Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Infect Control Hosp Epidemiol ; 44(11): 1760-1768, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37088696

ABSTRACT

OBJECTIVE: Although needleless connectors (NCs) are widely used in clinical practice, they carry significant risk of bloodstream infection (BSI). In this study, we quantified differences in bacterial transfer and biofilm formation between various NCs. DESIGN: Prospective, clinically simulated in vitro experimental study. METHODS: We tested 20 NCs in a 5-day clinical simulation of Staphylococcus aureus inoculations onto NC septum surfaces, which were then flushed with saline and cultured for bacterial transfer. Biofilm formation was measured through destructive sampling of the connector-catheter system. Moreover, 8 NC design factors were evaluated for their influence on bacterial transfer and biofilm formation. This study was designed without a disinfection protocol to ascertain the intrinsic risk of each NC. RESULTS: Clave Neutron and MicroClave had the lowest overall mean log density of bacteria in the flush compared to other NCs (P < .05), except there were no statistically significant differences between Clave Neutron, Microclave, SafeTouch, and SafeAccess (P ≥ .05). The amount of biofilm in the NC was positively associated with bacteria in the flush (P < .0005). Among 8 design factors, flow path was most important, with the internal cannula associated with a statistically significant 1 log reduction (LR) in bacteria in the flush (R2 = 49%) and 0.5-2 LR in the connector (R2 = 34%). All factors together best explained bacteria in the flush (R2 = 65%) and biofilm in the connector (R2 = 48%). CONCLUSIONS: Bacterial transfer and biofilm formation in the connector-catheter system varied statistically significantly between the 20 NCs, suggesting that NC choice can lower the risk of developing catheter-related BSIs.


Subject(s)
Bacteria , Disinfection , Humans , Prospective Studies , Disinfection/methods , Catheters , Biofilms
2.
Antibiotics (Basel) ; 12(3)2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36978402

ABSTRACT

Considering the prevalence and pathogenicity of biofilms in wounds, this study was designed to evaluate the anti-biofilm capabilities of eight commercially available wound care products using established in vitro assays for biofilms. The products evaluated included dressings with multiple delivery formats for ionic silver including nanocrystalline, gelling fibers, polyurethane (PU) foam, and polymer matrix. Additionally, non-silver-based products including an extracellular polymeric substance (EPS)-dissolving antimicrobial wound gel (BDWG), a collagenase-based debriding ointment and a fish skin-based skin substitute were also evaluated. The products were evaluated on Staphylococcus aureus and Pseudomonas aeruginosa mixed-species biofilms grown using colony drip flow reactor (CDFR) and standard drip flow reactor (DFR) methodologies. Anti-biofilm efficacy was measured by viable plate counts and confocal scanning laser microscopy (CSLM). Four of the eight wound care products tested were efficacious in inhibiting growth of new biofilm when compared with untreated controls. These four products were further evaluated against mature biofilms. BDWG was the only product that achieved greater than 2-log growth reduction (5.88 and 6.58 for S. aureus and P. aeruginosa, respectively) of a mature biofilm. Evaluating both biofilm prevention and mature biofilm disruption capacity is important to a comprehensive understanding of the anti-biofilm efficacy of wound care products.

3.
Antimicrob Agents Chemother ; 66(4): e0002122, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35266829

ABSTRACT

Is there a universal genetically programmed defense providing tolerance to antibiotics when bacteria grow as biofilms? A comparison between biofilms of three different bacterial species by transcriptomic and metabolomic approaches uncovered no evidence of one. Single-species biofilms of three bacterial species (Pseudomonas aeruginosa, Staphylococcus aureus, and Acinetobacter baumannii) were grown in vitro for 3 days and then challenged with respective antibiotics (ciprofloxacin, daptomycin, and tigecycline) for an additional 24 h. All three microorganisms displayed reduced susceptibility in biofilms compared to planktonic cultures. Global transcriptomic profiling of gene expression comparing biofilm to planktonic and antibiotic-treated biofilm to untreated biofilm was performed. Extracellular metabolites were measured to characterize the utilization of carbon sources between biofilms, treated biofilms, and planktonic cells. While all three bacteria exhibited a species-specific signature of stationary phase, no conserved gene, gene set, or common functional pathway could be identified that changed consistently across the three microorganisms. Across the three species, glucose consumption was increased in biofilms compared to planktonic cells, and alanine and aspartic acid utilization were decreased in biofilms compared to planktonic cells. The reasons for these changes were not readily apparent in the transcriptomes. No common shift in the utilization pattern of carbon sources was discerned when comparing untreated to antibiotic-exposed biofilms. Overall, our measurements do not support the existence of a common genetic or biochemical basis for biofilm tolerance against antibiotics. Rather, there are likely myriad genes, proteins, and metabolic pathways that influence the physiological state of individual microorganisms in biofilms and contribute to antibiotic tolerance.


Subject(s)
Anti-Bacterial Agents , Biofilms , Anti-Bacterial Agents/pharmacology , Carbon , Plankton/genetics , Pseudomonas aeruginosa/genetics , Staphylococcus aureus/genetics
4.
J Arthroplasty ; 37(7S): S647-S652, 2022 07.
Article in English | MEDLINE | ID: mdl-35210150

ABSTRACT

BACKGROUND: Numerous studies have examined the use of topical and irrigation-related adjuvants to decrease the risk of periprosthetic joint infection (PJI) after total hip arthroplasty. Many issues related to their use remain to be investigated. These include cost, antibiotic stewardship, bactericidal effect on planktonic bacteria, host cytotoxicity, necessity to irrigate/dilute potentially cytotoxic agents after their application, and impact on biofilm. METHODS: Bacterial strains of microorganisms were grown in optimal medium. After the growth phase, the organisms were exposed to the novel irrigation solution (XPerience) or phosphate buffer solution (PBS) for 5 minutes before a neutralizing broth was added. The colony-forming units per milliliter and the log reduction in colony-forming units in the treated sample vs the control were then determined. Subsequently, biofilms of microorganisms were grown on hydroxyapatite-coated glass slides. Each slide was then exposed to irrigation solutions for various contact times. Biofilm quantification was performed and the log10 density of each organism was obtained. RESULTS: In vitro testing of the irrigant demonstrated 6-log reductions in planktonic bacteria in 5 minutes, and 4-log to 8-log reductions in biofilms. Laboratory tissue testing has demonstrated minimal cytotoxic effects to host tissue allowing for solution to remain in contact with the host without need for subsequent irrigation, creating a barrier to biofilm for up to 5 hours after its application. CONCLUSION: This novel irrigant demonstrates high efficacy against both planktonic bacteria and bacterial biofilms in laboratory testing. Large series in vivo data are necessary to further establish its efficacy in reducing primary and recurrent surgical site infections.


Subject(s)
Arthroplasty, Replacement, Hip , Prosthesis-Related Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria , Biofilms , Humans , Plankton , Prosthesis-Related Infections/microbiology
5.
Orthopedics ; 45(2): 122-127, 2022.
Article in English | MEDLINE | ID: mdl-34978511

ABSTRACT

Prosthetic joint infections (PJIs) are among the most devastating complications after joint replacement. There is limited evidence regarding the efficacy of different antiseptic solutions in reducing planktonic microorganism burden. The purpose of this study was to test the efficacy of different antiseptic solutions against clinically relevant planktonic microorganisms. We designed an experiment examining the efficacy of several antiseptic solutions against clinically relevant planktonic microorganisms in vitro. Regarding planktonic microorganisms, povidone-iodine had 99.9% or greater reduction for all microorganisms tested except for methicillin-resistant Staphylococcus aureus, which was reduced by 60.44%. Irrisept (Irrimax Corp) had 99.9% or greater reduction for all microorganisms except Staphylococcus epidermidis (98.31%) and Enterococcus faecalis (48.61%). Bactisure (Zimmer Surgical Inc) had 99.9% or greater reduction for all microorganisms tested. Various measures exist for PJI prevention, one of which is intraoperative irrigation. We tested irrigants against clinically relevant planktonic microorganisms in vitro and found significant differences in efficacy among them. Further clinical outcome data are necessary to determine whether these solutions can impact PJI in vivo. [Orthopedics. 2022;45(2):122-127.].


Subject(s)
Anti-Infective Agents, Local , Methicillin-Resistant Staphylococcus aureus , Anti-Infective Agents, Local/pharmacology , Biofilms , Humans , Plankton , Povidone-Iodine
6.
Bone Joint J ; 103-B(5): 908-915, 2021 May.
Article in English | MEDLINE | ID: mdl-33934664

ABSTRACT

AIMS: Periprosthetic joint infections (PJIs) are among the most devastating complications after joint arthroplasty. There is limited evidence on the efficacy of different antiseptic solutions on reducing biofilm burden. The purpose of the present study was to test the efficacy of different antiseptic solutions against clinically relevant microorganisms in biofilm. METHODS: We conducted an in vitro study examining the efficacy of several antiseptic solutions against clinically relevant microorganisms. We tested antiseptic irrigants against nascent (four-hour) and mature (three-day) single-species biofilm created in vitro using a drip-flow reactor model. RESULTS: With regard to irrigant efficacy against biofilms, Povidone-iodine treatment resulted in greater reductions in nascent MRSA biofilms (logarithmic reduction (LR) = 3.12; p < 0.001) compared to other solutions. Bactisure treatment had the greatest reduction of mature Pseudomonas aeruginosa biofilms (LR = 1.94; p = 0.032) and a larger reduction than Vashe or Irrisept for mature Staphylococcus epidermidis biofilms (LR = 2.12; p = 0.025). Pooled data for all biofilms tested resulted in Bactisure and Povidone-iodine with significantly greater reductions compared to Vashe, Prontosan, and Irrisept solutions (p < 0.001). CONCLUSION: Treatment failure in PJI is often due to failure to clear the biofilm; antiseptics are often used as an adjunct to biofilm clearance. We tested irrigants against clinically relevant microorganisms in biofilm in vitro and showed significant differences in efficacy among the different solutions. Further clinical outcome data is necessary to determine whether these solutions can impact PJI outcome in vivo. Cite this article: Bone Joint J 2021;103-B(5):908-915.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Infective Agents, Local/pharmacology , Arthroplasty, Replacement , Biofilms/drug effects , Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/prevention & control , Acetic Acid/pharmacology , Benzalkonium Compounds/pharmacology , Betaine/pharmacology , Biguanides/pharmacology , Chlorhexidine/analogs & derivatives , Chlorhexidine/pharmacology , Hydrogen Peroxide/pharmacology , In Vitro Techniques , Methicillin-Resistant Staphylococcus aureus/drug effects , Povidone-Iodine/pharmacology , Propionibacteriaceae/drug effects , Pseudomonas aeruginosa/drug effects , Sodium Hypochlorite/pharmacology , Staphylococcus epidermidis/drug effects
7.
Int J Dermatol ; 60(5): 589-596, 2021 May.
Article in English | MEDLINE | ID: mdl-33615460

ABSTRACT

BACKGROUND: The ability of Cutibacterium acnes strains to form biofilms has been correlated with their virulence. OBJECTIVE: This study examined biofilm and skin microbiota in acne patients in order to understand their role in the development of acne lesions. METHODS: Thin sections of punch biopsy specimens of (i) uninflamed comedones, (ii) inflammatory lesions, and (iii) uninvolved adjacent skin of acne patients were examined. Epiflourescence and confocal laser scanning microscopy were used for biofilm detection, and pyrosequencing with taxonomic classification of 16s rRNA gene amplicons was used for microbiota analysis. RESULTS: Of the 39 skin specimens from patients with mild-moderate acne (n = 13) that were studied, nine (23%) contained biofilm. Among these specimens, biofilm was most frequently detected in comedones (55.6%) and less frequently in inflammatory papules (22.2%) and uninvolved skin (22.2%). Comedones demonstrated the highest mean alpha diversity of all the lesion subtypes. The relative abundance of Staphylococcus was significantly higher in comedones (11.400% ± 12.242%) compared to uninvolved skin (0.073% ± 0.185%, P = 0.024). CONCLUSIONS: The microenvironment of the comedone differs from that of inflammatory lesions and unaffected skin. The increased frequency of biofilm in comedones may account for the lack of host inflammatory response to these lesions.


Subject(s)
Acne Vulgaris , Microbiota , Biofilms , Humans , Propionibacterium acnes , RNA, Ribosomal, 16S/genetics
8.
Front Microbiol ; 11: 565914, 2020.
Article in English | MEDLINE | ID: mdl-33013797

ABSTRACT

One of the major causes of prosthetic joint failure is infection. Recently, coagulase negative Staphylococcus epidermidis has been identified as an emergent, nosocomial pathogen involved in subclinical prosthetic joint infections (PJIs). The diagnosis of PJIs mediated by S. epidermidis is usually complex and difficult due to the absence of acute clinical signs derived from the host immune system response. Therefore, analysis of protein patterns in biofilm-producing S. epidermidis allows for the examination of the molecular basis of biofilm formation. Thus, in the present study, the proteome of a clinical isolate S. epidermidis was analyzed when cultured in its planktonic or sessile form to examine protein expression changes depending on culture conditions. After 24 h of culture, sessile bacteria exhibited increased gene expression for ribosomal activity and for production of proteins related to the initial attachment phase, involved in the capsular polysaccharide/adhesin, surface associated proteins and peptidoglycan biosynthesis. Likewise, planktonic S. epidermidis was able to aggregate after 24 h, synthesizing the accumulation associate protein and cell-wall molecules through the activation of the YycFG and ArlRS, two component regulatory pathways. Prolonged culture under vigorous agitation generated a stressful growing environment triggering aggregation in a biofilm-like matrix as a mechanism to survive harsh conditions. Further studies will be essential to support these findings in order to further delineate the complex mechanisms of biofilm formation of S. epidermidis and they could provide the groundwork for the development of new drugs against biofilm-related infections, as well as the identification of novel biomarkers of subclinical or chronic infections mediated by these emerging, low virulence pathogens.

10.
Microorganisms ; 8(2)2020 Jan 31.
Article in English | MEDLINE | ID: mdl-32023892

ABSTRACT

The study of bacterial interaction between Streptococcus mutans and Actinomyces naeslundii may disclose important features of biofilm interspecies relationships. The aim of this study was to characterize-with an emphasis on biofilm formation and composition and metabolic activity-single- and dual-species biofilms of S. mutans or A. naeslundii, and to use a drip flow reactor (DFR) to evaluate biofilm stress responses to 0.2% chlorhexidine diacetate (CHX). Single- and dual-species biofilms were grown for 24 h. The following factors were evaluated: cell viability, biomass and total proteins in the extracellular matrix, 2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide-"XTT"-reduction and lactic acid production. To evaluate stress response, biofilms were grown in DFR. Biofilms were treated with CHX or 0.9% sodium chloride (NaCl; control). Biofilms were plated for viability assessment. Confocal laser-scanning microscopy (CLSM) was also performed. Data analysis was carried out at 5% significance level. S. mutans viability and lactic acid production in dual-species biofilms were significantly reduced. S. mutans showed a higher resistance to CHX in dual-species biofilms. Total protein content, biomass and XTT reduction showed no significant differences between single- and dual-species biofilms. CLSM images showed the formation of large clusters in dual-species biofilms. In conclusion, dual-species biofilms reduced S. mutans viability and lactic acid production and increased S. mutans' resistance to chlorhexidine.

11.
J Bacteriol ; 201(22)2019 11 15.
Article in English | MEDLINE | ID: mdl-31501280

ABSTRACT

Transcriptomic, metabolomic, physiological, and computational modeling approaches were integrated to gain insight into the mechanisms of antibiotic tolerance in an in vitro biofilm system. Pseudomonas aeruginosa biofilms were grown in drip flow reactors on a medium composed to mimic the exudate from a chronic wound. After 4 days, the biofilm was 114 µm thick with 9.45 log10 CFU cm-2 These biofilms exhibited tolerance, relative to exponential-phase planktonic cells, to subsequent treatment with ciprofloxacin. The specific growth rate of the biofilm was estimated via elemental balances to be approximately 0.37 h-1 and with a reaction-diffusion model to be 0.32 h-1, or one-third of the maximum specific growth rate for planktonic cells. Global analysis of gene expression indicated lower transcription of ribosomal genes and genes for other anabolic functions in biofilms than in exponential-phase planktonic cells and revealed the induction of multiple stress responses in biofilm cells, including those associated with growth arrest, zinc limitation, hypoxia, and acyl-homoserine lactone quorum sensing. Metabolic pathways for phenazine biosynthesis and denitrification were transcriptionally activated in biofilms. A customized reaction-diffusion model predicted that steep oxygen concentration gradients will form when these biofilms are thicker than about 40 µm. Mutant strains that were deficient in Psl polysaccharide synthesis, the stringent response, the stationary-phase response, and the membrane stress response exhibited increased ciprofloxacin susceptibility when cultured in biofilms. These results support a sequence of phenomena leading to biofilm antibiotic tolerance, involving oxygen limitation, electron acceptor starvation and growth arrest, induction of associated stress responses, and differentiation into protected cell states.IMPORTANCE Bacteria in biofilms are protected from killing by antibiotics, and this reduced susceptibility contributes to the persistence of infections such as those in the cystic fibrosis lung and chronic wounds. A generalized conceptual model of biofilm antimicrobial tolerance with the following mechanistic steps is proposed: (i) establishment of concentration gradients in metabolic substrates and products; (ii) active biological responses to these changes in the local chemical microenvironment; (iii) entry of biofilm cells into a spectrum of states involving alternative metabolisms, stress responses, slow growth, cessation of growth, or dormancy (all prior to antibiotic treatment); (iv) adaptive responses to antibiotic exposure; and (v) reduced susceptibility of microbial cells to antimicrobial challenges in some of the physiological states accessed through these changes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Drug Resistance, Bacterial , Pseudomonas aeruginosa/drug effects , Ciprofloxacin/pharmacology , Diffusion , Drug Resistance, Bacterial/genetics , Gene Expression , Models, Biological , Oxygen/metabolism , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/physiology , Stress, Physiological
12.
Aesthetic Plast Surg ; 43(2): 490-497, 2019 04.
Article in English | MEDLINE | ID: mdl-30276456

ABSTRACT

BACKGROUND: Bacterial biofilms have been implicated with breast implant complications including capsular contracture and anaplastic large-cell lymphoma. The actual mechanisms for either are still under active investigation and are not clear. Due to their increased surface area, implants with textured surfaces may harbor greater biofilm loads than those with smooth surfaces. METHODS: Biofilm formation on the outer surface material was compared using implants with various surface areas and roughness, including Natrelle® (Smooth), SmoothSilk®/SilkSurface® (Silk), VelvetSurface ® (Velvet), Siltex®, and Biocell®. The roughness and surface area of each material were assessed using non-contact profilometry. Bacterial attachment (2 h) and biofilm formation (24 h) were evaluated for Staphylococcus epidermidis, Pseudomonas aeruginosa, and Ralstonia pickettii over nine independent experiments using a CDC biofilm reactor and viable plate counts (VPCs) as well as confocal scanning laser microscopy. VPCs of the textured implants were compared relative to the Smooth implant. RESULTS: Surface areas increased with roughness and were similar among the three least rough implants (Smooth, Silk, and Velvet) and among the roughest implants (Siltex and Biocell). Overall, VPC indicated there was significantly more bacterial attachment and biofilm formation on the Siltex and Biocell implants than the Silk or Velvet implants, although there were differences between species and time points. CSLM confirmed the formation of thicker biofilms on the implants with rougher surface textures. CONCLUSION: This in vitro study confirmed that implant surfaces with rougher texture, resulting in more surface area, harbored greater biofilm loads than those with smoother surfaces. NO LEVEL ASSIGNED: This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Subject(s)
Bacterial Adhesion , Biofilms , Breast Implants/microbiology , Pseudomonas aeruginosa/physiology , Ralstonia pickettii/physiology , Staphylococcus epidermidis/physiology , Prosthesis Design
13.
J Pharm Biomed Anal ; 152: 1-11, 2018 Apr 15.
Article in English | MEDLINE | ID: mdl-29413999

ABSTRACT

This work explains the motivation, advantages, and novel approach of using velocity magnetic resonance imaging (MRI) for studying the hydrodynamics in a complicated structural biomedical device such as an intravenous catheter needleless connector (NC). MRI was applied as a non-invasive and non-destructive technique to evaluate the fluid dynamics associated with various internal designs of the NC. Spatial velocity maps of fluid flow at specific locations within these medical devices were acquired. Dynamic MRI is demonstrated as an effective method to quantify flow patterns and fluid dynamic dependence on structural features of NCs. These spatial velocity maps could be used as a basis for groundtruthing computational fluid dynamics (CFD) methods that could impact the design of NCs.


Subject(s)
Equipment and Supplies , Catheters , Hydrodynamics , Magnetic Resonance Spectroscopy/methods
14.
J Antimicrob Chemother ; 73(1): 102-108, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29029221

ABSTRACT

BACKGROUND: Clostridium difficile, a spore-forming Gram-positive anaerobic bacillus, is the most common causative agent of healthcare-associated diarrhoea. Formation of biofilms may protect C. difficile against antibiotics, potentially leading to treatment failure. Furthermore, bacterial spores or vegetative cells may linger in biofilms in the gut causing C. difficile infection recurrence. OBJECTIVES: In this study, we evaluated and compared the efficacy of four antibiotics (fidaxomicin, surotomycin, vancomycin and metronidazole) in penetrating C. difficile biofilms and killing vegetative cells. METHODS: C. difficile biofilms grown initially for 48 or 72 h using the colony biofilm model were then treated with antibiotics at a concentration of 25 × MIC for 24 h. Vegetative cells and spores were enumerated. The effect of treatment on biofilm structure was studied by scanning electron microscopy (SEM). The ability of fidaxomicin and surotomycin to penetrate biofilms was studied using fluorescently tagged antibiotics. RESULTS: Both surotomycin and fidaxomicin were significantly more effective than vancomycin or metronidazole (P < 0.001) at killing vegetative cells in established biofilms. Fidaxomicin was more effective than metronidazole at reducing viable spore counts in biofilms (P < 0.05). Fluorescently labelled surotomycin and fidaxomicin penetrated C. difficile biofilms in < 1 h. After 24 h of treatment, SEM demonstrated that both fidaxomicin and surotomycin disrupted the biofilm structure, while metronidazole had no observable effect. CONCLUSIONS: Fidaxomicin is effective in disrupting C. difficile biofilms, killing vegetative cells and decreasing spore counts.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Clostridioides difficile/drug effects , Lipopeptides/pharmacology , Metronidazole/pharmacology , Peptides, Cyclic/pharmacology , Vancomycin/pharmacology , Biological Transport/physiology , Clostridioides difficile/growth & development , Colony Count, Microbial , Fidaxomicin , Humans , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Spores, Bacterial/drug effects
15.
Anal Biochem ; 539: 144-148, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29107579

ABSTRACT

Monitoring patients with burn wounds for infection is standard practice because failure to rapidly and specifically identify a pathogen can result in poor clinical outcomes, including death. Therefore, a method that facilitates detection and identification of pathogens in situ within minutes of biopsy would be a significant benefit to clinicians. Mass spectrometry is rapidly becoming a standard tool in clinical settings, capable of identifying specific pathogens from complex samples. Imaging mass spectrometry (IMS) expands the information content by enabling spatial resolution of biomarkers in tissue samples as in histology, without the need for specific stains/antibodies. Herein, a murine model of thermal injury was used to study infection of burn tissue by Pseudomonas aeruginosa. This is the first use of IMS to detect P. aeruginosa infection in situ from thermally injured tissue. Multiple molecular features could be spatially resolved to infected or uninfected tissue. This demonstrates the potential use of IMS in a clinical setting to aid doctors in identifying both presence and species of pathogens in tissue.


Subject(s)
Biomarkers/analysis , Burns/microbiology , Pseudomonas aeruginosa/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Animals , Burns/complications , Burns/pathology , Carboxymethylcellulose Sodium/chemistry , Disease Models, Animal , Gelatin/chemistry , Mice , Optical Imaging , Pseudomonas Infections/complications , Pseudomonas Infections/microbiology
16.
Wound Repair Regen ; 25(5): 744-757, 2017 09.
Article in English | MEDLINE | ID: mdl-28960634

ABSTRACT

BACKGROUND: Despite a growing consensus that biofilms contribute to a delay in the healing of chronic wounds, conflicting evidence pertaining to their identification and management can lead to uncertainty regarding treatment. This, in part, has been driven by reliance on in vitro data or animal models, which may not directly correlate to clinical evidence on the importance of biofilms. Limited data presented in human studies have further contributed to the uncertainty. Guidelines for care of chronic wounds with a focus on biofilms are needed to help aid the identification and management of biofilms, providing a clinical focus to support clinicians in improving patient care through evidence-based medicine. METHODS: A Global Wound Biofilm Expert Panel, comprising 10 clinicians and researchers with expertise in laboratory and clinical aspects of biofilms, was identified and convened. A modified Delphi process, based on published scientific data and expert opinion, was used to develop consensus statements that could help identify and treat biofilms as part of the management of chronic nonhealing wounds. Using an electronic survey, panel members rated their agreement with statements about biofilm identification and treatment, and the management of chronic nonhealing wounds. Final consensus statements were agreed on in a face-to-face meeting. RESULTS: Participants reached consensus on 61 statements in the following topic areas: understanding biofilms and the problems they cause clinicians; current diagnostic options; clinical indicators of biofilms; future options for diagnostic tests; treatment strategies; mechanical debridement; topical antiseptics; screening antibiofilm agents; and levels of evidence when choosing antibiofilm treatments. CONCLUSION: This consensus document attempts to clarify misunderstandings about the role of biofilms in clinical practice, and provides a basis for clinicians to recognize biofilms in chronic nonhealing wounds and manage patients optimally. A new paradigm for wound care, based on a stepped-down treatment approach, was derived from the consensus statements.


Subject(s)
Anti-Infective Agents, Local/administration & dosage , Biofilms , Consensus , Wound Healing/physiology , Wound Infection/therapy , Administration, Topical , Animals , Chronic Disease , Humans
17.
PLoS One ; 12(4): e0174518, 2017.
Article in English | MEDLINE | ID: mdl-28369127

ABSTRACT

BACKGROUND: In previous studies, Propionibacterium acnes was cultured from intervertebral disc tissue of ~25% of patients undergoing microdiscectomy, suggesting a possible link between chronic bacterial infection and disc degeneration. However, given the prominence of P. acnes as a skin commensal, such analyses often struggled to exclude the alternate possibility that these organisms represent perioperative microbiologic contamination. This investigation seeks to validate P. acnes prevalence in resected disc cultures, while providing microscopic evidence of P. acnes biofilm in the intervertebral discs. METHODS: Specimens from 368 patients undergoing microdiscectomy for disc herniation were divided into several fragments, one being homogenized, subjected to quantitative anaerobic culture, and assessed for bacterial growth, and a second fragment frozen for additional analyses. Colonies were identified by MALDI-TOF mass spectrometry and P. acnes phylotyping was conducted by multiplex PCR. For a sub-set of specimens, bacteria localization within the disc was assessed by microscopy using confocal laser scanning and FISH. RESULTS: Bacteria were cultured from 162 discs (44%), including 119 cases (32.3%) with P. acnes. In 89 cases, P. acnes was cultured exclusively; in 30 cases, it was isolated in combination with other bacteria (primarily coagulase-negative Staphylococcus spp.) Among positive specimens, the median P. acnes bacterial burden was 350 CFU/g (12 - ~20,000 CFU/g). Thirty-eight P. acnes isolates were subjected to molecular sub-typing, identifying 4 of 6 defined phylogroups: IA1, IB, IC, and II. Eight culture-positive specimens were evaluated by fluorescence microscopy and revealed P. acnes in situ. Notably, these bacteria demonstrated a biofilm distribution within the disc matrix. P. acnes bacteria were more prevalent in males than females (39% vs. 23%, p = 0.0013). CONCLUSIONS: This study confirms that P. acnes is prevalent in herniated disc tissue. Moreover, it provides the first visual evidence of P. acnes biofilms within such specimens, consistent with infection rather than microbiologic contamination.


Subject(s)
Biofilms/growth & development , Intervertebral Disc Displacement/microbiology , Intervertebral Disc/microbiology , Propionibacterium acnes/isolation & purification , Propionibacterium acnes/physiology , Adult , Aged , Aged, 80 and over , Diskectomy , Female , Gram-Positive Bacterial Infections/complications , Gram-Positive Bacterial Infections/microbiology , Humans , Intervertebral Disc/surgery , Intervertebral Disc Degeneration/etiology , Intervertebral Disc Degeneration/microbiology , Intervertebral Disc Displacement/etiology , Intervertebral Disc Displacement/surgery , Male , Middle Aged , Phenotype , Propionibacterium acnes/pathogenicity , Young Adult
18.
APMIS ; 125(4): 344-352, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28407431

ABSTRACT

Chronic wounds are characterized as wounds that have failed to proceed through the well-orchestrated healing process and have remained open for months to years. Open wounds are at risk for colonization by opportunistic pathogens. Bacteria that colonize the open wound bed form surface-attached, multicellular communities called biofilms, and chronic wound biofilms can contain a diverse microbiota. Investigators are just beginning to elucidate the role of biofilms in chronic wound pathogenesis, and have simplified the complex wound environment using in vitro models to obtain a fundamental understanding of the impact of biofilms on wound-healing cell types. The intent of this review is to describe current in vitro methodologies and their results. Investigations started with one host cell-type and single species biofilms and demonstrated that biofilms, or their secretions, had deleterious effects on wound-healing cells. More complex systems involved the use of multiple host cell/tissue types and single species biofilms. Using human skin-equivalent tissues, investigators demonstrated that a number of different species can grow on the tissue and elicit an inflammatory response from the tissue. A full understanding of how biofilms impact wound-healing cells and host tissues will have a profound effect on how chronic wounds are treated.


Subject(s)
Bacterial Physiological Phenomena , Biofilms , Wound Infection/microbiology , Animals , Bacteria/genetics , Bacteria/isolation & purification , Humans , Wound Healing , Wound Infection/physiopathology
19.
Wound Repair Regen ; 24(2): 373-83, 2016 03.
Article in English | MEDLINE | ID: mdl-26748963

ABSTRACT

Biofilms have been implicated in delayed wound healing, although the mechanisms by which biofilms impair wound healing are poorly understood. Many species of bacteria produce exotoxins and exoenzymes that may inhibit healing. In addition, oxygen consumption by biofilms and by the responding leukocytes, may impede wound healing by depleting the oxygen that is required for healing. In this study, oxygen microsensors to measure oxygen transects through in vitro cultured biofilms, biofilms formed in vivo within scabs from a diabetic (db/db) mouse wound model, and ex vivo human chronic wound specimens was used. The results showed that oxygen levels within mouse scabs had steep gradients that reached minima ranging from 17 to 72 mmHg on live mice and from 6.4 to 1.1 mmHg on euthanized mice. The oxygen gradients in the mouse scabs were similar to those observed for clinical isolates cultured in vitro and for human ex vivo specimens. To characterize the metabolic activities of the bacteria in the mouse scabs, transcriptomics analyses of Pseudomonas aeruginosa biofilms associated with the db/db mice wounds was performed. The results demonstrated that the bacteria expressed genes for metabolic activities associated with cell growth. Interestingly, the transcriptome results also indicated that the bacteria within the wounds experienced oxygen-limitation stress. Among the bacterial genes that were expressed in vivo were genes associated with the Anr-mediated hypoxia-stress response. Other bacterial stress response genes highly expressed in vivo were genes associated with stationary-phase growth, osmotic stress, and RpoH-mediated heat shock stress. Overall, the results supported the hypothesis that bacterial biofilms in chronic wounds promote chronicity by contributing to the maintenance of localized low oxygen tensions, through their metabolic activities and through their recruitment of cells that consume oxygen for host defensive processes.


Subject(s)
Biofilms/growth & development , Biosensing Techniques , Diabetes Mellitus, Experimental/metabolism , Oxygen/metabolism , Pseudomonas Infections/microbiology , Transcriptome/physiology , Wound Infection/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Humans , Mice , Osmotic Pressure , Pseudomonas Infections/pathology , Wound Healing/physiology , Wound Infection/pathology
20.
Int J Antimicrob Agents ; 46(4): 451-5, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26213381

ABSTRACT

Due to the emergence of staphylococcal biofilm infections, the need for advanced antibiotics is crucial. The aim of this investigation was to evaluate the potency and penetration of telavancin against staphylococcal biofilms using two different biofilm models. Multiple staphylococcal strains, including meticillin-sensitive Staphylococcus aureus ATCC 29213, vancomycin-intermediate S. aureus ATCC 700787, heterogeneously vancomycin-intermediate S. aureus ATCC 700698 and meticillin-sensitive Staphylococcus epidermidis ATCC 12228, were grown and treated in drip-flow reactors to determine log reductions due to telavancin treatment. After 3 days of biofilm growth and 24 h of treatment, mean log reductions for telavancin ranged from 1.65 to 2.17 depending on the bacterial strain tested. Penetration was evaluated qualitatively using confocal scanning laser microscopy to image the infiltration of fluorescently labelled antibiotic into a staphylococcal biofilm grown in a flow cell. Fluorescently labelled telavancin rapidly penetrated the biofilms with no alteration in the biofilm structure.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Staphylococcus aureus/drug effects , Staphylococcus epidermidis/drug effects , Humans , Lipoglycopeptides , Staphylococcus aureus/physiology , Staphylococcus epidermidis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...