Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Tipo de estudio
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.
PLoS One ; 15(2): e0228591, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32023304

RESUMEN

Biofilms are currently considered as a predominant lifestyle of many bacteria in nature. While they promote survival of microbes, biofilms also potentially increase the threats to animal and public health in case of pathogenic species. They not only facilitate bacteria transmission and persistence, but also promote spreading of antibiotic resistance leading to chronic infections. In the case of Francisella tularensis, the causative agent of tularemia, biofilms have remained largely enigmatic. Here, applying live and static confocal microscopy, we report growth and ultrastructural organization of the biofilms formed in vitro by these microorganisms over the early transition from coccobacillary into coccoid shape during biofilm assembly. Using selective dispersing agents, we provided evidence for extracellular DNA (eDNA) being a major and conserved structural component of mature biofilms formed by both F. subsp. novicida and a human clinical isolate of F. philomiragia. We also observed a higher physical robustness of F. novicida biofilm as compared to F. philomiragia one, a feature likely promoted by specific polysaccharides. Further, F. novicida biofilms resisted significantly better to ciprofloxacin than their planktonic counterparts. Importantly, when grown in biofilms, both Francisella species survived longer in cold water as compared to free-living bacteria, a trait possibly associated with a gain in fitness in the natural aquatic environment. Overall, this study provides information on survival of Francisella when embedded with biofilms that should improve both the future management of biofilm-related infections and the design of effective strategies to tackle down the problematic issue of bacteria persistence in aquatic ecosystems.


Asunto(s)
Biopelículas , Farmacorresistencia Bacteriana , Francisella/fisiología , Agua Dulce/microbiología , Adaptación Fisiológica , Antibacterianos/farmacología , Ciprofloxacina/farmacología , Secuencia Conservada , ADN Bacteriano/química , Francisella/efectos de los fármacos , Francisella/genética , Francisella/patogenicidad , Infecciones por Bacterias Gramnegativas/microbiología , Humanos
3.
J Biomed Mater Res A ; 105(8): 2171-2181, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28380658

RESUMEN

P15-CSP is a biomimetic cationic fusion peptide that stimulates osteogenesis and inhibits bacterial biofilm formation when coated on 2-D surfaces. This study tested the hypothesis that P15-CSP coatings enhance 3-D osteogenesis in a porous but otherwise hydrophobic poly-(ɛ-caprolactone) (PCL) scaffold. Scaffolds of 84 µm and 141 µm average pore size were coated or not with Layer-by-Layer polyelectrolytes followed by P15-CSP, seeded with adult primary human mesenchymal stem cells (MSCs), and cultured 10 days in proliferation medium, then 21 days in osteogenic medium. Atomic analyses showed that P15-CSP was successfully captured by LbL. After 2 days of culture, MSCs adhered and spread more on P15-CSP coated pores than PCL-only. At day 10, all constructs contained nonmineralized tissue. At day 31, all constructs became enveloped in a "skin" of tissue that, like 2-D cultures, underwent sporadic mineralization in areas of high cell density that extended into some 141 µm edge pores. By quantitative histomorphometry, 2.5-fold more tissue and biomineral accumulated in edge pores versus inner pores. P15-CSP specifically promoted tissue-scaffold integration, fourfold higher overall biomineralization, and more mineral deposits in the outer 84 µm and inner 141 µm pores than PCL-only (p < 0.05). 3-D Micro-CT revealed asymmetric mineral deposition consistent with histological calcium staining. This study provides proof-of-concept that P15-CSP coatings are osteoconductive in PCL pore surfaces with 3-D topography. Biomineralization deeper than 150 µm from the scaffold edge was optimally attained with the larger 141 µm peptide-coated pores. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2171-2181, 2017.


Asunto(s)
Materiales Biocompatibles Revestidos/química , Colágeno/química , Células Madre Mesenquimatosas/citología , Osteogénesis , Fragmentos de Péptidos/química , Poliésteres/química , Andamios del Tejido/química , Adulto , Diferenciación Celular , Células Cultivadas , Humanos , Porosidad , Ingeniería de Tejidos
4.
J Biomed Mater Res A ; 103(12): 3736-46, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26097095

RESUMEN

In the context of porous bone void filler for oral bone reconstruction, peptides that suppress microbial growth and promote osteoblast function could be used to enhance the performance of a porous bone void filler. We tested the hypothesis that P15-CSP, a novel fusion peptide containing collagen-mimetic osteogenic peptide P15, and competence-stimulating peptide (CSP), a cationic antimicrobial peptide, has emerging properties not shared by P15 or CSP alone. Peptide-coated surfaces were tested for antimicrobial activity toward Streptoccocus mutans, and their ability to promote human mesenchymal stem cell (MSC) attachment, spreading, metabolism, and osteogenesis. In the osteogenesis assay, peptides were coated on tissue culture plastic and on thin films generated by plasma-enhanced chemical vapor deposition to have hydrophilic or hydrophobic character (water contact angles 63°, 42°, and 92°, respectively). S. mutans planktonic growth was specifically inhibited by CSP, whereas biofilm formation was inhibited by P15-CSP. MSC adhesion and actin stress fiber formation was strongly enhanced by CSP, P15-CSP, and fibronectin coatings and modestly enhanced by P15 versus uncoated surfaces. Metabolic assays revealed that CSP was slightly cytotoxic to MSCs. MSCs developed alkaline phosphatase activity on all surfaces, with or without peptide coatings, and consistently deposited the most biomineralized matrix on hydrophilic surfaces coated with P15-CSP. Hydrophobic thin films completely suppressed MSC biomineralization, consistent with previous findings of suppressed osteogenesis on hydrophobic bioplastics. Collective data in this study provide new evidence that P15-CSP has unique dual capacity to suppress biofilm formation, and to enhance osteogenic activity as a coating on hydrophilic surfaces.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Biopelículas/efectos de los fármacos , Materiales Biocompatibles Revestidos/química , Colágeno/farmacología , Osteogénesis/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Streptococcus/efectos de los fármacos , Secuencia de Aminoácidos , Péptidos Catiónicos Antimicrobianos/administración & dosificación , Péptidos Catiónicos Antimicrobianos/química , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/química , Proteínas Bacterianas/farmacología , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Colágeno/administración & dosificación , Colágeno/química , Proteínas de Unión al ADN/administración & dosificación , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/farmacología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Datos de Secuencia Molecular , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/farmacología , Infecciones Estreptocócicas/prevención & control , Streptococcus/fisiología , Humectabilidad
5.
Microbiol Insights ; 7: 9-13, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24826078

RESUMEN

Chronic wounds including diabetic foot ulcers, pressure ulcers, and venous leg ulcers are a worldwide health problem. As the traditional methods of treatment have proven ineffective against chronic wounds involving biofilms, there is an unmet clinical need for developing products with an antibiofilm component that inhibits and/or disrupts biofilms and thus make the biofilm-embedded bacteria more susceptible to antimicrobial therapy. We developed a DispersinB® antibiofilm enzyme-based wound spray for treating chronic wounds in conjunction with an antimicrobial. Under in vitro conditions, the DispersinB® and Acticoat™ combination performed significantly better (P < 0.05) than Acticoat™ alone, indicating the synergy between the two compounds because of DispersinB® enhancing the antimicrobial activity of Acticoat™. Furthermore, DispersinB® wound spray enhanced the antimicrobial activity of Acticoat™ in a chronic wound mouse model of methicillin-resistant Staphylococcus aureus (MRSA) infection. Thus, this novel combination of DispersinB® and Acticoat™, an antimicrobial dressing, prompts clinical evaluation for potential applications in biofilm-based chronic wound management.

6.
ACS Appl Mater Interfaces ; 4(9): 4708-16, 2012 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-22909396

RESUMEN

We developed a highly efficient, biocompatible surface coating that disperses bacterial biofilms through enzymatic cleavage of the extracellular biofilm matrix. The coating was fabricated by binding the naturally existing enzyme dispersin B (DspB) to surface-attached polymer matrices constructed via a layer-by-layer (LbL) deposition technique. LbL matrices were assembled through electrostatic interactions of poly(allylamine hydrochloride) (PAH) and poly(methacrylic acid) (PMAA), followed by chemical cross-linking with glutaraldehyde and pH-triggered removal of PMAA, producing a stable PAH hydrogel matrix used for DspB loading. The amount of DspB loaded increased linearly with the number of PAH layers in surface hydrogels. DspB was retained within these coatings in the pH range from 4 to 7.5. DspB-loaded coatings inhibited biofilm formation by two clinical strains of Staphylococcus epidermidis. Biofilm inhibition was ≥98% compared to mock-loaded coatings as determined by CFU enumeration. In addition, DspB-loaded coatings did not inhibit attachment or growth of cultured human osteoblast cells. We suggest that the use of DspB-loaded multilayer coatings presents a promising method for creating biocompatible surfaces with high antibiofilm efficiency, especially when combined with conventional antimicrobial treatment of dispersed bacteria.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Biopelículas/efectos de los fármacos , Materiales Biocompatibles Revestidos/farmacología , Glicósido Hidrolasas/metabolismo , Antibacterianos/química , Antibacterianos/toxicidad , Proteínas Bacterianas/química , Diferenciación Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/toxicidad , Glicósido Hidrolasas/química , Humanos , Hidrogeles/química , Ácidos Polimetacrílicos/química , Staphylococcus epidermidis/fisiología , Propiedades de Superficie
7.
Appl Environ Microbiol ; 77(23): 8303-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21984237

RESUMEN

We demonstrated the production of poly-ß-1,6-N-acetylglucosamine (PNAG) polysaccharide in the biofilms of Burkholderia multivorans, Burkholderia vietnamiensis, Burkholderia ambifaria, Burkholderia cepacia, and Burkholderia cenocepacia using an immunoblot assay for PNAG. These results were confirmed by further studies, which showed that the PNAG hydrolase, dispersin B, eliminated immunoreactivity of extracts from the species that were tested (B. cenocepacia and B. multivorans). Dispersin B also inhibited biofilm formation and dispersed preformed biofilms of Burkholderia species. These results imply a role for PNAG in the maintenance of Burkholderia biofilm integrity. While PNAG was present in biofilms of all of the wild-type test organisms, a ΔpgaBC mutant of B. multivorans (Mu5) produced no detectable PNAG, indicating that these genes are needed for Burkholderia PNAG formation. Furthermore, restoration of PNAG production in PNAG negative E. coli TRXWMGΔC (ΔpgaC) by complementation with B. multivorans pgaBCD confirmed the involvement of these genes in Burkholderia PNAG production. While the confocal scanning laser microscopy of untreated wild-type B. multivorans showed thick, multilayered biofilm, Mu5 and dispersin B-treated wild-type biofilms were thin, poorly developed, and disrupted, confirming the involvement of PNAG in B. multivorans biofilm formation. Thus, PNAG appears to be an important component of Burkholderia biofilms, potentially contributing to its resistance to multiple antibiotics and persistence during chronic infections, including cystic fibrosis-associated infection.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Burkholderia/fisiología , beta-Glucanos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Burkholderia/crecimiento & desarrollo , Burkholderia/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Eliminación de Gen , Prueba de Complementación Genética , Glicósido Hidrolasas/metabolismo , Immunoblotting , Microscopía Confocal
8.
Plant Biotechnol J ; 7(7): 631-44, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19702755

RESUMEN

Nearly one-third of the world's population, mostly women and children, suffer from iron malnutrition and its consequences, such as anaemia or impaired mental development. Iron fortification of food is difficult because soluble iron is either unstable or unpalatable, and non-soluble iron is not bioavailable. Genetic engineering of crop plants to increase iron content has therefore emerged as an alternative for iron biofortification. To date, strategies to increase iron content have relied on single genes, with limited success. Our work focuses on rice as a model plant, because it feeds one-half of the world's population, including the majority of the iron-malnourished population. Using the targeted expression of two transgenes, nicotianamine synthase and ferritin, we increased the iron content of rice endosperm by more than six-fold. Analysis of transgenic rice lines confirmed that, in combination, they provide a synergistic effect on iron uptake and storage. Laser ablation-inductively coupled plasma-mass spectrometry showed that the iron in the endosperm of the transgenic rice lines accumulated in spots, most probably as a consequence of spatially restricted ferritin accumulation. Agronomic evaluation of the high-iron rice lines did not reveal a yield penalty or significant changes in trait characters, except for a tendency to earlier flowering. Overall, we have demonstrated that rice can be engineered with a small number of genes to achieve iron biofortification at a dietary significant level.


Asunto(s)
Transferasas Alquil y Aril/metabolismo , Ferritinas/metabolismo , Hierro/metabolismo , Oryza/enzimología , Oryza/metabolismo , Plantas Modificadas Genéticamente/metabolismo , Transferasas Alquil y Aril/genética , Ferritinas/genética , Oryza/genética , Plantas Modificadas Genéticamente/enzimología , Plantas Modificadas Genéticamente/genética , Semillas/genética , Semillas/metabolismo
9.
J Ind Microbiol Biotechnol ; 36(10): 1297-305, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19597857

RESUMEN

beta-1,6-N-Acetylglucosaminidase (dispersin B), which cleaves poly-ss-(1,6)-linked N-acetylglucosamine, is encoded by dspB of Aggregatibacter actinomycetemcomitans. To enhance the production of dispersin B, we engineered dspB to transcribe mRNAs devoid of the trinucleotide ACA. Transcription and translation levels of ACA-less and wild-type dspB expressed in Escherichia coli (E. coli) under T5 and T7 promoters were analyzed by real-time RT-PCR and protein quantification, respectively. The ACA-less dspB mRNA level was significantly higher (P < 0.01) and produced 77.6 and 34.9% more dispersin B than wild-type dspB expressed under T7 and T5 promoters, respectively. Dispersin B expression under T7 promoter caused a 98-99.5% drop in the glyceraldehyde-3-phosphate dehydrogenase (gapA) mRNA level, which was not observed with T5 promoter. Fusion of green fluorescent protein (GFP) with dispersin B allowed rapid quantification of dispersin B production by measuring fluorescence intensity in culture broth. Although the cultures containing 0.1% glucose showed sustained increase in dispersin B-GFP production until 12 h, no significant increase in dispersin B activity was observed beyond 4 and 6 h after induction when expressed under T7 and T5 promoters, respectively. This study demonstrates the effectiveness of ACA-less mRNA and the advantage of GFP tagging for enhanced dispersin B production and quantification, which could be adapted for improving the production of other commercially important proteins in E. coli.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Escherichia coli/enzimología , Escherichia coli/genética , Glicósido Hidrolasas/biosíntesis , Glicósido Hidrolasas/genética , Fusión Artificial Génica , Bacteriófago T7/genética , Secuencia de Bases , Perfilación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Datos de Secuencia Molecular , Mutación , Nucleótidos/genética , Regiones Promotoras Genéticas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Siphoviridae/genética , Transcripción Genética
10.
Antimicrob Agents Chemother ; 50(5): 1835-40, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16641457

RESUMEN

The colonization of uropathogenic bacteria on urinary catheters resulting in biofilm formation frequently leads to the infection of surrounding tissue and often requires removal of the catheter. Infections associated with biofilms are difficult to treat since they may be more than 1,000 times more resistant to antibiotics than their planktonic counterparts. We have developed an antibiofilm composition comprising an N-acetyl-D-glucosamine-1-phosphate acetyltransferase (GlmU) inhibitor and protamine sulfate, a cationic polypeptide. The antibiofilm activity of GlmU inhibitors, such as iodoacetamide (IDA), N-ethyl maleimide (NEM), and NEM analogs, including N-phenyl maleimide, N,N'-(1,2-phenylene)dimaleimide (oPDM), and N-(1-pyrenyl)maleimide (PyrM), was tested against that of catheter-associated uropathogens. Both IDA and NEM inhibited biofilm formation in Escherichia coli. All NEM analogs showed antibiofilm activity against clinical isolates of E. coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, Staphylococcus epidermidis, and Enterococcus faecalis. The combination of oPDM with protamine sulfate (PS) enhanced its antibiofilm activity and reduced its effective concentration to as low as 12.5 microM. In addition, we found that the in vitro inhibitory activity of oPDM-plus-PS-coated silicone catheters against P. aeruginosa and S. epidermidis colonization was superior to that of catheters coated with silver hydrogel. Confocal scanning laser microscopy further confirmed that the oPDM-plus-PS-coated silicone catheters were almost free from bacterial colonization. Thus, a broad-spectrum antibiofilm composition comprising a GlmU inhibitor and protamine sulfate shows promise for use in anti-infective coatings for medical devices, including urinary catheters.


Asunto(s)
Biopelículas , Catéteres de Permanencia/microbiología , Inhibidores Enzimáticos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Infecciones Urinarias/tratamiento farmacológico , Acetiltransferasas/antagonistas & inhibidores , Péptidos Catiónicos Antimicrobianos/farmacología , Bacterias Gramnegativas/aislamiento & purificación , Bacterias Grampositivas/aislamiento & purificación , Humanos , Pruebas de Sensibilidad Microbiana , Microscopía Confocal , Protaminas/farmacología , Cateterismo Urinario/efectos adversos , Infecciones Urinarias/microbiología
11.
Plant Sci ; 171(4): 488-96, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25193646

RESUMEN

Callus is an important intermediate tissue for the propagation of plants through tissue culture, as it can form new shoots, and hence plants, through somatic embryogenesis, a process whose underlying biochemical mechanisms are poorly understood. The involvement of kinases suggests that signalling through phosphorylation cascades may play an important role. In this report methods for labelling wheat (Triticum aestivum L.) callus phosphoproteins in vivo through the uptake of [(32)P]orthophosphate and subsequently revealing the phosphoproteome of this tissue by two-dimensional electrophoresis and autoradiography are described. Labelled proteins from 2D gels were identified by tandem mass spectrometry analysis using the 'Mascot' search engine, and by de novo sequencing in combination with BLAST and MS-BLAST, using a cross-species protein identification approach. Eight putative phosphoproteins from 10 spots were identified, confirming the potential utility of this method; three of these have not previously shown to be phosphorylated. Furthermore, the phosphorylation site for one of the proteins was identified. Protein synthesis functions were tentatively assigned to five of the phosphoproteins, one reactive oxygen species (ROS)-detoxifying enzyme ascorbate peroxidase, and two could not be identified reliably from current database matches.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA