Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
3.
Materials (Basel) ; 14(10)2021 May 12.
Article in English | MEDLINE | ID: mdl-34065926

ABSTRACT

Surface texturing has brought significant improvements in the functional properties of parts and components. Sinker electro discharge machining (SEDM) is one of the processes which generates great texturing results at different scale. An electrode is needed to reproduce the geometry to be textured. Some geometries are difficult or impossible to achieve on an electrode using conventional and even unconventional machining methods. This work sets out the advances made in the manufacturing of copper electrodes for electro erosion by additive manufacturing, and their subsequent application to the functional texturing of Al-Cu UNS A92024-T3 alloy. A combined procedure of digital light processing (DLP) additive manufacturing, sputtering and micro-electroforming (AMSME), has been used to produce electrodes. Also, a specific laboratory equipment has been developed to reproduce details on a microscopic scale. Shells with outgoing spherical geometries pattern have been manufactured. AMSME process has shown ability to copper electrodes manufacturing. A highly detailed surface on a micrometric scale have been achieved. Copper shells with minimum thickness close to 300 µm have been tested in sinker electro discharge machining (SEDM) and have been shown very good performance in surface finishing operations. The method has shown great potential for use in surfaces texturing.

5.
Methods Mol Biol ; 2118: 269-279, 2020.
Article in English | MEDLINE | ID: mdl-32152986

ABSTRACT

Nanomaterials have become increasingly important in medicine, manufacturing, and consumer products. A fundamental understanding of the effects of nanoparticles (NPs) and their interactions with biomolecules and organismal systems has yet to be achieved. In this chapter, we firstly provide a brief review of the interactions between nanoparticles and biological systems. We then provide an example by describing a novel method to assess the effects of NPs on biological systems, using insects as a model. Nanoparticles were injected into the central nervous system of the discoid cockroach (Blaberus discoidalis). It was found that insects became hyperactive compared to negative control (water injections). Our method could provide a generic method of assessing nanoparticles toxicity.


Subject(s)
Cockroaches/physiology , Motor Activity/drug effects , Nanoparticles/toxicity , Animals , Central Nervous System/chemistry , Central Nervous System/drug effects , Central Nervous System/physiology , Cockroaches/drug effects , Female , Male , Models, Animal , Nanoparticles/administration & dosage
6.
Med Mycol ; 55(3): 334-343, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-27601610

ABSTRACT

Soft-tissue invasive fungal infections are increasingly recognized as significant entities directly contributing to morbidity and mortality. They complicate clinical care, requiring aggressive surgical debridement and systemic antifungal therapy. To evaluate new topical approaches to therapy, we examined the antifungal activity and cytotoxicity of Manuka Honey (MH) and polyhexamethylene biguanide (PHMB). The activities of multiple concentrations of MH (40%, 60%, 80%) and PHMB (0.01%, 0.04%, 0.1%) against 13 clinical mould isolates were evaluated using a time-kill assay between 5 min and 24 h. Concentrations were selected to represent current clinical use. Cell viability was examined in parallel for human epidermal keratinocytes, dermal fibroblasts and osteoblasts, allowing determination of the 50% viability (LD50) concentration. Antifungal activity of both agents correlated more closely with exposure time than concentration. Exophiala and Fusarium growth was completely suppressed at 5 min for all PHMB concentrations, and at 12 and 6 h, respectively, for all MH concentrations. Only Lichtheimia had persistent growth to both agents at 24 h. Viability assays displayed concentration-and time-dependent toxicity for PHMB. For MH, exposure time predicted cytotoxicity only when all cell types were analyzed in aggregate. This study demonstrates that MH and PHMB possess primarily time-dependent antifungal activity, but also exert in vitro toxicity on human cells which may limit clinical use. Further research is needed to determine ideal treatment strategies to optimize antifungal activity against moulds while limiting cytotoxicity against host tissues in vivo.


Subject(s)
Biguanides/pharmacology , Disinfectants/pharmacology , Fibroblasts/drug effects , Fungi/drug effects , Honey , Keratinocytes/drug effects , Osteoblasts/drug effects , Biguanides/toxicity , Cell Line , Cell Survival/drug effects , Disinfectants/toxicity , Fibroblasts/physiology , Fungi/physiology , Humans , Keratinocytes/physiology , Lethal Dose 50 , Microbial Sensitivity Tests , Osteoblasts/physiology , Time Factors
7.
Adv Exp Med Biol ; 973: 53-70, 2017.
Article in English | MEDLINE | ID: mdl-27864804

ABSTRACT

Biofilm formation is a major virulence factor for numerous pathogenic bacteria and is cited as a central event in the pathogenesis of chronic human infections, which is in large part due to excessive extracellular matrix secretion and metabolic changes that occur within the biofilm rendering them highly tolerant to antimicrobial treatments. Polyamines, including norspermidine, play central roles in bacterial biofilm development, but have also recently been shown to inhibit biofilm formation in select strains of various pathogenic bacteria. The aim of this study was to evaluate in vitro the biofilm dispersive and inhibitory activities of norspermidine against multidrug-resistant clinical isolates of Acinetobacter baumannii(n = 4), Klebsiella pneumoniae (n = 3), Pseudomonas aeruginosa (n = 5) and Staphylococcus aureus (n = 4) associated with chronic extremity wound infections using the semi-quantitative 96-well plate method and confocal laser microscopy. In addition to the antibiofilm activity, biocompatibility of norspermidine was also evaluated by measuring toxicity in vitro to human cell lines and whole porcine tissue explants using MTT viability assay and histological analysis. Norspermidine (5-20 mM) had variable dispersive and inhibitory activity on biofilms which was dependent on both the strain and species. Of the clinical bacterial species evaluated herein, A. baumannii isolates were the most sensitive to the effect of norspermidine, which was in part due to the inhibitory effects of norspermidine on bacterial motility and expression of genes involved in the production of homoserine lactones and quorum sensing molecules both essential for biofilm formation. Importantly, exposure of cell lines and whole tissues to norspermidine for prolonged periods of time (≥24 h) was observed to reduce viability and alter tissue histology in a time and concentration dependent manner, with 20 mM exposure having the greatest negative effects on both tissues and individual cell lines. Collectively our findings demonstrate that, similar to other polyamines, norspermidine displays both inhibitory and dispersive activities on biofilms of clinical multidrug-resistant bacterial isolates, in particular for strains of A. baumannii. Additionally our findings suggest that direct application may be considered on tissues, albeit for limited exposure times.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Drug Resistance, Multiple, Bacterial , Pseudomonas aeruginosa/drug effects , Spermidine/analogs & derivatives , Staphylococcus aureus/drug effects , Wound Infection/microbiology , Acinetobacter baumannii/physiology , Humans , Pseudomonas aeruginosa/physiology , Quorum Sensing/drug effects , Spermidine/pharmacology , Staphylococcus aureus/physiology
8.
BMC Res Notes ; 9: 216, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27071769

ABSTRACT

BACKGROUND: Bacterial infections are a common clinical problem in both acute and chronic wounds. With growing concerns over antibiotic resistance, treatment of bacterial infections should only occur after positive diagnosis. Currently, diagnosis is delayed due to lengthy culturing methods which may also fail to identify the presence of bacteria. While newer costly bacterial identification methods are being explored, a simple and inexpensive diagnostic tool would aid in immediate and accurate treatments for bacterial infections. Histologically, hematoxylin and eosin (H&E) and Gram stains have been employed, but are far from optimal when analyzing tissue samples due to non-specific staining. The goal of the current study was to develop a modification of the Gram stain that enhances the contrast between bacteria and host tissue. FINDINGS: A modified Gram stain was developed and tested as an alternative to Gram stain that improves the contrast between Gram positive bacteria, Gram negative bacteria and host tissue. Initially, clinically relevant strains of Pseudomonas aeruginosa and Staphylococcus aureus were visualized in vitro and in biopsies of infected, porcine burns using routine Gram stain, and immunohistochemistry techniques involving bacterial strain-specific fluorescent antibodies as validation tools. H&E and Gram stain of serial biopsy sections were then compared to a modification of the Gram stain incorporating a counterstain that highlights collagen found in tissue. The modified Gram stain clearly identified both Gram positive and Gram negative bacteria, and when compared to H&E or Gram stain alone provided excellent contrast between bacteria and non-viable burn eschar. Moreover, when applied to surgical biopsies from patients that underwent burn debridement this technique was able to clearly detect bacterial morphology within host tissue. CONCLUSIONS: We describe a modification of the Gram stain that provides improved contrast of Gram positive and Gram negative microorganisms within host tissue. The samples used in this study demonstrate that this staining technique has laboratory and clinical applicability. This modification only adds minutes to traditional Gram stain with reusable reagents, and results in a cost- and time-efficient technique for identifying bacteria in any clinical biopsy containing connective tissue.


Subject(s)
Bacterial Infections/diagnosis , Bacterial Infections/microbiology , Gram-Positive Bacteria/physiology , Staining and Labeling/methods , Animals , Burns/microbiology , Eosine Yellowish-(YS) , Gentian Violet , Hematoxylin , Host-Pathogen Interactions , Humans , Immunohistochemistry , Microscopy, Video , Phenazines , Pseudomonas aeruginosa/physiology , Reproducibility of Results , Sensitivity and Specificity , Skin/microbiology , Staphylococcus aureus/physiology , Swine
9.
J Orthop Trauma ; 30(10): 531-7, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27124826

ABSTRACT

OBJECTIVES: To evaluate the effectiveness of locally applied vancomycin powder at different times postinfection in a contaminated traumatic animal model. METHODS: This study used an established segmental defect rat femur model contaminated with Staphylococcus aureus UAMS-1 followed by treatment at 6 or 24 hours postinfection. Three treatments were evaluated: debridement and irrigation alone (control group) or in combination with either vancomycin powder or vancomycin-impregnated poly(methyl methacrylate) beads. Serum vancomycin levels were determined at scheduled time points over 14 days; bone, surrounding muscle, and implants were harvested for bacterial and inflammatory analyses. RESULTS: Locally applied vancomycin powder and impregnated beads significantly reduced bacteria both within the bone and implant when treatment was performed at 6 hours. Delaying treatment to 24 hours significantly reduced the therapeutic efficacy of locally applied vancomycin of both groups. Serum vancomycin levels were detectable in all animals treated with vancomycin powder at 24 hours, but absorption was negligible from beads. At 14 days, vancomycin was detectable in the surrounding musculature of all animals and in serum of 20% of animals treated with vancomycin powder. CONCLUSIONS: This study suggests that vancomycin powder is a promising adjunctive therapy for preventing infection in traumatic wounds when treatment is performed early. This time-dependent effectiveness of vancomycin powder is similar to that observed with systemic and other local delivery adjuncts, which is likely attributable to biofilm formation after contamination, conferring intrinsic recalcitrance to antimicrobials.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Femoral Fractures/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/isolation & purification , Vancomycin/administration & dosage , Wound Infection/prevention & control , Animals , Debridement , Disease Models, Animal , Femoral Fractures/surgery , Femur/microbiology , Femur/surgery , Fractures, Open/microbiology , Fractures, Open/surgery , Microspheres , Polymethyl Methacrylate , Powders , Rats , Rats, Inbred Lew , Staphylococcal Infections/microbiology , Staphylococcal Infections/surgery , Staphylococcus aureus/drug effects , Therapeutic Irrigation , Time Factors , Wound Infection/microbiology , Wound Infection/surgery
10.
Pharmaceuticals (Basel) ; 9(1)2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26999163

ABSTRACT

Acinetobacter baumannii is a challenging pathogen due to antimicrobial resistance and biofilm development. The role of iron in bacterial physiology has prompted the evaluation of iron-modulation as an antimicrobial strategy. The non-reducible iron analog gallium(III) nitrate, Ga(NO3)3, has been shown to inhibit A. baumannii planktonic growth; however, utilization of heme-iron by clinical isolates has been associated with development of tolerance. These observations prompted the evaluation of iron-heme sources on planktonic and biofilm growth, as well as antimicrobial activities of gallium meso- and protoporphyrin IX (Ga-MPIX and Ga-PPIX), metal heme derivatives against planktonic and biofilm bacteria of multidrug-resistant (MDR) clinical isolates of A. baumannii in vitro. Ga(NO3)3 was moderately effective at reducing planktonic bacteria (64 to 128 µM) with little activity against biofilms (≥512 µM). In contrast, Ga-MPIX and Ga-PPIX were highly active against planktonic bacteria (0.25 to 8 µM). Cytotoxic effects in human fibroblasts were observed following exposure to concentrations exceeding 128 µM of Ga-MPIX and Ga-PPIX. We observed that the gallium metal heme conjugates were more active against planktonic and biofilm bacteria, possibly due to utilization of heme-iron as demonstrated by the enhanced effects on bacterial growth and biofilm formation.

11.
Biomed Res Int ; 2016: 7078989, 2016.
Article in English | MEDLINE | ID: mdl-26885514

ABSTRACT

Antibiotic-loaded bone cements, including poly(methyl methacrylate) (PMMA) and calcium sulfate (CaSO4), are often used for treatment of orthopaedic infections involving Staphylococcus spp., although the effectiveness of this treatment modality may be limited due to the emergence of antimicrobial resistance and/or the development of biofilms within surgical sites. Gallium(III) is an iron analog capable of inhibiting essential iron-dependent pathways, exerting broad antimicrobial activity against multiple microorganisms, including Staphylococcus spp. Herein, we evaluated PMMA and CaSO4 as carriers for delivery of gallium(III) nitrate (Ga(NO3)3) to infected surgical sites by assessing the release kinetics subsequent to incorporation and antimicrobial activity against S. aureus and S. epidermidis. PMMA and to a lesser extent CaSO4 were observed to be compatible as carriers for Ga(NO3)3, eluting concentrations with antimicrobial activity against planktonic bacteria, inhibiting bacterial growth, and preventing bacterial colonization of beads, and effective against established bacterial biofilms of S. aureus and S. epidermidis. Collectively, our in vitro results indicate that PMMA is a more suitable carrier compared to CaSO4 for delivery of Ga(NO3)3; moreover they provide evidence for the potential use of Ga(NO3)3 with PMMA as a strategy for the prevention and/or treatment for orthopaedic infections.


Subject(s)
Drug Carriers/administration & dosage , Methicillin-Resistant Staphylococcus aureus/drug effects , Polymethyl Methacrylate/administration & dosage , Staphylococcal Infections/drug therapy , Biofilms/drug effects , Bone Cements/chemistry , Bone Cements/therapeutic use , Calcium Sulfate/administration & dosage , Drug Carriers/chemistry , Gallium/administration & dosage , Humans , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Polymethyl Methacrylate/chemistry , Staphylococcal Infections/pathology , Staphylococcal Infections/surgery
12.
Antimicrob Agents Chemother ; 59(12): 7205-13, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26324277

ABSTRACT

Periostitis, which is characterized by bony pain and diffuse periosteal ossification, has been increasingly reported with prolonged clinical use of voriconazole. While resolution of clinical symptoms following discontinuation of therapy suggests a causative role for voriconazole, the biological mechanisms contributing to voriconazole-induced periostitis are unknown. To elucidate potential mechanisms, we exposed human osteoblasts in vitro to voriconazole or fluconazole at 15 or 200 µg/ml (reflecting systemic or local administration, respectively), under nonosteogenic or osteogenic conditions, for 1, 3, or 7 days and evaluated the effects on cell proliferation (reflected by total cellular DNA) and osteogenic differentiation (reflected by alkaline phosphatase activity, calcium accumulation, and expression of genes involved in osteogenic differentiation). Release of free fluoride, vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF) was also measured in cell supernatants of osteoblasts exposed to triazoles, with an ion-selective electrode (for free fluoride) and enzyme-linked immunosorbent assays (ELISAs) (for VEGF and PDGF). Voriconazole but not fluconazole significantly enhanced the proliferation and differentiation of osteoblasts. In contrast to clinical observations, no increases in free fluoride levels were detected following exposure to either voriconazole or fluconazole; however, significant increases in the expression of VEGF and PDGF by osteoblasts were observed following exposure to voriconazole. Our results demonstrate that voriconazole can induce osteoblast proliferation and enhance osteogenic activity in vitro. Importantly, and in contrast to the previously proposed mechanism of fluoride-stimulated osteogenesis, our findings suggest that voriconazole-induced periostitis may also occur through fluoride-independent mechanisms that enhance the expression of cytokines that can augment osteoblastic activity.


Subject(s)
Antifungal Agents/pharmacology , Fluconazole/pharmacology , Osteoblasts/drug effects , Osteogenesis/drug effects , Voriconazole/pharmacology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Gene Expression/drug effects , Humans , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/genetics , Platelet-Derived Growth Factor/agonists , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Primary Cell Culture , Signal Transduction , Sodium Fluoride/pharmacology , Vascular Endothelial Growth Factor A/agonists , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
13.
Clin Orthop Relat Res ; 473(12): 3951-61, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26201421

ABSTRACT

BACKGROUND: Infectious complications of musculoskeletal trauma are an important factor contributing to patient morbidity. Biofilm-dispersive bone grafts augmented with D-amino acids (D-AAs) prevent biofilm formation in vitro and in vivo, but the effects of D-AAs on osteocompatibility and new bone formation have not been investigated. QUESTIONS/PURPOSES: We asked: (1) Do D-AAs hinder osteoblast and osteoclast differentiation in vitro? (2) Does local delivery of D-AAs from low-viscosity bone grafts inhibit new bone formation in a large-animal model? METHODS: Methicillin-sensitive Staphylococcus aureus and methicillin-resistant S aureus clinical isolates, mouse bone marrow stromal cells, and osteoclast precursor cells were treated with an equal mass (1:1:1) mixture of D-Pro:D-Met:D-Phe. The effects of the D-AA dose on biofilm inhibition (n = 4), biofilm dispersion (n = 4), and bone marrow stromal cell proliferation (n = 3) were quantitatively measured by crystal violet staining. Osteoblast differentiation was quantitatively assessed by alkaline phosphatase staining, von Kossa staining, and quantitative reverse transcription for the osteogenic factors a1Col1 and Ocn (n = 3). Osteoclast differentiation was quantitatively measured by tartrate-resistant acid phosphatase staining (n = 3). Bone grafts augmented with 0 or 200 mmol/L D-AAs were injected in ovine femoral condyle defects in four sheep. New bone formation was evaluated by µCT and histology 4 months later. An a priori power analysis indicated that a sample size of four would detect a 7.5% difference of bone volume/total volume between groups assuming a mean and SD of 30% and 5%, respectively, with a power of 80% and an alpha level of 0.05 using a two-tailed t-test between the means of two independent samples. RESULTS: Bone marrow stromal cell proliferation, osteoblast differentiation, and osteoclast differentiation were inhibited at D-AAs concentrations of 27 mmol/L or greater in a dose-responsive manner in vitro (p < 0.05). In methicillin-sensitive and methicillin-resistant S aureus clinical isolates, D-AAs inhibited biofilm formation at concentrations of 13.5 mmol/L or greater in vitro (p < 0.05). Local delivery of D-AAs from low-viscosity grafts did not inhibit new bone formation in a large-animal model pilot study (0 mmol/L D-AAs: bone volume/total volume = 26.9% ± 4.1%; 200 mmol/L D-AAs: bone volume/total volume = 28.3% ± 15.4%; mean difference with 95% CI = -1.4; p = 0.13). CONCLUSIONS: D-AAs inhibit biofilm formation, bone marrow stromal cell proliferation, osteoblast differentiation, and osteoclast differentiation in vitro in a dose-responsive manner. Local delivery of D-AAs from bone grafts did not inhibit new bone formation in vivo at clinically relevant doses. CLINICAL RELEVANCE: Local delivery of D-AAs is an effective antibiofilm strategy that does not appear to inhibit bone repair. Longitudinal studies investigating bacterial burden, bone formation, and bone remodeling in contaminated defects as a function of D-AA dose are required to further support the use of D-AAs in the clinical management of infected open fractures.


Subject(s)
Amino Acids/pharmacology , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Bone Transplantation/methods , Femur/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Osteogenesis/drug effects , Animals , Anti-Bacterial Agents/toxicity , Biofilms/growth & development , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Female , Femur/diagnostic imaging , Femur/metabolism , Femur/surgery , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Methicillin-Resistant Staphylococcus aureus/growth & development , Methionine/pharmacology , Mice , Models, Animal , Osseointegration/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Phenylalanine/pharmacology , Proline/pharmacology , Sheep, Domestic , Time Factors , X-Ray Microtomography
14.
BMC Infect Dis ; 15: 223, 2015 Jun 07.
Article in English | MEDLINE | ID: mdl-26049931

ABSTRACT

BACKGROUND: Biofilms are associated with persistent infection. Reports characterizing clinical infectious outcomes and patient risk factors for colonization or infection with biofilm forming isolates are scarce. Our institution recently published a study examining the biofilm forming ability of 205 randomly selected clinical isolates. This present study aims to identify potential risk factors associated with these isolates and assess clinical infectious outcomes. METHODS: 221 clinical isolates collected from 2005 to 2012 and previously characterized for biofilm formation were studied. Clinical information from the associated patients, including demographics, comorbidities, antibiotic usage, laboratory values, and clinical infectious outcomes, was determined retrospectively through chart review. Duplicate isolates and non-clinical isolates were excluded from analysis. Associations with biofilm forming isolates were determined by univariate analysis and multivariate analysis. RESULTS: 187 isolates in 144 patients were identified for analysis; 113 were biofilm producers and 74 were not biofilm producers. Patients were primarily male (78 %) military members (61 %) with combat trauma (52 %). On multivariate analysis, the presence of methicillin-resistant Staphylococcus aureus (p < 0.01, OR 5.09, 95 % CI 1.12, 23.1) and Pseudomonas aeruginosa (p = 0.02, OR 3.73, 95 % CI 1.46, 9.53) were the only characteristics more likely to be present in the biofilm producing isolate group. Infectious outcomes of patients with non-biofilm forming isolates, including cure, relapse/reinfection, and chronic infection, were similar to infectious outcomes of patients with biofilm-forming isolates. Mortality with initial infection was higher in the biofilm producing isolate group (16 % vs 5 %, p = 0.01) but attributable mortality was low (1 of 14). No characteristics examined in this study were found to be associated with relapse/reinfection or chronic infection on multivariate analysis. CONCLUSIONS: Bacteria species, but not clinical characteristics, were associated with biofilm formation on multivariate analysis. Biofilm forming isolates and non-biofilm forming isolates had similar infectious outcomes in this study.


Subject(s)
Bacterial Infections/pathology , Biofilms/growth & development , Methicillin-Resistant Staphylococcus aureus/physiology , Pseudomonas aeruginosa/physiology , Adult , Bacterial Infections/microbiology , Demography , Female , Humans , Male , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Middle Aged , Multivariate Analysis , Pseudomonas aeruginosa/isolation & purification , Recurrence , Retrospective Studies , Risk Factors
15.
BMC Microbiol ; 15: 75, 2015 Mar 28.
Article in English | MEDLINE | ID: mdl-25886581

ABSTRACT

BACKGROUND: Chronic, non-healing wounds are often characterized by the persistence of bacteria within biofilms - aggregations of cells encased within a self-produced polysaccharide matrix. Biofilm bacteria exhibit unique characteristics from planktonic, or culture-grown, bacterial phenotype, including diminished responses to antimicrobial therapy and persistence against host immune responses. Mesenchymal stromal cells (MSCs) are host cells characterized by their multifunctional ability to undergo differentiation into multiple cell types and modulation of host-immune responses by secreting factors that promote wound healing. While these characteristics make MSCs an attractive therapeutic for wounds, these pro-healing activities may be differentially influenced in the context of an infection (i.e., biofilm related infections) within chronic wounds. Herein, we evaluated the effect of soluble factors derived from biofilms of clinical isolates of Staphylococcus aureus and Pseudomonas aeruginosa on the viability, differentiation, and paracrine activity of human MSCs to evaluate the influence of biofilms on MSC activity in vitro. RESULTS: Exposure of MSCs to biofilm-conditioned medias of S. aureus and P. aeruginosa resulted in reductions in cell viability, in part due to activation of apoptosis. Similarly, exposure to soluble factors from biofilms was also observed to diminish the migration ability of cells and to hinder multi-lineage differentiation of MSCs. In contrast to these findings, exposure of MSCs to soluble factors from biofilms resulted in significant increases in the release of paracrine factors involved in inflammation and wound healing. CONCLUSIONS: Collectively, these findings demonstrate that factors produced by biofilms can negatively impact the intrinsic properties of MSCs, in particular limiting the migratory and differentiation capacity of MSCs. Consequently, these studies suggest use/application of stem-cell therapies in the context of infection may have a limited therapeutic effect.


Subject(s)
Mesenchymal Stem Cells/drug effects , Organic Chemicals/toxicity , Pseudomonas aeruginosa/chemistry , Staphylococcus aureus/chemistry , Wound Infection/microbiology , Biofilms/growth & development , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Culture Media, Conditioned , Cytokines/metabolism , Humans , Mesenchymal Stem Cells/physiology , Neovascularization, Physiologic/drug effects , Organic Chemicals/isolation & purification , Organic Chemicals/metabolism , Pseudomonas aeruginosa/isolation & purification , Pseudomonas aeruginosa/physiology , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/physiology
16.
Clin Orthop Relat Res ; 473(9): 2874-84, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25896136

ABSTRACT

BACKGROUND: Local antimicrobial delivery through polymethylmethacrylate beads (PMMA), commonly vancomycin, is used for the treatment of contaminated open fractures but has limited activity against Staphylococcus aureus biofilms, which occur commonly in such fractures. Rifamycins have activity against biofilms and are an effective treatment for osteoarticular infections involving staphylococcal biofilms, but there are limited studies evaluating the activity of rifamycin derivatives, other than rifampin, against biofilms of S. aureus and evaluating incorporation of these drugs into PMMA for treatment of contaminated open fractures. QUESTIONS/PURPOSES: (1) Are rifamycin derivatives effective against established biofilms of clinical isolates of S. aureus? (2) Can PMMA be used as a carrier for rifamycin derivatives? METHODS: Biofilms were developed and evaluated for susceptibility to a panel of antimicrobials in vitro using the minimum biofilm eradication concentration high-throughput model. Susceptibility was assessed by measuring bacterial recovery at 6 and 24 hours after antimicrobial treatment. Activity of rifamycin derivatives against intracellular bacteria was also evaluated using a gentamicin protection assay. Evaluation of PMMA as a carrier for rifampin and rifamycin derivatives was determined by assessing the curing time subsequent to loading of rifamycins and characterizing the release kinetics of rifamycins at daily intervals for 14 days from PMMA by performing bioassays. RESULTS: Rifamycin derivatives between 1 and 8 µg/mL reduced bacteria within biofilms 5- to 9-logs and prevented bacterial recovery up to 24 hours post-treatment, indicating near to complete eradication of biofilms. Rifamycin derivatives at 32 µg/mL had activity against intracellular staphylococci, significantly reducing the number of internalized bacteria with limited effects on osteoblast viability. Rifampin was the only rifamycin observed to have a suitable release profile from PMMA, releasing 49% of the total antibiotic and maintaining a sustained released profile up to 14 days at a mean 28 ± 6 µg/mL. CONCLUSIONS: Rifampin can be incorporated into PMMA and eluted at concentrations effective against biofilms and intracellular staphylococci. CLINICAL RELEVANCE: Our in vitro findings suggest that local delivery of rifampin may be an effective strategy for the prevention and/or treatment of open fractures where S. aureus biofilms might develop. Clinical studies are needed to characterize what role this approach might have in the prevention and treatment of infections involving biofilms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Drug Carriers , Polymethyl Methacrylate/chemistry , Rifamycins/pharmacology , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Biofilms/growth & development , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Kinetics , Microbial Sensitivity Tests , Rifamycins/administration & dosage , Rifamycins/chemistry , Solubility , Staphylococcus aureus/growth & development
17.
J R Soc Interface ; 12(105)2015 Apr 06.
Article in English | MEDLINE | ID: mdl-25740855

ABSTRACT

Natural systems retain significant advantages over engineered systems in many aspects, including size and versatility. In this research, we develop a hybrid robotic system using American (Periplaneta americana) and discoid (Blaberus discoidalis) cockroaches that uses the natural locomotion and robustness of the insect. A tethered control system was firstly characterized using American cockroaches, wherein implanted electrodes were used to apply an electrical stimulus to the prothoracic ganglia. Using this approach, larger discoid cockroaches were engineered into a remotely controlled hybrid robotic system. Locomotion control was achieved through electrical stimulation of the prothoracic ganglia, via a remotely operated backpack system and implanted electrodes. The backpack consisted of a microcontroller with integrated transceiver protocol, and a rechargeable battery. The hybrid discoid roach was able to walk, and turn in response to an electrical stimulus to its nervous system with high repeatability of 60%.


Subject(s)
Cockroaches/physiology , Hybridization, Genetic , Locomotion/physiology , Robotics/methods , Animals , Cockroaches/genetics , Electric Stimulation
18.
Med Mycol ; 53(3): 285-94, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25631479

ABSTRACT

Invasive fungal wound infections (IFIs) are increasingly reported in trauma patients and cause considerable morbidity and mortality despite standard of care treatment in trauma centers by experienced medical personnel. Topical agents such as oil of melaleuca, also known as tea tree oil (TTO), have been proposed for adjunctive treatment of IFIs. We evaluated the activity of TTO against filamentous fungi associated with IFIs by testing 13 clinical isolates representing nine species via time-kill assay with seven concentrations of TTO (100%, 75%, 50%, 25%, 10%, 5%, and 1%). To ascertain the safety of topical application to wounds, cell viability assays were performed in vitro using human fibroblasts, keratinocytes, osteoblasts, and umbilical vein endothelial cells with 10 concentrations of TTO (75%, 50%, 25%, 10%, 5%, and 10-fold serial dilutions from 1 to 0.0001%) at five time points (5, 15, 30, 60, and 180 min). Compatibility of TTO with explanted porcine tissues was also assessed with eight concentrations of TTO (100%, 75%, 50%, 25%, 10%, 5%, 1%, and 0.1%) at the time points used for cellular assays and at 24 h. The time-kill studies showed that fungicidal activity was variable between isolates. The effect of TTO on cell viability was primarily concentration dependent with significant cytotoxicity at concentrations of ≥ 10% and ≥ 50% for cells lines and whole tissue, respectively. Our findings demonstrate that TTO possesses antifungal activity against filamentous fungi associated with IFIs; furthermore that negligible effects on whole tissues, in contrast to individual cells, were observed following exposure to TTO. Collectively, these findings indicate a potential use of TTO as topical treatment of IFIs.


Subject(s)
Antifungal Agents/pharmacology , Antifungal Agents/toxicity , Fungi/drug effects , Melaleuca/chemistry , Tea Tree Oil/pharmacology , Tea Tree Oil/toxicity , Animals , Antifungal Agents/isolation & purification , Cell Line , Cell Survival/drug effects , Endothelial Cells/drug effects , Fibroblasts/drug effects , Fungi/isolation & purification , Humans , Keratinocytes/drug effects , Mycoses/microbiology , Osteoblasts/drug effects , Swine , Tea Tree Oil/isolation & purification , Wound Infection/microbiology
19.
J Surg Res ; 192(2): 692-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25130774

ABSTRACT

BACKGROUND: Macrophages are important in wound defense and healing. Dakin's solution (DS), buffered sodium hypochlorite, has been used since World War I as a topical antimicrobial for wound care. DS has been shown to be toxic to host cells, but effects on immune cells are not well documented. MATERIALS AND METHODS: DS at 0.5%, 0.125%, and ten-fold serial dilutions from 0.25%-0.00025% were evaluated for cellular toxicity on murine macrophages (J774A.1). The effect of DS on macrophage adhesion, phagocytosis, and generation of reactive oxygen species was examined. Macrophage polarization following DS exposure was determined by gene expression using quantitative real-time polymerase chain reaction. RESULTS: Concentrations of DS >0.0025% reduced macrophage viability to <5% in exposure times as short as 30 s. Similarly, phagocytosis of Staphylococcus aureus, Pseudomonas aeruginosa, and Aspergillus flavus were significantly reduced at all tested concentrations by macrophages pretreated with DS. H2O2 production was reduced by 8%-38% following treatment with 0.00025%-0.125% DS. Macrophage adherence was significantly increased with >0.0025% DS after 15 min of exposure compared with controls. Quantitative real-time polymerase chain reaction demonstrated that DS exposure resulted in classical macrophage activation, with increased expression of inducible nitric oxide synthase 2, interferon-γ, and interleukin (IL)-1ß. CONCLUSIONS: DS at clinically used concentrations (0.025%-0.25%) was detrimental to macrophage survival and function. For optimal clinical use, understanding the impact of DS on macrophages is important as depletion may result in impaired pathogen clearance and delayed healing. These findings indicate that 0.00025% DS is a safe starting dose; however, optimal use of DS requires further validation with in vivo models.


Subject(s)
Disinfectants/pharmacology , Macrophages/cytology , Macrophages/drug effects , Sodium Hypochlorite/pharmacology , Surgical Wound Infection/drug therapy , Wound Healing/drug effects , Adult , Animals , Aspergillosis/drug therapy , Aspergillosis/immunology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Line , Cell Polarity/drug effects , Cell Polarity/immunology , Cell Survival/drug effects , Cell Survival/immunology , Female , Humans , Mice , Phagocytosis/drug effects , Phagocytosis/immunology , Pseudomonas Infections/drug therapy , Pseudomonas Infections/immunology , Solutions/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/immunology , Surgical Wound Infection/immunology , Wound Healing/immunology
20.
BMC Res Notes ; 7: 457, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25034276

ABSTRACT

BACKGROUND: Microbial biofilms have been associated with the development of chronic human infections and represent a clinical challenge given their increased antimicrobial tolerance. Staphylococcus aureus is a major human pathogen causing a diverse range of diseases, of which biofilms are often involved. Staphylococcal attachment and the formation of biofilms have been shown to be facilitated by host factors that accumulate on surfaces. To better understand how host factors enhance staphylococcal biofilm formation, we evaluated the effect of whole human plasma on biofilm formation in clinical isolates of S. aureus and the expression of seven microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) known to be involved in biofilm formation by quantitative real-time PCR. We also evaluated whether plasma augmented changes in S. aureus biofilm morphology and antimicrobial resistance. RESULTS: Exposure of clinical isolates of S. aureus to human plasma (10%) within media, and to a lesser extent when coated onto plates, significantly enhanced biofilm formation in all of the clinical isolates tested. Compared to biofilms grown under non-supplemented conditions, plasma-augmented biofilms displayed significant changes in both the biofilm phenotype and cell morphology as determined by confocal scanning laser microscopy (CLSM) and scanning electron microscopy (SEM), respectively. Exposure of bacteria to plasma resulted in a significant fold-increase in MSCRAMM expression in both a time and isolate-dependent manner. Additionally, plasma-augmented biofilms displayed an increased tolerance to vancomycin compared to biofilms grown in non-supplemented media. CONCLUSIONS: Collectively, these studies support previous findings demonstrating a role for host factors in biofilm formation and provide further insight into how plasma, a preferred growth medium for staphylococcal biofilm formation enhances as well as augments other intrinsic properties of S. aureus biofilms. Consequently, these findings indicate that incorporation of host factors may be necessary to better replicate in vivo conditions and for the best utility of a clinical biofilm assay to evaluate the process of biofilm formation and treatments.


Subject(s)
Biofilms/drug effects , Culture Media/pharmacology , Plasma/chemistry , Staphylococcus aureus/drug effects , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Biofilms/growth & development , Culture Media/chemistry , Gene Expression , Humans , Microbial Sensitivity Tests , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development , Staphylococcus aureus/ultrastructure , Vancomycin/pharmacology , Vancomycin Resistance
SELECTION OF CITATIONS
SEARCH DETAIL
...