Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Microbiol ; 15: 1386017, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38751716

RESUMEN

Background: The commensal skin bacterium Cutibacterium acnes plays a role in the pathogenesis of acne vulgaris and also causes opportunistic infections of implanted medical devices due to its ability to form biofilms on biomaterial surfaces. Poly-ß-(1→6)-N-acetyl-D-glucosamine (PNAG) is an extracellular polysaccharide that mediates biofilm formation and biocide resistance in a wide range of bacterial pathogens. The objective of this study was to determine whether C. acnes produces PNAG, and whether PNAG contributes to C. acnes biofilm formation and biocide resistance in vitro. Methods: PNAG was detected on the surface of C. acnes cells by fluorescence confocal microscopy using the antigen-specific human IgG1 monoclonal antibody F598. PNAG was detected in C. acnes biofilms by measuring the ability of the PNAG-specific glycosidase dispersin B to inhibit biofilm formation and sensitize biofilms to biocide killing. Results: Monoclonal antibody F598 bound to the surface of C. acnes cells. Dispersin B inhibited attachment of C. acnes cells to polystyrene rods, inhibited biofilm formation by C. acnes in glass and polypropylene tubes, and sensitized C. acnes biofilms to killing by benzoyl peroxide and tetracycline. Conclusion: C. acnes produces PNAG, and PNAG contributes to C. acnes biofilm formation and biocide resistance in vitro. PNAG may play a role in C. acnes skin colonization, biocide resistance, and virulence in vivo.

2.
Molecules ; 28(14)2023 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-37513185

RESUMEN

To address the high tolerance of biofilms to antibiotics, it is urgent to develop new strategies to fight against these bacterial consortia. An innovative antibiofilm nanovector drug delivery system, consisting of Dispersin B-permethylated-ß-cyclodextrin/ciprofloxacin adamantyl (DspB-ß-CD/CIP-Ad), is described here. For this purpose, complexation assays between CIP-Ad and (i) unmodified ß-CD and (ii) different derivatives of ß-CD, which are 2,3-O-dimethyl-ß-CD, 2,6-O-dimethyl-ß-CD, and 2,3,6-O-trimethyl-ß-CD, were tested. A stoichiometry of 1/1 was obtained for the ß-CD/CIP-Ad complex by NMR analysis. Isothermal Titration Calorimetry (ITC) experiments were carried out to determine Ka, ΔH, and ΔS thermodynamic parameters of the complex between ß-CD and its different derivatives in the presence of CIP-Ad. A stoichiometry of 1/1 for ß-CD/CIP-Ad complexes was confirmed with variable affinity according to the type of methylation. A phase solubility study showed increased CIP-Ad solubility with CD concentration, pointing out complex formation. The evaluation of the antibacterial activity of CIP-Ad and the 2,3-O-dimethyl-ß-CD/CIP-Ad or 2,3,6-O-trimethyl-ß-CD/CIP-Ad complexes was performed on Staphylococcus epidermidis (S. epidermidis) strains. The Minimum Inhibitory Concentration (MIC) studies showed that the complex of CIP-Ad and 2,3-O-dimethyl-ß-CD exhibited a similar antimicrobial activity to CIP-Ad alone, while the interaction with 2,3,6-O-trimethyl-ß-CD increased MIC values. Antimicrobial assays on S. epidermidis biofilms demonstrated that the synergistic effect observed with the DspB/CIP association was partly maintained with the 2,3-O-dimethyl-ß-CDs/CIP-Ad complex. To obtain this "all-in-one" drug delivery system, able to destroy the biofilm matrix and release the antibiotic simultaneously, we covalently grafted DspB on three carboxylic permethylated CD derivatives with different-length spacer arms. The strategy was validated by demonstrating that a DspB-permethylated-ß-CD/ciprofloxacin-Ad system exhibited efficient antibiofilm activity.


Asunto(s)
Ciclodextrinas , Ciclodextrinas/química , Ciprofloxacina/farmacología , Ciprofloxacina/química , Antibacterianos/farmacología , Antibacterianos/química , Termodinámica , Staphylococcus epidermidis
3.
J Mol Model ; 29(6): 182, 2023 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-37209272

RESUMEN

CONTEXT: A bacterial biofilm is a cluster of bacterial cells embedded in a self-produced matrix of extracellular polymeric substances such as DNA, proteins, and polysaccharides. Several diseases have been reported to cause by bacterial biofilms, and difficulties in treating these infections are of concern. This work aimed to identify the inhibitor with the highest binding affinity for the receptor protein by screening various inhibitors obtained from Azorella species for a potential target to inhibit dispersin B. This work shows that azorellolide has the highest binding affinity (- 8.2 kcal/mol) among the compounds tested, followed by dyhydroazorellolide, mulinone A, and 7-acetoxy-mulin-9,12-diene which all had a binding affinity of - 8.0 kcal/mol. To the best of our knowledge, this is the first study to evaluate and contrast several diterpene compounds as antibacterial biofilm chemicals. METHODS: Here, molecular modelling techniques tested 49 diterpene compounds of Azorella and six FDA-approved antibiotics medicines for antibiofilm activity. Since protein-like interactions are crucial in drug discovery, AutoDock Vina was initially employed to carry out structure-based virtual screening. The drug-likeness and ADMET properties of the chosen compounds were examined to assess the antibiofilm activity further. Lipinski's rule of five was then applied to determine the antibiofilm activity. Then, molecular electrostatic potential was used to determine the relative polarity of a molecule using the Gaussian 09 package and GaussView 5.08. Following three replica molecular dynamic simulations (using the Schrodinger program, Desmond 2019-4 package) that each lasted 100 ns on the promising candidates, binding free energy was estimated using MM-GBSA. Structural visualisation was used to test the binding affinity of each compound to the crystal structure of dispersin B protein (PDB: 1YHT), a well-known antibiofilm compound.


Asunto(s)
Apiaceae , Diterpenos , Simulación del Acoplamiento Molecular , Apiaceae/química , Simulación de Dinámica Molecular , Antibacterianos/farmacología , Diterpenos/farmacología , Diterpenos/química
4.
FEBS J ; 290(4): 1049-1059, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36083143

RESUMEN

Bacterial biofilms consist of bacterial cells embedded within a self-produced extracellular polymeric substance (EPS) composed of exopolysaccharides, extra cellular DNA, proteins and lipids. The enzyme Dispersin B (DspB) is a CAZy type 20 ß-hexosaminidase enzyme that catalyses the hydrolysis of poly-N-acetylglucosamine (PNAG), a major biofilm polysaccharide produced by a wide variety of biofilm-forming bacteria. Native PNAG is partially de-N-acetylated, and the degree of deacetylation varies between species and dependent on the environment. We have previously shown that DspB is able to perform both endo- and exo-glycosidic bond cleavage of PNAG depending on the de-N-acetylation patterns present in the PNAG substrate. Here, we used a combination of synthetic PNAG substrate analogues, site-directed mutagenesis and in vitro biofilm dispersal assay to investigate the molecular basis for the endo-glycosidic cleavage activity of DspB and the importance of this activity for dispersal of PNAG-dependent Staphylococcus epidermidis biofilms. We found that D242 contributes to the endoglycosidase activity of DspB through electrostatic interactions with cationic substrates in the -2 binding site. A DspBD242N mutant was highly deficient in endoglycosidase activity while maintaining exoglycosidase activity. When used to disperse S. epidermidis biofilms, this DspBD242N mutant resulted in an increase in residual biofilm biomass after treatment when compared to wild-type DspB. These results suggest that the de-N-acetylation of PNAG in S. epidermidis biofilms is not uniformly distributed and that the endoglycosidase activity of DspB is required for efficient biofilm dispersal.


Asunto(s)
Acetilglucosamina , Glicósido Hidrolasas , Glicósido Hidrolasas/química , Proteínas Bacterianas/metabolismo , Matriz Extracelular de Sustancias Poliméricas/metabolismo , Electricidad Estática , Staphylococcus epidermidis/metabolismo , Biopelículas
5.
Methods Enzymol ; 665: 209-231, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35379435

RESUMEN

Bacterial biofilms consist of surface-attached communities that secrete polymeric substances to form a biofilm matrix, generating a local microenvironment which helps protect from external factors. One such matrix component produced by a diverse list of microorganisms is the polysaccharide poly-ß-1,6-N-acetylglucosamine (PNAG). Dispersin B is a PNAG-specific glycosyl hydrolase, which by leveraging its unique specificity, can be used to design a macromolecular fluorescent PNAG binding probe. An active site mutant of Dispersin B was fused to a fluorescent protein, to generate a probe that bound PNAG but did not hydrolyze its polysaccharide target. The ease and versatility of this strategy has made it possible to study PNAG in the context of maturing biofilms, as the probe tends to sequester in regions of high PNAG density. In this chapter, typical workflows from probe construction to cell-binding and imaging experiments are described.


Asunto(s)
Proteínas Bacterianas , Biopelículas , Acetilglucosamina , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Polisacáridos
6.
Appl Biochem Biotechnol ; 194(2): 737-747, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34524634

RESUMEN

Dispersin B (DspB) is a member of glycoside hydrolase family 20 (GH20) and catalyzes degradation of biofilms forming by pathogenic bacteria such as Staphylococcus aureus. Magnetoreceptor (MagR) is a magnetic protein that can be used as a fusion partner for functionally immobilizing proteins on magnetic surfaces. In the present study, a recombinant protein DspB-MagR was constructed by fusing MagR to the C-terminus of DspB and expressed in Escherichia coli. Magnetic immobilization of purified DspB-MagR on magnetic core-shell structured Fe3O4@SiO2 nanoparticles was achieved and characterized by means of various techniques including SDS-PAGE, Fourier transform infrared spectroscopy, thermogravimetric analysis, zeta potential measurement, and scanning electron microscopy. It was evaluated the influence of temperature, pH, and storage time on the performance of immobilized DspB-MagR on Fe3O4@SiO2 nanoparticles. Removal of biofilms forming by Staphylococcus aureus and other medical sourced bacterial species was achieved by using Fe3O4@SiO2 nanoparticles loading with DspB-MagR. This work promoted potential applications of DspB and similar enzymes for medical purposes.


Asunto(s)
Dióxido de Silicio
7.
Artículo en Inglés | MEDLINE | ID: mdl-33468484

RESUMEN

Prosthetic joint infections (PJI) are frequent complications of arthroplasties. Their treatment is made complex by the rapid formation of bacterial biofilms, limiting the effectiveness of antibiotic therapy. In this study, we explore the effect of a tri-enzymatic cocktail (TEC) consisting of an endo-1,4-ß-d-glucanase, a ß-1,6-hexosaminidase, and an RNA/DNA nonspecific endonuclease combined with antibiotics of different classes against biofilms of Staphylococcus aureus, Staphylococcus epidermidis, and Escherichia coli grown on Ti-6Al-4V substrates. Biofilms were grown in Trypticase soy broth (TSB) with 10 g/liter glucose and 20 g/liter NaCl (TGN). Mature biofilms were assigned to a control group or treated with the TEC for 30 min and then either analyzed or reincubated for 24 h in TGN or TGN with antibiotics. The cytotoxicity of the TEC was assayed against MG-63 osteoblasts, primary murine fibroblasts, and J-774 macrophages using the lactate dehydrogenase (LDH) release test. The TEC dispersed 80.3 to 95.2% of the biofilms' biomass after 30 min. The reincubation of the treated biofilms with antibiotics resulted in a synergistic reduction of the total culturable bacterial count (CFU) compared to that of biofilms treated with antibiotics alone in the three tested species (additional reduction from 2 to more than 3 log10 CFU). No toxicity of the TEC was observed against the tested cell lines after 24 h of incubation. The combination of pretreatment with TEC followed by 24 h of incubation with antibiotics had a synergistic effect against biofilms of S. aureus, S. epidermidis, and E. coli Further studies should assess the potential of the TEC as an adjuvant therapy in in vivo models of PJI.


Asunto(s)
Antibacterianos , Infecciones Estafilocócicas , Animales , Antibacterianos/farmacología , Biopelículas , Escherichia coli , Ratones , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus , Staphylococcus epidermidis
8.
J Biol Chem ; 296: 100203, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33334876

RESUMEN

The exopolysaccharide poly-ß-(1→6)-N-acetylglucosamine (PNAG) is a major structural determinant of bacterial biofilms responsible for persistent and nosocomial infections. The enzymatic dispersal of biofilms by PNAG-hydrolyzing glycosidase enzymes, such as Dispersin B (DspB), is a possible approach to treat biofilm-dependent bacterial infections. The cationic charge resulting from partial de-N-acetylation of native PNAG is critical for PNAG-dependent biofilm formation. We recently demonstrated that DspB has increased catalytic activity on de-N-acetylated PNAG oligosaccharides, but the molecular basis for this increased activity is not known. Here, we analyze the role of anionic amino acids surrounding the catalytic pocket of DspB in PNAG substrate recognition and hydrolysis using a combination of site-directed mutagenesis, activity measurements using synthetic PNAG oligosaccharide analogs, and in vitro biofilm dispersal assays. The results of these studies support a model in which bound PNAG is weakly associated with a shallow anionic groove on the DspB protein surface with recognition driven by interactions with the -1 GlcNAc residue in the catalytic pocket. An increased rate of hydrolysis for cationic PNAG was driven, in part, by interaction with D147 on the anionic surface. Moreover, we identified that a DspB mutant with improved hydrolysis of fully acetylated PNAG oligosaccharides correlates with improved in vitro dispersal of PNAG-dependent Staphylococcus epidermidis biofilms. These results provide insight into the mechanism of substrate recognition by DspB and suggest a method to improve DspB biofilm dispersal activity by mutation of the amino acids within the anionic binding surface.


Asunto(s)
Aggregatibacter actinomycetemcomitans/metabolismo , Aminoácidos/metabolismo , Proteínas Bacterianas/metabolismo , Glicósido Hidrolasas/metabolismo , beta-Glucanos/metabolismo , Biopelículas , Hidrólisis , Modelos Moleculares
9.
Artículo en Inglés | MEDLINE | ID: mdl-32671021

RESUMEN

Regenerative medicine has become an extremely valuable tool offering an alternative to conventional therapies for the repair and regeneration of tissues. The re-establishment of tissue and organ functions can be carried out by tissue engineering strategies or by using medical devices such as implants. However, with any material being implanted inside the human body, one of the conundrums that remains is the ease with which these materials can get contaminated by bacteria. Bacterial adhesion leads to the formation of mature, alive and complex three-dimensional biofilm structures, further infection of surrounding tissues and consequent development of complicated chronic infections. Hence, novel tissue engineering strategies delivering biofilm-targeted therapies, while at the same time allowing tissue formation are highly relevant. In this study our aim was to develop surface modified polyhydroxyalkanoate-based fiber meshes with enhanced bacterial anti-adhesive and juvenile biofilm disrupting properties for tissue regeneration purposes. Using reactive and amphiphilic star-shaped macromolecules as an additive to a polyhydroxyalkanoate spinning solution, a synthetic antimicrobial peptide, Amhelin, with strong bactericidal and anti-biofilm properties, and Dispersin B, an enzyme promoting the disruption of exopolysaccharides found in the biofilm matrix, were covalently conjugated to the fibers by addition to the solution before the spinning process. Staphylococcus epidermidis is one of the most problematic pathogens responsible for tissue-related infections. The initial antibacterial screening showed that Amhelin proved to be strongly bactericidal at 12 µg/ml and caused >50% reductions of biofilm formation at 6 µg/ml, while Dispersin B was found to disperse >70% of pre-formed biofilms at 3 µg/ml. Regarding the cytotoxicity of the agents toward L929 murine fibroblasts, a CC50 of 140 and 115 µg/ml was measured for Amhelin and Dispersin B, respectively. Optimization of the electrospinning process resulted in aligned fibers. Surface activated fibers with Amhelin and Dispersin B resulted in 83% reduction of adhered bacteria on the surface of the fibers. Additionally, the materials developed were found to be cytocompatible toward L929 murine fibroblasts. The strategy reported in this preliminary study suggests an alternative approach to prevent bacterial adhesion and, in turn biofilm formation, in materials used in regenerative medicine applications such as tissue engineering.

10.
Int J Biol Macromol ; 118(Pt A): 419-426, 2018 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-29908271

RESUMEN

Dispersin B (DspB) has shown a great potential for the hydrolysis of polymeric ß-1,6-N-acetyl-d-glucosamine (PNAG) to disperse the biofilms formed by various bacteria but with no killing activity. Here we have investigated whether a silver-binding peptide (AgBP) fused to DspB can induce the in situ formation of silver nanoparticles (AgNP) and conjugated to the structure of DspB so that the bacteria cells released from the dispersed biofilm will be killed by the conjugated AgNP. However, the desired conjugate could be obtained because of the silver ions itself was found to precipitate DspB. But, the fusion of AgBP2 to DspB (AgBP2-DspB) could generate at least 2 fold higher activity against soluble substrate 4-nitrophenyl N-acetyl-ß-D-glucosaminide (NP-GlcNAc). By applying to a preformed Staphylococcus epidermidis biofilm, AgBP2-DspB could clear 69% of the biofilm while only 37% could be cleared by DspB as observed by fluorescent microscope. As measured by crystal violet staining, biofilm could be eradicated to the same extent by loading AgBP2-DspB activity level approximately 20 fold lower than that of DspB. The biofilm formation could be prevented on a AgBP2-DspB immobilized surface as observed by confocal laser microscope.


Asunto(s)
Proteínas Bacterianas/farmacología , Biopelículas/efectos de los fármacos , Glicósido Hidrolasas/farmacología , Péptidos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Acetilglucosamina/análogos & derivados , Acetilglucosamina/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Escherichia coli/química , Escherichia coli/genética , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Humanos , Hidrólisis , Nanopartículas del Metal/química , Péptidos/química , Péptidos/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Plata/química , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/crecimiento & desarrollo , Staphylococcus epidermidis/patogenicidad , beta-Glucanos/química
11.
Adv Healthc Mater ; 7(11): e1701059, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29577666

RESUMEN

Bioconjugation of enzymes on coatings based on polydopamine (PDA) layers is an appealing approach to control biological responses on biomedical implant surfaces. As alternative to PDA wet deposition, a fast, solvent-free, and dynamic deposition approach based on atmospheric-pressure plasma dielectric barrier discharge process is considered to deposit on metallic surfaces acrylic-based interlayers containing highly chemically reactive catechol/quinone groups. A biomimetic approach based on covalent immobilization of Dispersin B, an enzyme with antibiofilm properties, shows the bioconjugation potential of the novel plasma polymer layers. The excellent antibiofilm activity against Staphylococcus epidermidis is comparable to the PDA-based layers prepared by wet chemical methods with slow deposition rates. A study of preosteoblastic MG-63 human cell line viability and adhesion properties on plasma polymer layers demonstrates early interaction required for biomedical applications.


Asunto(s)
Benzoquinonas , Biopelículas/efectos de los fármacos , Catecoles , Indoles , Metacrilatos , Gases em Plasma/química , Polímeros , Animales , Benzoquinonas/química , Benzoquinonas/farmacología , Biopelículas/crecimiento & desarrollo , Catecoles/química , Catecoles/farmacología , Línea Celular , Indoles/química , Indoles/farmacología , Ensayo de Materiales , Metacrilatos/química , Metacrilatos/farmacología , Ratones , Polímeros/química , Polímeros/farmacología , Staphylococcus epidermidis/fisiología
12.
ACS Biomater Sci Eng ; 3(12): 3654-3661, 2017 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33445400

RESUMEN

One major factor inhibiting natural wound-healing processes is infection through bacterial biofilms, particularly in the case of chronic wounds. In this study, the micro/nanostructure of a wound dressing was optimized in order to obtain a more efficient antibiofilm protein-release profile for biofilm inhibition and/or detachment. A 3D substrate was developed with asymmetric polyhydroxyalkanoate (PHA) membranes to entrap Dispersin B (DB), the antibiofilm protein. The membranes were prepared using wet-induced phase separation (WIPS). By modulating the concentration and the molecular weight of the porogen polymer, polyvinylpyrrolidone (PVP), asymmetric membranes with controlled porosity were obtained. PVP was added at 10, 30, and 50% w/w, relative to the total polymer concentration. The physical and kinetic properties of the quaternary nonsolvent/solvent/PHA/PVP systems were studied and correlated with the membrane structures obtained. The results show that at high molecular weight (Mw = 360 kDa) and high PVP content (above 30%), pore size decreased and the membrane became extremely brittle with serious loss of physical integrity. This brittle effect was not observed for low molecular weight PVP (Mw = 40 kDa) at comparable contents. Whatever the molecular weight, porogen content up to 30% increased membrane surface porosity and consequently protein uptake. Above 30% porogen content, the pore size and the physical integrity/mechanical robustness both decreased. The PHA membranes were loaded with DB and their antibiofilm activity was evaluated against Staphylococcus epidermidis biofilms. When the bacterial biofilms were exposed to the DB-loaded PHA membrane, up to 33% of the S. epidermidis biofilm formation was inhibited, while 26% of the biofilm already formed was destroyed. These promising results validate our approach based on the development of bioactive-protein-loaded asymmetric membranes for antibiofilm strategies in situations where traditional antibiotic therapies are ineffective.

13.
Microb Drug Resist ; 23(3): 384-390, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27754780

RESUMEN

Staphylococcus aureus in biofilms is highly resistant to the treatment with antibiotics, to which the planktonic cells are susceptible. This is likely to be due to the biofilm creating a protective barrier that prevents antibiotics from accessing the live pathogens buried in the biofilm. S. aureus biofilms consist of an extracellular matrix comprising, but not limited to, extracellular bacterial DNA (eDNA) and poly-ß-1, 6-N-acetyl-d-glucosamine (PNAG). Our study revealed that despite inferiority of dispersin B (an enzyme that degrades PNAG) to DNase I that cleaves eDNA, in dispersing the biofilm of S. aureus, both enzymes were equally efficient in enhancing the antibacterial efficiency of tobramycin, a relatively narrow-spectrum antibiotic against infections caused by gram-positive and gram-negative pathogens, including S. aureus, used in this investigation. However, a combination of these two biofilm-degrading enzymes was found to be significantly less effective in enhancing the antimicrobial efficacy of tobramycin than the individual application of the enzymes. These findings indicate that combinations of different biofilm-degrading enzymes may compromise the antimicrobial efficacy of antibiotics and need to be carefully assessed in vitro before being used for treating medical devices or in pharmaceutical formulations for use in the treatment of chronic ear or respiratory infections.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Biopelículas/efectos de los fármacos , Desoxirribonucleasa I/metabolismo , Staphylococcus aureus/efectos de los fármacos , Tobramicina/farmacología , ADN Bacteriano/genética , Pruebas de Sensibilidad Microbiana/métodos , Staphylococcus aureus/metabolismo
14.
Mol Oral Microbiol ; 31(4): 329-39, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26280561

RESUMEN

Among the various proteins expressed by the periodontopathogen Aggregatibacter actinomycetemcomitans, two proteins play important roles for survival in the oral cavity. The autotransporter Aae facilitates the attachment of the pathogen to oral epithelial cells, which act as a reservoir, while the biofilm-degrading glycoside hydrolase dispersin B facilitates the movement of daughter cells from the mature biofilm to a new site. The objective of this study was to use the potential of these two proteins to control biofilms. To this end, we generated a hybrid construct between the Aae C-terminal translocating domain and dispersin B, and mobilized it into Escherichia coli Rosetta (DE3) pLysS cells. Immunofluorescence analysis of the modified E. coli cells confirmed the presence of dispersin B on the surface. Further, the membrane localization of the displayed dispersin B was confirmed with Western blot analysis. The integrity of the E. coli cells displaying the dispersin B was confirmed through FACS analysis. The hydrolytic activity of the surface-displayed dispersin B was confirmed by using 4-methylumbelliferyl-ß-d-glucopyranoside as the substrate. The detachment ability of the dispersin B surface-displaying E. coli cells was shown using Staphylococcus epidermidis and Actinobacillus pleuropneumoniae biofilms in a microtiter assay. We concluded that the Aae ß-domain is sufficient to translocate foreign enzymes in the native folded form and that the method of Aae-mediated translocation of surface displayed enzymes might be useful for control of biofilms.


Asunto(s)
Aggregatibacter actinomycetemcomitans/fisiología , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Escherichia coli/genética , Glicósido Hidrolasas/metabolismo , Sistemas de Secreción Tipo V/metabolismo , Actinobacillus pleuropneumoniae/fisiología , Aggregatibacter actinomycetemcomitans/enzimología , Aggregatibacter actinomycetemcomitans/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Técnicas de Visualización de Superficie Celular , Escherichia coli/química , Escherichia coli/metabolismo , Citometría de Flujo , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Staphylococcus epidermidis/fisiología , Sistemas de Secreción Tipo V/química , Sistemas de Secreción Tipo V/genética
15.
Colloids Surf B Biointerfaces ; 136: 56-63, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26363267

RESUMEN

It is known for roughly a decade that bacterial communities (called biofilms) are responsible for significant enhanced antibiotherapy resistance. Biofilms are involved in tissue persistent infection, causing direct or collateral damage leading to chronic wounds development and impairing natural wound healing. In this study, we are interested in the development of supported protein materials which consist of asymmetric membranes as reservoir supports for the incorporation and controlled release of biomolecules capable of dissolving biofilms (or preventing their formation) and their use as wound dressing for chronic wound treatment. In a first step, polyhydroxyalkanoates (PHAs) asymmetric membranes were prepared using wet phase inversion technique. Scanning microscopy (SEM) analysis has showed the influence of different processing parameters. In a second step, the porous side of the membranes were functionalized with a surface treatment and then loaded with the antibiofilm agent (dispersin B). In a third step, the properties and antibiofilm performance of the loaded-membranes were evaluated. Exposure of Staphylococcus epidermidis biofilms to such systems weakly inhibited biofilm formation (weak preventive effect) but caused their detachment and disaggregation (strong curative effect). These initial results are promising since they open the way to a new generation of effective tools in the struggle against persistent bacterial infections exhibiting enhanced antibiotherapy resistance, and in particular in the case of infected chronic wounds.


Asunto(s)
Biopelículas , Materiales Biocompatibles , Membranas Artificiales , Microscopía Electrónica de Rastreo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...