Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Appl Environ Microbiol ; 83(12)2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28411222

RESUMO

The prevalence and structure of Staphylococcus aureus and Staphylococcus epidermidis within multispecies biofilms were found to depend sensitively on physical environment and antibiotic dosage. Although these species commonly infect similar sites, such as orthopedic implants, little is known about their behavior in multispecies communities, particularly in response to treatment. This research establishes that S. aureus is much more prevalent than S. epidermidis when simultaneously seeded and grown under unstressed conditions (pH 7, 37°C) in both laboratory and clinical strains. In multispecies communities, S. epidermidis is capable of growing a more confluent biofilm when the addition of S. aureus is delayed 4 to 6 h during 18 h of growth. Different vancomycin dosages generate various behaviors: S. epidermidis is more prevalent at a dose of 1.0 µg/ml vancomycin, but reduced growth of both species occurs at 1.9 µg/ml vancomycin. This variability is consistent with the different MICs of S. aureus and S. epidermidis Growth at higher temperature (45°C) results in an environment where S. aureus forms porous biofilms. This porosity allows S. epidermidis to colonize more of the surface, resulting in detectable S. epidermidis biomass. Variations in pH result in increased prevalence of S. epidermidis at low pH (pH 5 and 6), while S. aureus remains dominant at high pH (pH 8 and 9). This work establishes the structural variability of multispecies staphylococcal biofilms as they undergo physical and antimicrobial treatments. It provides a basis for understanding the structure of these communities at infection sites and how treatments disrupt their multispecies behaviors.IMPORTANCEStaphylococcus aureus and Staphylococcus epidermidis are two species of bacteria that are commonly responsible for biofilm infections on medical devices. Biofilms are structured communities of bacteria surrounded by polysaccharides, proteins, and DNA; bacteria are more resistant to antimicrobials as part of a biofilm than as individual cells. This work investigates the structure and prevalence of these two organisms when grown together in multispecies biofilms and shows shifts in the behavior of the polymicrobial community when grown in various concentrations of vancomycin (an antibiotic commonly used to treat staphylococcal infections), in a high-temperature environment (a condition previously shown to lead to cell disruption and death), and at low and high pH (a change that has been previously shown to soften the mechanical properties of staphylococcal biofilms). These shifts in community structure demonstrate the effect such treatments may have on multispecies staphylococcal infections.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus epidermidis/efeitos dos fármacos , Humanos , Testes de Sensibilidade Microbiana , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/fisiologia , Staphylococcus epidermidis/crescimento & desenvolvimento , Staphylococcus epidermidis/fisiologia
2.
Mol Microbiol ; 99(1): 123-34, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26365835

RESUMO

Persistent staphylococcal infections often involve surface-associated communities called biofilms. Staphylococcus aureus biofilm development is mediated by the co-ordinated production of the biofilm matrix, which can be composed of polysaccharides, extracellular DNA (eDNA) and proteins including amyloid fibers. The nature of the interactions between matrix components, and how these interactions contribute to the formation of matrix, remain unclear. Here we show that the presence of eDNA in S. aureus biofilms promotes the formation of amyloid fibers. Conditions or mutants that do not generate eDNA result in lack of amyloids during biofilm growth despite the amyloidogeneic subunits, phenol soluble modulin peptides, being produced. In vitro studies revealed that the presence of DNA promotes amyloid formation by PSM peptides. Thus, this work exposes a previously unacknowledged interaction between biofilm matrix components that furthers our understanding of functional amyloid formation and S. aureus biofilm biology.


Assuntos
Amiloide/metabolismo , Toxinas Bacterianas/metabolismo , Biofilmes/crescimento & desenvolvimento , DNA Bacteriano/metabolismo , Staphylococcus aureus/fisiologia
3.
Curr Genet ; 62(1): 137-41, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26515441

RESUMO

Bacterial cells are most often found in the form of multicellular aggregates commonly referred to as biofilms. Biofilms offer their member cells several benefits, such as resistance to killing by antimicrobials and predation. During biofilm formation there is a production of extracellular substances that, upon assembly, constitute an extracellular matrix. The ability to generate a matrix encasing the microbial cells is a common feature of biofilms, but there is diversity in matrix composition and in interaction between matrix components. The different components of bacterial biofilm extracellular matrixes, known as matrix interactions, and resulting implications are discussed in this review.


Assuntos
Biofilmes , Matriz Extracelular/metabolismo , Proteínas de Bactérias/metabolismo , DNA Bacteriano , Espaço Extracelular , Genoma Bacteriano , Polissacarídeos Bacterianos/metabolismo , Staphylococcus aureus/fisiologia
5.
mBio ; 6(4): e00705, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-26173697

RESUMO

UNLABELLED: Bacterial infection can trigger cellular stress programs, such as the unfolded protein response (UPR), which occurs when misfolded proteins accumulate within the endoplasmic reticulum (ER). Here, we used the human pathogen methicillin-resistant Staphylococcus aureus (MRSA) as an infection model to probe how ER stress promotes antimicrobial function. MRSA infection activated the most highly conserved unfolded protein response sensor, inositol-requiring enzyme 1α (IRE1α), which was necessary for robust bacterial killing in vitro and in vivo. The macrophage IRE1-dependent bactericidal activity required reactive oxygen species (ROS). Viable MRSA cells excluded ROS from the nascent phagosome and strongly triggered IRE1 activation, leading to sustained generation of ROS that were largely Nox2 independent. In contrast, dead MRSA showed early colocalization with ROS but was a poor activator of IRE1 and did not trigger sustained ROS generation. The global ROS stimulated by IRE1 signaling was necessary, but not sufficient, for MRSA killing, which also required the ER resident SNARE Sec22B for accumulation of ROS in the phagosomal compartment. Taken together, these results suggest that IRE1-mediated persistent ROS generation might act as a fail-safe mechanism to kill bacterial pathogens that evade the initial macrophage oxidative burst. IMPORTANCE: Cellular stress programs have been implicated as important components of the innate immune response to infection. The role of the IRE1 pathway of the ER stress response in immune secretory functions, such as antibody production, is well established, but its contribution to innate immunity is less well defined. Here, we show that infection of macrophages with viable MRSA induces IRE1 activation, leading to bacterial killing. IRE1-dependent bactericidal activity required generation of reactive oxygen species in a sustained manner over hours of infection. The SNARE protein Sec22B, which was previously demonstrated to control ER-phagosome trafficking, was dispensable for IRE1-driven global ROS production but necessary for late ROS accumulation in bacteria-containing phagosomes. Our study highlights a key role for IRE1 in promoting macrophage bactericidal capacity and reveals a fail-safe mechanism that leads to the concentration of antimicrobial effector molecules in the macrophage phagosome.


Assuntos
Endorribonucleases/metabolismo , Interações Hospedeiro-Patógeno , Macrófagos/imunologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/imunologia , Viabilidade Microbiana/efeitos dos fármacos , Oxidantes/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Linhagem Celular , Estresse do Retículo Endoplasmático , Perfilação da Expressão Gênica , Humanos , Macrófagos/microbiologia , Staphylococcus aureus Resistente à Meticilina/fisiologia , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Espécies Reativas de Oxigênio/metabolismo , Análise de Sequência de DNA
6.
Infect Immun ; 83(9): 3428-37, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26077761

RESUMO

Staphylococcus aureus is a human commensal that colonizes the skin. While it is normally innocuous, it has strong associations with atopic dermatitis pathogenesis and has become the leading cause of skin and soft tissue infections in the United States. The factors that dictate the role of S. aureus in disease are still being determined. In this work, we utilized primary keratinocyte culture and an epidermal murine colonization model to investigate the role of S. aureus phenol-soluble modulins (PSMs) in proinflammatory cytokine release and inflammation induction. We demonstrated that many species of Staphylococcus are capable of causing release of interleukin 18 (IL-18) from keratinocytes and that S. aureus PSMs are necessary and sufficient to stimulate IL-18 release from keratinocytes independently of caspase 1. Further, after 7 days of epicutaneous exposure to wild-type S. aureus, but not S. aureus Δpsm, we saw dramatic changes in gross pathology, as well as systemic release of proinflammatory cytokines. This work demonstrates the importance of PSM peptides in S. aureus-mediated inflammatory cytokine release from keratinocytes in vitro and in vivo and further implicates PSMs as important contributors to pathogenesis.


Assuntos
Toxinas Bacterianas/imunologia , Interleucina-18/biossíntese , Queratinócitos/imunologia , Infecções Cutâneas Estafilocócicas/imunologia , Animais , Toxinas Bacterianas/metabolismo , Linhagem Celular , Citocinas/biossíntese , Dermatite/imunologia , Dermatite/metabolismo , Dermatite/microbiologia , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Humanos , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real , Staphylococcus aureus/imunologia
7.
Antimicrob Agents Chemother ; 59(8): 4497-503, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25987623

RESUMO

Annually, medical device infections are associated with >250,000 catheter-associated bloodstream infections (CLABSI), with up to 25% mortality. Staphylococcus aureus, a primary pathogen in these infections, is capable of biofilm production, allowing organism persistence in harsh environments, offering antimicrobial protection. With increases in S. aureus isolates with reduced susceptibility to current agents, ceftaroline (CPT) offers a therapeutic alternative. Therefore, we evaluated whether CPT would have a role against biofilm-producing methicillin-resistant S. aureus (MRSA), including those with decreased susceptibilities to alternative agents. In this study, we investigated CPT activity alone or combined with daptomycin (DAP) or rifampin (RIF) against 3 clinical biofilm-producing MRSA strains in an in vitro biofilm pharmacokinetic/pharmacodynamic (PK/PD) model. Simulated antimicrobial regimens were as follows: 600 mg of CPT every 8 h (q8h) (free maximum concentration of drug [fCmax], 17.04 mg/liter; elimination half-life [t1/2], 2.66 h), 12 mg/kg of body weight/day of DAP (fCmax, 14.7 mg/liter; t1/2, 8 h), and 450 mg of RIF q12h (fCmax, 3.5 mg/liter; t1/2, 3.4 h), CPT plus DAP, and CPT plus RIF. Samples were obtained and plated to determine colony counts. Differences in log10 CFU/cm(2) were evaluated by analysis of variance with Tukey's post hoc test. The strains were CPT and vancomycin susceptible and DAP nonsusceptible (DNS). CPT displayed activity throughout the experiment. DAP demonstrated initial activity with regrowth at 24 h in all strains. RIF was comparable to the drug-free control, and little benefit was observed when combined with CPT. CPT plus DAP displayed potent activity, with an average log10 CFU/cm(2) reduction of 3.33 ± 1.01 from baseline. CPT demonstrated activity against biofilm-producing DNS MRSA. CPT plus DAP displayed therapeutic enhancement over monotherapy, providing a potential option for difficult-to-treat medical device infections.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Cefalosporinas/farmacologia , Daptomicina/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Meticilina/farmacologia , Vancomicina/farmacologia , Antibacterianos/farmacocinética , Cefalosporinas/farmacocinética , Daptomicina/farmacocinética , Quimioterapia Combinada/métodos , Meia-Vida , Testes de Sensibilidade Microbiana/métodos , Rifampina/farmacocinética , Rifampina/farmacologia , Vancomicina/farmacocinética , Ceftarolina
8.
Appl Environ Microbiol ; 80(22): 7079-87, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25192998

RESUMO

Enteric bacteria, such as Escherichia coli, are exposed to a variety of stresses in the nonhost environment. The development of biofilms provides E. coli with resistance to environmental insults, such as desiccation and bleach. We found that biofilm formation, specifically production of the matrix components curli and cellulose, protected E. coli against killing by the soil-dwelling nematode Caenorhabditis elegans and the predatory bacterium Myxococcus xanthus. Additionally, matrix-encased bacteria at the air-biofilm interface exhibited ∼40-fold-increased survival after C. elegans and M. xanthus killing compared to the non-matrix-encased cells that populate the interior of the biofilm. To determine if nonhost Enterobacteriaceae reservoirs supported biofilm formation, we grew E. coli on media composed of pig dung or commonly contaminated foods, such as beef, chicken, and spinach. Each of these medium types provided a nutritional environment that supported matrix production and biofilm formation. Altogether, we showed that common, nonhost reservoirs of E. coli supported the formation of biofilms that subsequently protected E. coli against predation.


Assuntos
Biofilmes , Caenorhabditis elegans/fisiologia , Escherichia coli/fisiologia , Carne/microbiologia , Myxococcus xanthus/fisiologia , Verduras/microbiologia , Animais , Bovinos , Galinhas , Contaminação de Alimentos/análise , Suínos
9.
Front Microbiol ; 5: 401, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25136340

RESUMO

Many bacteria produce cytolytic toxins that target host cells or other competing microbes. It is well known that environmental factors control toxin expression, however, recent work suggests that some bacteria manipulate the fold of these protein toxins to control their function. The ß-sheet rich amyloid fold is a highly stable ordered aggregate that many toxins form in response to specific environmental conditions. When in the amyloid state, toxins become inert, losing the cytolytic activity they display in the soluble form. Emerging evidence suggest that some amyloids function as toxin storage systems until they are again needed, while other bacteria utilize amyloids as a structural matrix component of biofilms. This amyloid matrix component facilitates resistance to biofilm disruptive challenges. The bacterial amyloids discussed in this review reveal an elegant system where changes in protein fold and solubility dictate the function of proteins in response to the environment.

10.
Biomacromolecules ; 15(8): 2933-43, 2014 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-25010735

RESUMO

The in vitro and in vivo antimicrobial activity of primary ammonium ethyl methacrylate homopolymers (AEMPs) was investigated. AEMPs with different degrees of polymerization (DP = 7.7-12) were prepared by reversible addition-fragmentation chain-transfer (RAFT) polymerization. The AEMPs showed higher inhibitory effects against Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA), than Gram-negative bacteria. The AEMPs also showed potent anti-S. aureus activity in the presence of fetal bovine serum, whereas the activity of the antibiotic mupirocin was reduced under the same conditions. The AEMPs showed very little or no hemolytic activity. The cytotoxicity of AEMPs against mammalian cells HEp-2 and COS-7 was concentration-dependent, and the cell viability significantly decreased at higher polymer concentrations. The AEMPs significantly reduced the number of viable S. aureus cells in the nasal environment of cotton rats when compared to that of the control. This study demonstrates that AEMPs have potential for use in treating topical S. aureus infections.


Assuntos
Antibacterianos , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Doenças Nasais/tratamento farmacológico , Ácidos Polimetacrílicos , Infecções Estafilocócicas/tratamento farmacológico , Administração Tópica , Animais , Antibacterianos/síntese química , Antibacterianos/química , Antibacterianos/farmacologia , Células COS , Bovinos , Chlorocebus aethiops , Bactérias Gram-Negativas/crescimento & desenvolvimento , Humanos , Mupirocina/química , Mupirocina/farmacologia , Doenças Nasais/microbiologia , Ácidos Polimetacrílicos/síntese química , Ácidos Polimetacrílicos/química , Ácidos Polimetacrílicos/farmacologia , Ratos , Sigmodontinae
11.
Infect Immun ; 82(9): 3837-44, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24980969

RESUMO

Staphylococcus aureus virulence is coordinated through the Agr quorum-sensing system to produce an array of secreted molecules. One important class of secreted virulence factors is the phenol-soluble modulins (PSMs). PSMs are small-peptide toxins that have recently been characterized for their roles in infection, biofilm development, and subversion of the host immune system. In this work, we demonstrate that the signal peptide of the S. aureus quorum-sensing signal, AgrD, shares structural and functional similarities with the PSM family of toxins. The efficacy of this peptide (termed N-AgrD) beyond AgrD propeptide trafficking has never been described before. We observe that N-AgrD, like the PSMs, is found in the amyloid fibrils of S. aureus biofilms and is capable of forming and seeding amyloid fibrils in vitro. N-AgrD displays cytolytic and proinflammatory properties that are abrogated after fibril formation. These data suggest that the N-AgrD leader peptide affects S. aureus biology in a manner similar to that described previously for the PSM peptide toxins. Taken together, our findings suggest that peptide cleavage products can affect cellular function beyond their canonical roles and may represent a class of virulence factors warranting further exploration.


Assuntos
Proteínas de Bactérias/metabolismo , Peptídeos Cíclicos/metabolismo , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Amiloide/genética , Amiloide/metabolismo , Proteínas de Bactérias/genética , Biofilmes/crescimento & desenvolvimento , Humanos , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Peptídeos Cíclicos/genética , Sinais Direcionadores de Proteínas/genética , Percepção de Quorum/genética , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Virulência/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
12.
mBio ; 5(2): e01015, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24713325

RESUMO

The biocide triclosan is used in many personal care products, including toothpastes, soaps, clothing, and medical equipment. Consequently, it is present as a contaminant in the environment and has been detected in some human fluids, including serum, urine, and milk. Staphylococcus aureus is an opportunistic pathogen that colonizes the noses and throats of approximately 30% of the population. Colonization with S. aureus is known to be a risk factor for several types of infection. Here we demonstrate that triclosan is commonly found in the nasal secretions of healthy adults and the presence of triclosan trends positively with nasal colonization by S. aureus. We demonstrate that triclosan can promote the binding of S. aureus to host proteins such as collagen, fibronectin, and keratin, as well as inanimate surfaces such as plastic and glass. Lastly, triclosan-exposed rats are more susceptible to nasal colonization with S. aureus. These data reveal a novel factor that influences the ability of S. aureus to bind surfaces and alters S. aureus nasal colonization. IMPORTANCE Triclosan has been used as a biocide for over 40 years, but the broader effects that it has on the human microbiome have not been investigated. We demonstrate that triclosan is present in nasal secretions of a large portion of a test population and its presence correlates with Staphylococcus aureus nasal colonization. Triclosan also promotes the binding of S. aureus to human proteins and increases the susceptibility of rats to nasal colonization by S. aureus. These findings are significant because S. aureus colonization is a known risk factor for the development of several types of infections. Our data demonstrate the unintended consequences of unregulated triclosan use and contribute to the growing body of research demonstrating inadvertent effects of triclosan on the environment and human health.


Assuntos
Anti-Infecciosos Locais/metabolismo , Portador Sadio/microbiologia , Cavidade Nasal/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Triclosan/metabolismo , Adulto , Animais , Aderência Bacteriana/efeitos dos fármacos , Humanos , Ratos , Sigmodontinae , Staphylococcus aureus/crescimento & desenvolvimento
13.
Biofouling ; 30(3): 337-46, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24564823

RESUMO

The human microbiome is influenced by a number of factors, including environmental exposure to microbes. Because many humans spend a large amount of time in built environments, it can be expected that the microbial ecology of these environments will influence the human microbiome. In an attempt to further understand the microbial ecology of built environments, the microbiota of car interiors was analyzed using culture dependent and culture independent methods. While it was found that the number and type of bacteria varied widely among the cars and sites tested, Staphylococcus and Propionibacterium were nearly always the dominant genera found at the locations sampled. Because Staphylococcus is of particular concern to human health, the characteristics of this genus found in car interiors were investigated. Staphylococcus epidermidis, S. aureus, and S. warnerii were the most prevalent staphylococcal species found, and 22.6% of S. aureus strains isolated from shared community vehicles were resistant to methicillin. The reduction in the prevalence of pathogenic bacteria in cars by using silver-based antimicrobial surface coatings was also evaluated. Coatings containing 5% silver ion additives were applied to steering wheels, placed in cars for five months and were found to eliminate the presence of culturable pathogenic bacteria recovered from these sites relative to controls. Together, these results provide new insight into the microbiota found in an important built environment, the automobile, and potential strategies for controlling the presence of human pathogens.


Assuntos
Anti-Infecciosos/farmacologia , Automóveis , Incrustação Biológica/prevenção & controle , Microbiota , Prata/farmacologia , Contagem de Colônia Microbiana , Exposição Ambiental/análise , Propriedades de Superfície
14.
Am J Respir Crit Care Med ; 189(7): 812-24, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24467627

RESUMO

RATIONALE: Progress has been made in understanding how the cystic fibrosis (CF) basic defect produces lung infection susceptibility. However, it remains unclear why CF exclusively leads to chronic infections that are noninvasive and highly resistant to eradication. Although biofilm formation has been suggested as a mechanism, recent work raises questions about the role of biofilms in CF. OBJECTIVES: To learn how airway conditions attributed to CF transmembrane regulator dysfunction could lead to chronic infection, and to determine if biofilm-inhibiting genetic adaptations that are common in CF isolates affect the capacity of Pseudomonas aeruginosa to develop chronic infection phenotypes. METHODS: We studied P. aeruginosa isolates grown in agar and mucus gels containing sputum from patients with CF and measured their susceptibility to killing by antibiotics and host defenses. We also measured the invasive virulence of P. aeruginosa grown in sputum gels using airway epithelial cells and a murine infection model. MEASUREMENTS AND MAIN RESULTS: We found that conditions likely to result from increased mucus density, hyperinflammation, and defective bacterial killing could all cause P. aeruginosa to grow in bacterial aggregates. Aggregated growth markedly increased the resistance of bacteria to killing by host defenses and antibiotics, and reduced their invasiveness. In addition, we found that biofilm-inhibiting mutations do not impede aggregate formation in gel growth environments. CONCLUSIONS: Our findings suggest that conditions associated with several CF pathogenesis hypotheses could cause the noninvasive and resistant infection phenotype, independently of the bacterial functions needed for biofilm formation.


Assuntos
Fibrose Cística/microbiologia , Infecções por Pseudomonas/etiologia , Pseudomonas aeruginosa/patogenicidade , Animais , Biofilmes , Biomarcadores/metabolismo , Doença Crônica , Fibrose Cística/complicações , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Farmacorresistência Bacteriana , Marcadores Genéticos , Humanos , Elastase de Leucócito/metabolismo , Camundongos , Testes de Sensibilidade Microbiana , Fenótipo , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/isolamento & purificação , Pseudomonas aeruginosa/fisiologia , Escarro/metabolismo , Escarro/microbiologia , Virulência
15.
Diabetes Metab Syndr Obes ; 6: 17-29, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23357897

RESUMO

BACKGROUND: Diabetic foot ulcers are a major source of morbidity, limb loss, and mortality. A prolonged inflammatory response, extracellular matrix degradation irregularities, and increased bacteria presence have all been hypothesized as major contributing factors in the delayed healing of diabetic wounds. Collagen components such as fibroblast and keratinocytes are fundamental to the process of wound healing and skin formation. Wound dressings that contain collagen products create a biological scaffold matrix that supports the regulation of extracellular components and promotes wound healing. METHODS: A systematic review of studies reporting collagen wound dressings used in the treatment of Diabetic foot ulcers was conducted. Comprehensive searches were run in Ovid MEDLINE, PubMed, EMBASE, and ISI Web of Science to capture citations pertaining to the use of collagen wound dressings in the treatment of diabetic foot ulcers. The searches were limited to human studies reported in English. RESULTS: Using our search strategy, 26 papers were discussed, and included 13 randomized designs, twelve prospective cohorts, and one retrospective cohort, representing 2386 patients with diabetic foot ulcers. Our design was not a formal meta-analysis. In those studies where complete epithelialization, 58% of collagen-treated wounds completely healed (weighted mean 67%). Only 23% of studies reported control group healing with 29% healing (weighted mean 11%) described for controls. CONCLUSION: Collagen-based wound dressings can be an effective tool in the healing of diabetic foot wounds. The current studies show an overall increase in healing rates despite limitations in study designs. This study suggests that future works focus on biofilms and extracellular regulation, and include high risk patients.

16.
Curr Opin Microbiol ; 16(1): 93-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23313395

RESUMO

The aggregation of proteins into amyloid fibers is a common characteristic of many neurodegenerative disorders including Alzheimer's, Parkinson's, and prion diseases. Amyloid formation was originally characterized in these systems and is traditionally viewed as a consequence of protein misfolding and aggregation. An emerging field of study brings functional amyloids, like those produced by bacteria, into the scientific mainstream, and demonstrates a ubiquitous role for amyloids in living systems. This review aims to summarize what is known about the bacterial amyloids and their interactions within various host environments.


Assuntos
Amiloide/metabolismo , Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Complexos Multiproteicos/metabolismo , Bactérias/patogenicidade , Interações Hospedeiro-Patógeno , Fatores de Virulência/metabolismo
17.
Infect Immun ; 81(2): 496-504, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23208606

RESUMO

Staphylococcus aureus is a human commensal and pathogen that is capable of forming biofilms on a variety of host tissues and implanted medical devices. Biofilm-associated infections resist antimicrobial chemotherapy and attack from the host immune system, making these infections particularly difficult to treat. In order to gain insight into environmental conditions that influence S. aureus biofilm development, we screened a library of small molecules for the ability to inhibit S. aureus biofilm formation. This led to the finding that the polyphenolic compound tannic acid inhibits S. aureus biofilm formation in multiple biofilm models without inhibiting bacterial growth. We present evidence that tannic acid inhibits S. aureus biofilm formation via a mechanism dependent upon the putative transglycosylase IsaA. Tannic acid did not inhibit biofilm formation of an isaA mutant. Overexpression of wild-type IsaA inhibited biofilm formation, whereas overexpression of a catalytically dead IsaA had no effect. Tannin-containing drinks like tea have been found to reduce methicillin-resistant S. aureus nasal colonization. We found that black tea inhibited S. aureus biofilm development and that an isaA mutant resisted this inhibition. Antibiofilm activity was eliminated from tea when milk was added to precipitate the tannic acid. Finally, we developed a rodent model for S. aureus throat colonization and found that tea consumption reduced S. aureus throat colonization via an isaA-dependent mechanism. These findings provide insight into a molecular mechanism by which commonly consumed polyphenolic compounds, such as tannins, influence S. aureus surface colonization.


Assuntos
Antígenos de Bactérias/metabolismo , Aderência Bacteriana/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Taninos/farmacologia , Animais , Antígenos de Bactérias/genética , Biofilmes/efeitos dos fármacos , Domínio Catalítico/efeitos dos fármacos , Feminino , Ratos , Sigmodontinae , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Staphylococcus aureus/fisiologia , Chá/metabolismo
18.
Biofouling ; 29(1): 53-68, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23194413

RESUMO

Showerheads support the development of multi-species biofilms that can be unsightly, produce malodor, and may harbor pathogens. The outer-surface spray-plates of many showerheads support visible biofilms that likely contain a mixture of bacteria from freshwater and potentially from human users. Coaggregation, a mechanism by which genetically distinct bacteria specifically recognize one another, may contribute to the retention and enrichment of different species within these biofilms. The aim of this work was to describe the bacterial composition of outer spray-plate biofilms of three domestic showerheads and to determine the intra- and inter-biofilm coaggregation ability of each culturable isolate. The bacterial composition of the three biofilms was determined by using bacterial tag-encoded FLX amplicon pyrosequencing (bTEFAP) and by culturing on R2A medium. An average of 31 genera per biofilm were identified using bTEFAP and a total of 30 isolates were cultured. Even though the microbial diversity of each showerhead biofilm differed, every cultured isolate was able to coaggregate with at least one other isolate from the same or different showerhead biofilm. Promiscuous coaggregating isolates belonged to the genera Brevundimonas, Micrococcus, and Lysobacter. This work suggests that coaggregation may be a common feature of showerhead biofilms. Characterization of the mechanisms mediating coaggregation, and the inter-species interactions they facilitate, may allow for novel strategies to inhibit biofilm development.


Assuntos
Bactérias/classificação , Fenômenos Fisiológicos Bacterianos , Banhos , Biofilmes/crescimento & desenvolvimento , Água Doce/microbiologia , Bactérias/genética , Bactérias/isolamento & purificação , Aderência Bacteriana , Proteínas de Bactérias/genética , Contagem de Colônia Microbiana , Interações Hidrofóbicas e Hidrofílicas , Microscopia Confocal , Filogenia , Reação em Cadeia da Polimerase , RNA Ribossômico 16S/genética , Alinhamento de Sequência , Análise de Sequência de DNA , Titânio/química , Abastecimento de Água
20.
PLoS Pathog ; 8(6): e1002744, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22685403

RESUMO

Staphylococcus aureus is an opportunistic pathogen that colonizes the skin and mucosal surfaces of mammals. Persistent staphylococcal infections often involve surface-associated communities called biofilms. Here we report the discovery of a novel extracellular fibril structure that promotes S. aureus biofilm integrity. Biochemical and genetic analysis has revealed that these fibers have amyloid-like properties and consist of small peptides called phenol soluble modulins (PSMs). Mutants unable to produce PSMs were susceptible to biofilm disassembly by matrix degrading enzymes and mechanical stress. Previous work has associated PSMs with biofilm disassembly, and we present data showing that soluble PSM peptides disperse biofilms while polymerized peptides do not. This work suggests the PSMs' aggregation into amyloid fibers modulates their biological activity and role in biofilms.


Assuntos
Amiloide/química , Toxinas Bacterianas/química , Biofilmes/crescimento & desenvolvimento , Staphylococcus aureus/fisiologia , Amiloide/metabolismo , Toxinas Bacterianas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA