of antibiotics active against Gram-positive bacteria to Gram-negative bacteria. Among the outer membrane-active compounds, the outer membrane-permeabilising peptides play a prominent role. They form a group of small cationic and amphipathic molecules with the ability to insert specifically into bacterial membranes, inducing their permeabilisation and/or disruption. Here we assessed the combined effect of several compounds belonging to the main antibiotic families and the cathelicidin close-to-nature outer membrane peptide D-11 against four clinically relevant Gram-negative bacteria. The results showed that peptide D-11 displays strong synergistic activity with several antibiotics belonging to different families, in particular against Klebsiella pneumoniae, even better than some other outer membrane-active peptides that are currently in clinical trials, such as SPR741. Notably, we observed this activity in vitro, ex vivo in a newly designed bacteraemia model, and in vivo in a mouse abscess infection model. Overall, our results suggest that D-11 is a good candidate to repurpose the activity of traditional antibiotics against K. pneumoniae.


Asunto(s)
Absceso/tratamiento farmacológico , Antibacterianos/farmacología , Membrana Externa Bacteriana/efectos de los fármacos , Catelicidinas/farmacología , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella pneumoniae/efectos de los fármacos , Absceso/microbiología , Infecciones por Acinetobacter/tratamiento farmacológico , Acinetobacter baumannii/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Quimioterapia Combinada , Escherichia coli/efectos de los fármacos , Infecciones por Escherichia coli/tratamiento farmacológico , Femenino , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Permeabilidad/efectos de los fármacos , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos
6.
mBio ; 12(5): e0228521, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34517753

RESUMEN

Bacteroidetocins are a family of antibacterial peptide toxins that are produced by and target members of the phylum Bacteroidetes. To date, 19 bacteroidetocins have been identified, and four have been tested and shown to kill diverse Bacteroidales species (M. J. Coyne, N. Béchon, L. M. Matano, V. L. McEneany, et al., Nat Commun 10:3460, 2019, https://doi.org/10.1038/s41467-019-11494-1). Here, we identify the target and likely mechanism of action of the bacteroidetocins. We selected seven spontaneous mutants of four different genera, all resistant to bacteroidetocin A (Bd-A) and found that all contained mutations in a single gene, bamA. Construction of three of these bamA mutants in the wild-type (WT) strains confirmed they confer resistance to Bd-A as well as to other bacteroidetocins. We identified an aspartate residue of BamA at the beginning of exterior loop 3 (eL3) that, when altered, renders strains resistant to Bd-A. Analysis of a panel of diverse Bacteroidales strains showed a correlation between the presence of this aspartate residue and Bd-A sensitivity. Fluorescence microscopy and transmission electron microscopy (TEM) analysis of Bd-A-treated cells showed cellular morphological changes consistent with a BamA defect. Transcriptomic analysis of Bd-A-treated cells revealed gene expression changes indicative of cell envelope stress. Studies in mice revealed that bacteroidetocin-resistant mutants are outcompeted by their WT strain in vivo. Analyses of longitudinal human gut isolates showed that bamA mutations leading to bacteroidetocin resistance do not become fixed in the human gut, even in bacteroidetocin-producing strains and nonproducing coresident strains. Together, these data lend further support to the applicability of the bacteroidetocins as therapeutic peptides in the treatment of maladies involving Bacteroidales species. IMPORTANCE The bacteroidetocins are a newly discovered class of bacteriocins specific to Bacteroidetes with a spectrum of targets extending from symbiotic gut Bacteroides, Parabacteroides, and Prevotella species to pathogenic oral and vaginal Prevotella species. We previously showed that one such bacteroidetocin, Bd-A, is active at nanomolar concentrations, is water soluble, and is bactericidal, all desirable features in a therapeutic antibacterial peptide. Here, we identify the target of several of the bacteroidetocins as the essential outer membrane protein BamA. Although mutations in bamA can be selected in bacteria grown in vitro, we show both in a mouse model and in human gut ecosystems that bamA mutants leading to Bd-A resistance are fitness attenuated and are not selected. These features further support the potential usefulness of the bacteroidetocins as therapeutics for maladies associated with pathogenic Prevotella species, such as recurrent bacterial vaginosis, for which there are few effective treatments.


Asunto(s)
Antibacterianos/farmacología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Bacteriocinas/farmacología , Bacteroidetes/efectos de los fármacos , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/efectos de los fármacos , Membrana Externa Bacteriana/metabolismo , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Bacteroidetes/química , Bacteroidetes/genética , Bacteroidetes/fisiología , Farmacorresistencia Bacteriana , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Infecciones por Bacterias Gramnegativas/microbiología , Humanos , Ratones , Alineación de Secuencia , Simbiosis
7.
Biomed Res Int ; 2021: 4134713, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34540993

RESUMEN

Previous work stated that Khaya senegalensis, Anacardium ouest L., Pterocarpus erinaceus, Diospyros mespiliformis, Ocimum gratissimum, Manihot esculenta, Vernonia amygdalina Delile, and Daniellia oliveri have a great potential for the fight against infectious diarrhea. However, data on their antibacterial activity on strains of bacteria responsible for infectious diarrhea are not available. This study is aimed at elucidating the mechanism of action of the antibacterial effect of these plants on some bacterial strains responsible for diarrheal infections. The design of the study included first evaluating the degree of sensitivity of Salmonella typhimurium 14028, Escherichia coli ATCC 25922, Shigella spp., and Salmonella spp. strains to aqueous and hydroethanolic extracts of each plant, followed by the determination of minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), and antibiotic power (Pa). This screening was completed with the evaluation of the possible mode of action of the extracts by testing the membrane permeability of these bacterial strains. The data collected indicate that the bacterial strains tested were sensitive to the extracts to varying degrees, except Cassia sieberiana DC and Pseudocedrela kotschyi extracts. For the active extracts, inhibition diameters ranged from 18.33 mm to 7 mm. With the exception of Escherichia coli, all strains were sensitive to the aqueous and hydroethanolic extracts of Anacardium occidentale. MICs vary between 3.37 and 25 mg/ml. Membrane permeability test data show that all active extracts affect the bacterial strains tested by attacking the stability of their outer membrane. For all active extracts, the high percentage of membrane destabilization of the bacteria is significantly (p < 0.05) better than that of cefixime used as a reference. Thus, it appears that these extracts can destroy Gram-negative bacteria and increase the fluidity and permeability of their cytoplasmic membrane. The knowledge of the mechanism of action of these extracts is an interesting contribution to the fundamental knowledge on the alternative that medicinal plants represent to antibiotics. These extracts can be used in the management of infectious diarrhea.


Asunto(s)
Membrana Externa Bacteriana/efectos de los fármacos , Disentería/tratamiento farmacológico , Extractos Vegetales/farmacología , África Occidental , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Membrana Externa Bacteriana/metabolismo , Diarrea/tratamiento farmacológico , Escherichia coli/efectos de los fármacos , Humanos , Medicinas Tradicionales Africanas/métodos , Pruebas de Sensibilidad Microbiana , Plantas Medicinales , Salmonella/efectos de los fármacos , Shigella/efectos de los fármacos
8.
Microbiol Spectr ; 9(2): e0000621, 2021 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-34468186

RESUMEN

Avian pathogenic Escherichia coli (APEC), a subgroup of extraintestinal pathogenic E. coli (ExPEC), causes colibacillosis in chickens and is reportedly implicated in urinary tract infections and meningitis in humans. A major limitation for the current ExPEC antibiotic therapy is the development of resistance, and antibacterial drugs that can circumvent this problem are critically needed. Here, we evaluated eight novel membrane-affecting anti-APEC small molecule growth inhibitors (GIs), identified in our previous study, against APEC infection in chickens. Among the GIs tested, GI-7 (the most effective), when administered orally (1 mg/kg of body weight), reduced the mortality (41.7%), severity of lesions (62.9%), and APEC load (2.6 log) in chickens. Furthermore, GI-7 administration at an optimized dose (60 mg/liter) in drinking water also reduced the mortality (14.7%), severity of lesions (29.5%), and APEC load (2.2 log) in chickens. The abundances of Lactobacillus and oleate were increased in the cecum and serum, respectively, of GI-7-treated chickens. Pharmacokinetic analysis revealed that GI-7 was readily absorbed with minimal accumulation in the tissues. Earlier, we showed that GI-7 induced membrane blebbing and increased membrane permeability in APEC, suggesting an effect on the APEC membrane. Consistent with this finding, the expression of genes essential for maintaining outer membrane (OM) integrity was downregulated in GI-7-treated APEC. Furthermore, decreased levels of lipopolysaccharide (LPS) transport (Lpt) proteins and LPS were observed in GI-7-treated APEC. However, the mechanism of action of GI-7 currently remains unknown and needs further investigation. Our studies suggest that GI-7 represents a promising novel lead compound that can be developed to treat APEC infection in chickens and related human ExPEC infections. IMPORTANCE APEC is a subgroup of ExPEC, and genetic similarities of APEC with human ExPECs, including uropathogenic E. coli (UPEC) and neonatal meningitis E. coli (NMEC), have been reported. Our study identified a novel small molecule growth inhibitor, GI-7, effective in reducing APEC infection in chickens with an efficacy similar to that of the currently used antibiotic sulfadimethoxine, notably with an 8-times-lower dose. GI-7 affects the OM integrity and decreases the Lpt protein and LPS levels in APEC, an antibacterial mechanism that can overcome the antibiotic resistance problem. Overall, GI-7 represents a promising lead molecule/scaffold for the development of novel antibacterial therapies that could have profound implications for treating APEC infections in chickens, as well as human infections caused by ExPECs and other related Gram-negative bacteria. Further elucidation of the mechanism of action of GI-7 and identification of its target(s) in APEC will benefit future novel antibacterial development efforts.


Asunto(s)
Antibacterianos/farmacología , Membrana Externa Bacteriana/patología , Infecciones por Escherichia coli/tratamiento farmacológico , Escherichia coli Patógena Extraintestinal/efectos de los fármacos , Inhibidores de Crecimiento/farmacología , Animales , Carga Bacteriana/efectos de los fármacos , Membrana Externa Bacteriana/efectos de los fármacos , Pollos/microbiología , Modelos Animales de Enfermedad , Escherichia coli Patógena Extraintestinal/crecimiento & desarrollo , Humanos , Enfermedades de las Aves de Corral/tratamiento farmacológico , Enfermedades de las Aves de Corral/microbiología , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología
9.
Biochem Biophys Res Commun ; 572: 145-150, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34364294

RESUMEN

4-Hydroxyphenylacetic acid (4-HPCA) is the major intestinal metabolite of kaempferol and polymeric proanthocyanidins whereas the effect of 4-HPCA on Listeria monocytogenes remains unknown. In this study, we investigated the effect and mechanism of action of 4-HPCA on the highly lethal foodborne pathogen Listeria monocytogenes. Our results indicated that 4-HPCA inhibited L. monocytogenes growth and proliferation in a dose-dependent manner. In particular, L. monocytogenes displayed negligible growth or proliferation after 4-HPCA treatment (15.61 mM) for 24 h. The impact of 4-HPCA on cell membrane structure and function was investigated in terms of fluorometric cell membrane integrity, zeta potential and relative electrical conductivity. We observed an approximately 15 % fluorescence reduction in the cell membrane after MIC treatment. The zeta potential of the bacteria shifted significantly from -49.74 to -43.70 mV, -36.65 mV and -37.97 mV after treatment with 4-HPCA at the MIC for 0 h, 3 h and 12 h, respectively. The absolute value of the relative electrical conductivities increased significantly following 3 h, 6 h, 9 h and 15 h of 4-HPCA treatment at the MIC level. The results of scanning electron microscopy (SEM) showed that cells treated with 4-HPCA displayed a wrinkled morphology and irregular shapes. Moreover, 4-HPCA obviously decreased the expression of three virulence genes (hlyA, prfA, and inlA) in L. monocytogenes after 12 h of treatment. All these results verified that 4-HPCA, as an effective antibacterial compound against L. monocytogenes, could cause cell death through cell membrane damage and decrease the expression of three virulence factors.


Asunto(s)
Antibacterianos/farmacología , Listeria monocytogenes/efectos de los fármacos , Fenilacetatos/farmacología , Membrana Externa Bacteriana/efectos de los fármacos , Pruebas de Sensibilidad Microbiana
10.
Int J Biol Macromol ; 185: 620-628, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34216663

RESUMEN

Mandarin (Citrus reticulata L.) essential oil (MEO) reportedly displays excellent antimicrobial properties. In this study, MEO was loaded into chitosan nanoparticles (CSNPs). The characteristics, antibacterial properties and benefit in pork preservation of MEO-CSNPs were evaluated. The MEO-CSNPs displayed an excellent encapsulation efficiency (EE) (67.32%-82.35%), the particle size values of 131.3 nm-161.9 nm, and the absolute zeta potential values above 30 mV. Fourier transform infrared spectroscopy (FTIR), X-ray diffraction (XRD) analysis, and thermogravimetric analysis (TGA) revealed that the MEO was incorporated into CSNPs without requiring a chemical reaction, the antibacterial activity of the MEO remained. Furthermore, the damage of MEO-chitosan nanoemulsions (MEO-CSs) to the cell membranes of Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) was confirmed by the change of bacterial cell morphology. The anti-biofilm assays verified that the MEO-CSs substantially inhibited biofilm formation and destroyed the mature biofilms. MEO-CSs were also applied to pork, proving a great potential for pork preservation. This study provides a potential approach for developing and utilizing MEO-CSs as natural antimicrobial agents in the food industry.


Asunto(s)
Quitosano/química , Citrus/química , Conservación de Alimentos/métodos , Aceites Volátiles/farmacología , Carne de Cerdo , Animales , Membrana Externa Bacteriana/efectos de los fármacos , Biopelículas/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Nanopartículas , Aceites Volátiles/química , Tamaño de la Partícula , Aceites de Plantas/química , Aceites de Plantas/farmacología , Espectroscopía Infrarroja por Transformada de Fourier , Staphylococcus aureus/efectos de los fármacos , Porcinos , Termogravimetría , Difracción de Rayos X
11.
Int J Mol Sci ; 22(11)2021 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-34072512

RESUMEN

Synthetic materials commonly used in the packaging industry generate a considerable amount of waste each year. Chitosan is a promising feedstock for the production of functional biomaterials. From a biological point of view, chitosan is very attractive for food packaging. The purposes of this study were to evaluate the antibacterial activity of a set of chitosan-metal oxide films and different chitosan-modified graphene (oxide) films against two foodborne pathogens: Campylobacter jejuni ATCC 33560 and Listeria monocytogenes 19115. Moreover, we wanted to check whether the incorporation of antimicrobial constituents such as TiO2, ZnO, Fe2O3, Ag, and graphene oxide (GO) into the polymer matrices can improve the antibacterial properties of these nanocomposite films. Finally, this research helps elucidate the interactions of these materials with eukaryotic cells. All chitosan-metal oxide films and chitosan-modified graphene (oxide) films displayed improved antibacterial (C. jejuni ATCC 33560 and L. monocytogenes 19115) properties compared to native chitosan films. The CS-ZnO films had excellent antibacterial activity towards L. monocytogenes (90% growth inhibition). Moreover, graphene-based chitosan films caused high inhibition of both tested strains. Chitosan films with graphene (GO, GOP, GOP-HMDS, rGO, GO-HMDS, rGOP), titanium dioxide (CS-TiO2 20:1a, CS-TiO2 20:1b, CS-TiO2 2:1, CS-TiO2 1:1a, CS-TiO2 1:1b) and zinc oxide (CS-ZnO 20:1a, CS-ZnO 20:1b) may be considered as a safe, non-cytotoxic packaging materials in the future.


Asunto(s)
Antiinfecciosos/química , Antiinfecciosos/farmacología , Materiales Biocompatibles , Quitosano , Microbiología de Alimentos , Embalaje de Alimentos , Membranas Artificiales , Antibacterianos/química , Antibacterianos/farmacología , Membrana Externa Bacteriana/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Quitosano/química , Metales/química , Polímeros
12.
Appl Environ Microbiol ; 87(17): e0056721, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34132592

RESUMEN

Avian pathogenic Escherichia coli (APEC), an extraintestinal pathogenic E. coli (ExPEC), causes colibacillosis in chickens and is reportedly associated with urinary tract infections and meningitis in humans. Development of resistance is a major limitation of current ExPEC antibiotic therapy. New antibacterials that can circumvent resistance problem such as antimicrobial peptides (AMPs) are critically needed. Here, we evaluated the efficacy of Lactobacillus rhamnosus GG (LGG)-derived peptides against APEC and uncovered their potential antibacterial targets. Three peptides (NPSRQERR [P1], PDENK [P2], and VHTAPK [P3]) displayed inhibitory activity against APEC. These peptides were effective against APEC in biofilm and chicken macrophage HD11 cells. Treatment with these peptides reduced the cecum colonization (0.5 to 1.3 log) of APEC in chickens. Microbiota analysis revealed two peptides (P1 and P2) decreased Enterobacteriaceae abundance with minimal impact on overall cecal microbiota of chickens. Bacterial cytological profiling showed peptides disrupt APEC membranes either by causing membrane shedding, rupturing, or flaccidity. Furthermore, gene expression analysis revealed that peptides downregulated the expression of ompC (>13.0-fold), ompF (>11.3-fold), and mlaA (>4.9-fold), genes responsible for the maintenance of outer membrane (OM) lipid asymmetry. Consistently, immunoblot analysis also showed decreased levels of OmpC and MlaA proteins in APEC treated with peptides. Alanine scanning studies revealed residues crucial (P1, N, E, R and P; P2, D and E; P3, T, P, and K) for their activity. Overall, our study identified peptides with a new antibacterial target that can be developed to control APEC infections in chickens, thereby curtailing poultry-originated human ExPEC infections. IMPORTANCE Avian pathogenic Escherichia coli (APEC) is a subgroup of extraintestinal pathogenic E. coli (ExPEC) and considered a foodborne zoonotic pathogen transmitted through consumption of contaminated poultry products. APEC shares genetic similarities with human ExPECs, including uropathogenic E. coli (UPEC) and neonatal meningitis E. coli (NMEC). Our study identified Lactobacillus rhamnosus GG (LGG)-derived peptides (P1 [NPSRQERR], P2 [PDENK], and P3 [VHTAPK]) effective in reducing APEC infection in chickens. Antimicrobial peptides (AMPs) are regarded as ideal candidates for antibacterial development because of their low propensity for resistance development and ability to kill resistant bacteria. Mechanistic studies showed peptides disrupt the APEC membrane by affecting the MlaA-OmpC/F system responsible for the maintenance of outer membrane (OM) lipid asymmetry, a promising new druggable target to overcome resistance problems in Gram-negative bacteria. Altogether, these peptides can provide a valuable approach for development of novel anti-ExPEC therapies, including APEC, human ExPECs, and other related Gram-negative pathogens. Furthermore, effective control of APEC infections in chickens can curb poultry-originated ExPEC infections in humans.


Asunto(s)
Infecciones por Escherichia coli/veterinaria , Proteínas de Escherichia coli/metabolismo , Escherichia coli Patógena Extraintestinal/efectos de los fármacos , Proteínas de Transferencia de Fosfolípidos/metabolismo , Proteínas Citotóxicas Formadoras de Poros/farmacología , Porinas/metabolismo , Enfermedades de las Aves de Corral/microbiología , Animales , Membrana Externa Bacteriana/efectos de los fármacos , Membrana Externa Bacteriana/metabolismo , Biopelículas/efectos de los fármacos , Pollos/microbiología , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Escherichia coli Patógena Extraintestinal/genética , Escherichia coli Patógena Extraintestinal/crecimiento & desarrollo , Escherichia coli Patógena Extraintestinal/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Proteínas de Transferencia de Fosfolípidos/genética , Porinas/genética , Enfermedades de las Aves de Corral/tratamiento farmacológico
13.
FEMS Microbiol Lett ; 368(11)2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34048543

RESUMEN

Gram-negative pathogens are a rapidly increasing threat to human health worldwide due to high rates of antibiotic resistance and the lack of development of novel antibiotics. The protective cell envelope of gram-negative bacteria is a major permeability barrier that contributes to the problem by restricting the uptake of antibiotics. On the other hand, its unique architecture also makes it a suitable target for antibiotic interference. In particular, essential multiprotein machines that are required for biogenesis of the outer membrane have attracted attention in antibacterial design strategies. Recently, significant progress has been made in the development of inhibitors of the ß-barrel assembly machine (BAM) complex. Here, we summarize the current state of drug development efforts targeting the BAM complex in pursuit of new antibiotics.


Asunto(s)
Antibacterianos/farmacología , Proteínas de la Membrana Bacteriana Externa/antagonistas & inhibidores , Antibacterianos/química , Membrana Externa Bacteriana/efectos de los fármacos , Membrana Externa Bacteriana/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Farmacorresistencia Bacteriana/genética , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/patogenicidad , Humanos , Mutación , Virulencia/efectos de los fármacos
14.
Biomolecules ; 11(5)2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33946460

RESUMEN

Formulations with lactate as an antimicrobial and high-pressure processing (HPP) as a lethal treatment are combined strategies used to control L. monocytogenes in cooked meat products. Previous studies have shown that when HPP is applied in products with lactate, the inactivation of L. monocytogenes is lower than that without lactate. The purpose of the present work was to identify the molecular mechanisms underlying the piezo-protection effect of lactate. Two L. monocytogenes strains (CTC1034 and EGDe) were independently inoculated in a cooked ham model medium without and with 2.8% potassium lactate. Samples were pressurized at 400 MPa for 10 min at 10 °C. Samples were subjected to RNA extraction, and a shotgun transcriptome sequencing was performed. The short exposure of L. monocytogenes cells to lactate through its inoculation in a cooked ham model with lactate 1h before HPP promoted a shift in the pathogen's central metabolism, favoring the metabolism of propanediol and ethanolamine together with the synthesis of the B12 cofactor. Moreover, the results suggest an activated methyl cycle that would promote modifications in membrane properties resulting in an enhanced resistance of the pathogen to HPP. This study provides insights on the mechanisms developed by L. monocytogenes in response to lactate and/or HPP and sheds light on the understanding of the piezo-protective effect of lactate.


Asunto(s)
Membrana Externa Bacteriana/efectos de los fármacos , Ácidos Grasos/metabolismo , Ácido Láctico/farmacología , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/metabolismo , Productos de la Carne/microbiología , Animales , Antibacterianos/farmacología , ADN Bacteriano , Etanolamina/metabolismo , Manipulación de Alimentos/métodos , Microbiología de Alimentos , Industria de Procesamiento de Alimentos/métodos , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Listeria monocytogenes/genética , Listeriosis/microbiología , Membranas/efectos de los fármacos , Redes y Vías Metabólicas , Presión , Glicoles de Propileno/metabolismo , Porcinos , Temperatura , Factores de Tiempo , Vitamina B 12/biosíntesis
15.
J Med Chem ; 64(11): 7630-7645, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34009979

RESUMEN

A class of quinazolone thiazoles was identified as new structural scaffolds for potential antibacterial conquerors to tackle dreadful resistance. Some prepared compounds exhibited favorable bacteriostatic efficiencies on tested bacteria, and the most representative 5j featuring the 4-trifluoromethylphenyl group possessed superior performances against Escherichia coli and Pseudomonas aeruginosa to norfloxacin. Further studies revealed that 5j with inappreciable hemolysis could hinder the formation of bacterial biofilms and trigger reactive oxygen species generation, which could take responsibility for emerging low resistance. Subsequent paralleled exploration discovered that 5j not only disintegrated outer and inner membranes to induce leakage of cytoplasmic contents but also broke the metabolism by suppressing dehydrogenase. Meanwhile, derivative 5j could intercalate into DNA to exert powerful antibacterial properties. Moreover, compound 5j gave synergistic effects against some Gram-negative bacteria in combination with norfloxacin. These findings indicated that this novel structural type of quinazolone thiazoles showed therapeutic foreground in struggling with Gram-negative bacterial infections.


Asunto(s)
Antibacterianos/farmacología , Azoles/química , Bacterias Gramnegativas/efectos de los fármacos , Tiazoles/química , Antibacterianos/química , Antibacterianos/metabolismo , Membrana Externa Bacteriana/efectos de los fármacos , Membrana Externa Bacteriana/metabolismo , Sitios de Unión , Unión Competitiva , Biopelículas/efectos de los fármacos , Cristalografía por Rayos X , Diseño de Fármacos , Farmacorresistencia Bacteriana/efectos de los fármacos , Escherichia coli/fisiología , Bacterias Grampositivas/efectos de los fármacos , L-Lactato Deshidrogenasa/antagonistas & inhibidores , L-Lactato Deshidrogenasa/metabolismo , Pruebas de Sensibilidad Microbiana , Conformación Molecular , Simulación del Acoplamiento Molecular , Pseudomonas aeruginosa/fisiología , Especies Reactivas de Oxígeno/metabolismo , Tiazoles/metabolismo , Tiazoles/farmacología
16.
Sci Rep ; 11(1): 10446, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34001940

RESUMEN

Lipophosphonoxins (LPPOs) are small modular synthetic antibacterial compounds that target the cytoplasmic membrane. First-generation LPPOs (LPPO I) exhibit an antimicrobial activity against Gram-positive bacteria; however they do not exhibit any activity against Gram-negatives. Second-generation LPPOs (LPPO II) also exhibit broadened activity against Gram-negatives. We investigated the reasons behind this different susceptibility of bacteria to the two generations of LPPOs using model membranes and the living model bacteria Bacillus subtilis and Escherichia coli. We show that both generations of LPPOs form oligomeric conductive pores and permeabilize the bacterial membrane of sensitive cells. LPPO activity is not affected by the value of the target membrane potential, and thus they are also active against persister cells. The insensitivity of Gram-negative bacteria to LPPO I is probably caused by the barrier function of the outer membrane with LPS. LPPO I is almost incapable of overcoming the outer membrane in living cells, and the presence of LPS in liposomes substantially reduces their activity. Further, the antimicrobial activity of LPPO is also influenced by the phospholipid composition of the target membrane. A higher proportion of phospholipids with neutral charge such as phosphatidylethanolamine or phosphatidylcholine reduces the LPPO permeabilizing potential.


Asunto(s)
Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Bacillus subtilis/efectos de los fármacos , Membrana Externa Bacteriana/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Antibacterianos/síntesis química , Péptidos Catiónicos Antimicrobianos/síntesis química , Bacillus subtilis/química , Bacillus subtilis/citología , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/metabolismo , Permeabilidad de la Membrana Celular , Escherichia coli/química , Escherichia coli/citología , Membrana Dobles de Lípidos , Potenciales de la Membrana/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Fosfatidilcolinas/análisis , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/análisis , Fosfatidiletanolaminas/metabolismo
17.
Sci Rep ; 11(1): 6603, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33758343

RESUMEN

New antimicrobial agents are urgently needed, especially to eliminate multidrug resistant Gram-negative bacteria that stand for most antibiotic-resistant threats. In the following study, we present superior properties of an engineered antimicrobial peptide, OMN6, a 40-amino acid cyclic peptide based on Cecropin A, that presents high efficacy against Gram-negative bacteria with a bactericidal mechanism of action. The target of OMN6 is assumed to be the bacterial membrane in contrast to small molecule-based agents which bind to a specific enzyme or bacterial site. Moreover, OMN6 mechanism of action is effective on Acinetobacter baumannii laboratory strains and clinical isolates, regardless of the bacteria genotype or resistance-phenotype, thus, is by orders-of-magnitude, less likely for mutation-driven development of resistance, recrudescence, or tolerance. OMN6 displays an increase in stability and a significant decrease in proteolytic degradation with full safety margin on erythrocytes and HEK293T cells. Taken together, these results strongly suggest that OMN6 is an efficient, stable, and non-toxic novel antimicrobial agent with the potential to become a therapy for humans.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Proteínas Citotóxicas Formadoras de Poros/farmacología , Antibacterianos/química , Péptidos Catiónicos Antimicrobianos/química , Membrana Externa Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple , Proteínas Citotóxicas Formadoras de Poros/química , Ingeniería de Proteínas , Estabilidad Proteica
18.
Structure ; 29(5): 444-456.e2, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33577754

RESUMEN

The periplasm of Gram-negative bacteria is a complex, highly crowded molecular environment. Little is known about how antibiotics move across the periplasm and the interactions they experience. Here, atomistic molecular dynamics simulations are used to study the antibiotic polymyxin B1 within models of the periplasm, which are crowded to different extents. We show that PMB1 is likely to be able to "hitchhike" within the periplasm by binding to lipoprotein carriers-a previously unreported passive transport route. The simulations reveal that PMB1 forms both transient and long-lived interactions with proteins, osmolytes, lipids of the outer membrane, and the cell wall, and is rarely uncomplexed when in the periplasm. Furthermore, it can interfere in the conformational dynamics of native proteins. These are important considerations for interpreting its mechanism of action and are likely to also hold for other antibiotics that rely on diffusion to cross the periplasm.


Asunto(s)
Antibacterianos/farmacología , Membrana Externa Bacteriana/efectos de los fármacos , Proteínas de Escherichia coli/química , Proteínas de Unión Periplasmáticas/química , Polimixinas/análogos & derivados , Antibacterianos/química , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/metabolismo , Escherichia coli , Proteínas de Escherichia coli/metabolismo , Simulación de Dinámica Molecular , Periplasma/metabolismo , Proteínas de Unión Periplasmáticas/metabolismo , Polimixinas/química , Polimixinas/farmacología
19.
Commun Biol ; 4(1): 31, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33398076

RESUMEN

The development and dissemination of antibiotic-resistant bacterial pathogens is a growing global threat to public health. Novel compounds and/or therapeutic strategies are required to face the challenge posed, in particular, by Gram-negative bacteria. Here we assess the combined effect of potent cell-wall synthesis inhibitors with either natural or synthetic peptides that can act on the outer-membrane. Thus, several linear peptides, either alone or combined with vancomycin or nisin, were tested against selected Gram-negative pathogens, and the best one was improved by further engineering. Finally, peptide D-11 and vancomycin displayed a potent antimicrobial activity at low µM concentrations against a panel of relevant Gram-negative pathogens. This combination was highly active in biological fluids like blood, but was non-hemolytic and non-toxic against cell lines. We conclude that vancomycin and D-11 are safe at >50-fold their MICs. Based on the results obtained, and as a proof of concept for the newly observed synergy, a Pseudomonas aeruginosa mouse infection model experiment was also performed, showing a 4 log10 reduction of the pathogen after treatment with the combination. This approach offers a potent alternative strategy to fight (drug-resistant) Gram-negative pathogens in humans and mammals.


Asunto(s)
Antibacterianos/farmacología , Membrana Externa Bacteriana/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Péptidos/farmacología , Uridina Difosfato Ácido N-Acetilmurámico/análogos & derivados , Antibacterianos/uso terapéutico , Quimioterapia Combinada , Pruebas de Sensibilidad Microbiana , Nisina/farmacología , Nisina/uso terapéutico , Péptidos/uso terapéutico , Uridina Difosfato Ácido N-Acetilmurámico/antagonistas & inhibidores , Vancomicina/farmacología , Vancomicina/uso terapéutico
20.
Nat Biomed Eng ; 5(5): 467-480, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33390588

RESUMEN

Precision antimicrobials aim to kill pathogens without damaging commensal bacteria in the host, and thereby cure disease without antibiotic-associated dysbiosis. Here we report the de novo design of a synthetic host defence peptide that targets a specific pathogen by mimicking key molecular features of the pathogen's channel-forming membrane proteins. By exploiting physical and structural vulnerabilities within the pathogen's cellular envelope, we designed a peptide sequence that undergoes instructed tryptophan-zippered assembly within the mycolic acid-rich outer membrane of Mycobacterium tuberculosis to specifically kill the pathogen without collateral toxicity towards lung commensal bacteria or host tissue. These mycomembrane-templated assemblies elicit rapid mycobactericidal activity and enhance the potency of antibiotics by improving their otherwise poor diffusion across the rigid M. tuberculosis envelope with respect to agents that exploit transmembrane protein channels for antimycobacterial activity. This biomimetic strategy may aid the design of other narrow-spectrum antimicrobial peptides.


Asunto(s)
Antibacterianos/farmacología , Proteínas de la Membrana/genética , Mycobacterium tuberculosis/efectos de los fármacos , Péptidos/farmacología , Membrana Externa Bacteriana/efectos de los fármacos , Proteínas Bacterianas/genética , Humanos , Pulmón/efectos de los fármacos , Pulmón/microbiología , Imitación Molecular , Péptidos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...