Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.279
1.
Ann Clin Microbiol Antimicrob ; 23(1): 39, 2024 May 03.
Article En | MEDLINE | ID: mdl-38702796

BACKGROUND: Non-surgical chronic wounds, including diabetes-related foot diseases (DRFD), pressure injuries (PIs) and venous leg ulcers (VLU), are common hard-to-heal wounds. Wound evolution partly depends on microbial colonisation or infection, which is often confused by clinicians, thereby hampering proper management. Current routine microbiology investigation of these wounds is based on in vitro culture, focusing only on a limited panel of the most frequently isolated bacteria, leaving a large part of the wound microbiome undocumented. METHODS: A literature search was conducted on original studies published through October 2022 reporting metagenomic next generation sequencing (mNGS) of chronic wound samples. Studies were eligible for inclusion if they applied 16 S rRNA metagenomics or shotgun metagenomics for microbiome analysis or diagnosis. Case reports, prospective, or retrospective studies were included. However, review articles, animal studies, in vitro model optimisation, benchmarking, treatment optimisation studies, and non-clinical studies were excluded. Articles were identified in PubMed, Google Scholar, Web of Science, Microsoft Academic, Crossref and Semantic Scholar databases. RESULTS: Of the 3,202 articles found in the initial search, 2,336 articles were removed after deduplication and 834 articles following title and abstract screening. A further 14 were removed after full text reading, with 18 articles finally included. Data were provided for 3,628 patients, including 1,535 DRFDs, 956 VLUs, and 791 PIs, with 164 microbial genera and 116 species identified using mNGS approaches. A high microbial diversity was observed depending on the geographical location and wound evolution. Clinically infected wounds were the most diverse, possibly due to a widespread colonisation by pathogenic bacteria from body and environmental microbiota. mNGS data identified the presence of virus (EBV) and fungi (Candida and Aspergillus species), as well as Staphylococcus and Pseudomonas bacteriophages. CONCLUSION: This study highlighted the benefit of mNGS for time-effective pathogen genome detection. Despite the majority of the included studies investigating only 16 S rDNA, ignoring a part of viral, fungal and parasite colonisation, mNGS detected a large number of bacteria through the included studies. Such technology could be implemented in routine microbiology for hard-to-heal wound microbiota investigation and post-treatment wound colonisation surveillance.


Bacteria , High-Throughput Nucleotide Sequencing , Metagenomics , Humans , Metagenomics/methods , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/classification , Wound Healing , Microbiota/genetics , Pressure Ulcer/microbiology , Diabetic Foot/microbiology , Wound Infection/microbiology , Varicose Ulcer/microbiology
2.
Carbohydr Polym ; 337: 122147, 2024 Aug 01.
Article En | MEDLINE | ID: mdl-38710554

Treatment of infected wound by simultaneously eliminating bacteria and inducing angiogenesis to promote wound tissue regeneration remains a clinical challenge. Dynamic and reversable hydrogels can adapt to irregular wound beds, which have raised great attention as wound dressings. Herein, a sprayable chitosan-based hydrogel (HPC/CCS/ODex-IGF1) was developed using hydroxypropyl chitosan (HPC), caffeic acid functionalized chitosan (CCS), oxidized dextran (ODex) to crosslink through the dynamic imine bond, which was pH-responsive to the acidic microenvironment and could controllably release insulin growth factor-1 (IGF1). The HPC/CCS/ODex-IGF1 hydrogels not only showed self-healing, self-adaptable and sprayable properties, but also exhibited excellent antibacterial ability, antioxidant property, low-cytotoxicity and angiogenetic activity. In vivo experiments demonstrated that hydrogels promoted tissue regeneration and healing of bacteria-infected wound with a rate of approximately 98.4 % on day 11 by eliminating bacteria, reducing inflammatory and facilitating angiogenesis, demonstrating its great potential for wound dressing.


Anti-Bacterial Agents , Chitosan , Hydrogels , Neovascularization, Physiologic , Wound Healing , Chitosan/chemistry , Chitosan/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Wound Healing/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Mice , Neovascularization, Physiologic/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Humans , Male , Insulin-Like Growth Factor I , Staphylococcus aureus/drug effects , Bandages , Wound Infection/drug therapy , Wound Infection/microbiology , Dextrans/chemistry , Dextrans/pharmacology , Angiogenesis
3.
PLoS One ; 19(5): e0304491, 2024.
Article En | MEDLINE | ID: mdl-38805522

Due to high tolerance to antibiotics and pronounced virulence, bacterial biofilms are considered a key factor and major clinical challenge in persistent wound infections. They are typically composed of multiple species, whose interactions determine the biofilm's structural development, functional properties and thus the progression of wound infections. However, most attempts to study bacterial biofilms in vitro solely rely on mono-species populations, since cultivating multi-species biofilms, especially for prolonged periods of time, poses significant challenges. To address this, the present study examined the influence of bacterial composition on structural biofilm development, morphology and spatial organization, as well as antibiotic tolerance and virulence on human skin cells in the context of persistent wound infections. By creating a wound-mimetic microenvironment, the successful cultivation of dual-species biofilms of two of the most prevalent wound pathogens, Pseudomonas aeruginosa and Staphylococcus aureus, was realized over a period of 72 h. Combining quantitative analysis with electron microscopy and label-free imaging enabled a comprehensive evaluation of the dynamics of biofilm formation and matrix secretion, revealing a twofold increased maturation of dual-species biofilms. Antibiotic tolerance was comparable for both mono-species cultures, however, dual-species communities showed a 50% increase in tolerance, mediated by a significantly reduced penetration of the applied antibiotic into the biofilm matrix. Further synergistic effects were observed, where dual-species biofilms exacerbated wound healing beyond the effects observed from either Pseudomonas or Staphylococcus. Consequently, predicting biofilm development, antimicrobial tolerance and virulence for multi-species biofilms based solely on the results from mono-species biofilms is unreliable. This study underscores the substantial impact of a multi-species composition on biofilm functional properties and emphasizes the need to tailor future studies reflecting the bacterial composition of the respective in vivo situation, leading to a more comprehensive understanding of microbial communities in the context of basic microbiology and the development of effective treatments.


Anti-Bacterial Agents , Biofilms , Pseudomonas aeruginosa , Staphylococcus aureus , Wound Infection , Biofilms/drug effects , Biofilms/growth & development , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Pseudomonas aeruginosa/pathogenicity , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Humans , Virulence/drug effects , Wound Infection/microbiology , Wound Infection/drug therapy , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Microbial Sensitivity Tests , Pseudomonas Infections/microbiology , Pseudomonas Infections/drug therapy
4.
Photobiomodul Photomed Laser Surg ; 42(5): 356-365, 2024 May.
Article En | MEDLINE | ID: mdl-38776546

Background: Nosocomial wound infection with Pseudomonas aeruginosa (PA) is a serious complication often responsible for the septic mortality of burn patients. Objective: High-intensity antimicrobial blue light (aBL) treatment may represent an alternative therapy for PA infections and will be investigated in this study. Methods: Antibacterial effects of a light-emitting diode array (450-460 nm; 300 mW/cm2; 15/30 min; 270/540 J/cm2) against PA were determined by suspension assay, biofilm assay, and a human skin wound model and compared with 15-min topically applied 3% citric acid (CA) and wound irrigation solution (Prontosan®; PRT). Results: aBL reduced the bacterial number [2.51-3.56 log10 colony-forming unit (CFU)/mL], whereas PRT or CA treatment achieved a 4.64 or 6.60 log10 CFU/mL reduction in suspension assays. aBL reduced biofilm formation by 60-66%. PRT or CA treatment showed reductions by 25% or 13%. Here, aBL reduced bacterial number in biofilms (1.30-1.64 log10 CFU), but to a lower extend than PRT (2.41 log10 CFU) or CA (2.48 log10 CFU). In the wound skin model, aBL (2.21-2.33 log10 CFU) showed a bacterial reduction of the same magnitude as PRT (2.26 log10 CFU) and CA (2.30 log10 CFU). Conclusions: aBL showed a significant antibacterial efficacy against PA and biofilm formation in a short time. However, a clinical application of aBL in wound therapy requires effective active skin cooling and eye protection, which in turn may limit clinical implementation.


Biofilms , Pseudomonas Infections , Pseudomonas aeruginosa , Wound Infection , Humans , Pseudomonas aeruginosa/radiation effects , Biofilms/radiation effects , Pseudomonas Infections/therapy , Pseudomonas Infections/radiotherapy , Wound Infection/therapy , Wound Infection/microbiology , Phototherapy , Blue Light
5.
BMC Microbiol ; 24(1): 166, 2024 May 16.
Article En | MEDLINE | ID: mdl-38755533

BACKGROUND: Bullet-related bacterial wound infection can be caused by high-velocity bullets and shrapnel injuries. In Ethiopia, significant injuries were reported that may cause severe wound infections, persistent systemic infections and may lead to amputation and mortality. The magnitude, antimicrobial susceptibility profiles, and factors associated with bacterial wound infections among patients with bullet-related injuries are not yet studied particularly at health facilities in Bahir Dar, Northwest Ethiopia. Therefore, this study was aimed to determine the prevalence, bacterial profiles, antimicrobial susceptibility profiles, and factors associated with bacterial infections among patients with bullet-related injuries at referral health facilities in Bahir Dar, Northwest Ethiopia. METHODS: A Hospital-based cross-sectional study was conducted among patients with bullet-related injuries at three referral health facilities in Bahir Dar from May 25 to July 27, 2022. A total of 384 patients with bullet-related injuries were included in the study. Sociodemographic and clinical data were collected using a structured questionnaire. Wound swabs were collected aseptically and cultured on Blood and MacConkey agar following bacteriological standards. Biochemical tests were performed to differentiate bacteria for positive cultivation and antimicrobial susceptibility profiles of the isolates were done on Muller Hinton agar using the Kirby-Bauer disk diffusion technique according to the 2021 Clinical Laboratory Standard Institute (CLSI) guideline. The data were entered using Epi-Info version 7.3 and analyzed using SPSS version 25. Descriptive data were presented using frequency, percentages, figures, and charts. Logistic regression was carried out to identify factors associated with bacterial wound infections. P-value < 0.05 was considered statistically significant. RESULTS: The prevalence of bullet-related bacterial wound infection among three referral hospitals in Bahir Dar city was 54.7%. The most commonly isolated Gram-negative organism was Klebsiella spps 49 (23.3%) while among Gram-positive organism, Staphylococcus aureus 58 (27.6%) and coagulase-negative staphylococci (CONS) 18 (8.6%). Contamination, hospitalization and smoking habit were significantly associated with the presence of bullet-related bacterial wound infections. Over 97% multidrug resistant (MDR) bacterial isolates were identified and of theses, E. coli, Proteus species, Citrobactor, and Staphylococcus aureus were highly drug resistant. CONCLUSION: Increased prevalence of bullet-related bacterial wound infection was noticed in this study. S. aureus followed by Klebsiella species were most commonly isolated bacteria. High frequency of resistance to Ampicillin, Oxacillin, Cefepime, Ceftriaxone, Ceftazidime, Vancomycin, and Norfloxacin was observed. Therefore, proper handling of bullet injuries, prompt investigation of bacterial infections, monitoring of drug sensitivity patterns and antibiotic usage are critical.


Anti-Bacterial Agents , Microbial Sensitivity Tests , Wound Infection , Humans , Ethiopia/epidemiology , Male , Cross-Sectional Studies , Adult , Female , Prevalence , Wound Infection/microbiology , Wound Infection/epidemiology , Anti-Bacterial Agents/pharmacology , Young Adult , Wounds, Gunshot/epidemiology , Wounds, Gunshot/microbiology , Bacterial Infections/epidemiology , Bacterial Infections/microbiology , Bacterial Infections/drug therapy , Middle Aged , Bacteria/drug effects , Bacteria/isolation & purification , Bacteria/classification , Emergency Service, Hospital/statistics & numerical data , Adolescent
6.
PLoS One ; 19(5): e0301201, 2024.
Article En | MEDLINE | ID: mdl-38743750

With the rise of AMR the management of wound infections are becoming a big challenge. This has been attributed to the fact that most wound bacterial isolates have been found to possess various virulence factors like enzymes, toxins & biofilms production. Therefore, need for discovery of new lead compounds is paramount as such factors make these microbes to be resistant to already existing arsenal of antibiotics or even the immune system. This study aimed at documenting the nutritional, physicochemical, phytochemical and antibacterial properties of stingless bee honey. Isolation and characterization of bacterial isolates from 34 samples obtained from wounds of outpatients and surgical wards of Nakuru County Referral Hospital, Kenya was done. Various bacterial isolates (43) were isolated Staphylococcus aureus (34.8%) being predominant, followed by Pseudomonas aeruginosa (27.9%), Klebsiella pneumoniae (23.3%) and Escherichia coli (14.0%). A total of 36 out of the total isolates were genotypically characterized using molecular techniques detecting the prevalence of the following virulence genes; 16 srRNA (756 bp), hla (229 bp), cnf1 (426 bp), cnf2 (543 bp), hlyA (1011 bp), rmpA (461 bp), lasL (600 bp), gyrB (411 bp), khe (77 bp) and magA (128 bp). An assessment of the in vitro antibacterial activity of 26 stingless bee honey samples collected from their cerumen egg-shaped pots in Marigat sub-County, Baringo County, Kenya was done. Antibacterial properties of the stingless bee honey was done with varying susceptibility patterns being observed at different concentrations of honey impregnated discs (10x104, 20x104, 50x104 and 75x104 ml µg/ ml) giving mean inhibition diameters of 18.23 ± 0.4 mm (Staphylococcus aureus), 17.49 ± 0.3 mm (Pseudomonas aeruginosa), 16.05 ± 0.6 mm (Klebsiella pneumoniae) and 10.19 ± 0.5 mm (Escherichia coli) with a mean range of 14.54 ± 2.0 mm to 17.58 ± 3 mm. Higher susceptibility to honey was recorded across all the bacterial isolates compared to conventional antibiotics while the mean MIC and MBC of the honey were recorded at 62.5 ml µg/ ml and 250 ml µg/ ml respectively. Control bacterial isolates Staphylococcus aureus ATCC 25923, Escherichia coli ATCC 25922, Klebsiella pneumoniae ATCC 27736 and Pseudomonas aeruginosa ATCC 27858 were used in the analysis. The stingless bee honey was found to be rich in various nutritive components like sugar (89.85 ± 5.07 g/100 g) and moisture (81.75 ± 10.35 mg/g) with a significant difference of P <0.05 as the main antibacterial components. Additionally, the stingless honey did possess water soluble vitamins, proteins and minerals of which potassium was the most dominant one. In regard to phytochemicals, on our preliminary analysis phenolic, flavonoid and carotenoid compounds were found to be present with phenolic compounds being the most dominant one. Stingless bee honey from Marigat, has antimicrobial properties which could be attributed to the rich phytochemicals it possesses and its physicochemical properties in addition to its high nutritive value.


Anti-Bacterial Agents , Honey , Microbial Sensitivity Tests , Honey/analysis , Anti-Bacterial Agents/pharmacology , Animals , Bees/microbiology , Humans , Bacteria/drug effects , Bacteria/isolation & purification , Phytochemicals/pharmacology , Wound Infection/microbiology , Wound Infection/prevention & control , Wound Infection/drug therapy , Virulence Factors
7.
Anal Chem ; 96(19): 7787-7796, 2024 May 14.
Article En | MEDLINE | ID: mdl-38702857

Microorganism are ubiquitous and intimately connected with human health and disease management. The accurate and fast identification of pathogenic microorganisms is especially important for diagnosing infections. Herein, three tetraphenylethylene derivatives (S-TDs: TBN, TPN, and TPI) featuring different cationic groups, charge numbers, emission wavelengths, and hydrophobicities were successfully synthesized. Benefiting from distinct cell wall binding properties, S-TDs were collectively utilized to create a sensor array capable of imaging various microorganisms through their characteristic fluorescent signatures. Furthermore, the interaction mechanism between S-TDs and different microorganisms was explored by calculating the binding energy between S-TDs and cell membrane/wall constituents, including phospholipid bilayer and peptidoglycan. Using a combination of the fluorescence sensor array and a deep learning model of residual network (ResNet), readily differentiation of Gram-negative bacteria (G-), Gram-positive bacteria (G+), fungi, and their mixtures was achieved. Specifically, by extensive training of two ResNet models with large quantities of images data from 14 kinds of microorganism stained with S-TDs, identification of microorganism was achieved at high-level accuracy: over 92.8% for both Gram species and antibiotic-resistant species, with 90.35% accuracy for the detection of mixed microorganism in infected wound. This novel method provides a rapid and accurate method for microbial classification, potentially aiding in the diagnosis and treatment of infectious diseases.


Deep Learning , Humans , Stilbenes/chemistry , Gram-Positive Bacteria/isolation & purification , Fluorescent Dyes/chemistry , Gram-Negative Bacteria/isolation & purification , Wound Infection/microbiology , Wound Infection/diagnosis , Fungi/isolation & purification
8.
AAPS PharmSciTech ; 25(5): 110, 2024 May 13.
Article En | MEDLINE | ID: mdl-38740721

Antimicrobial peptide LL37 is a promising antibacterial candidate due to its potent antimicrobial activity with no known bacterial resistance. However, intrinsically LL37 is susceptible to degradation in wound fluids limits its effectiveness. Bacterial toxins which are released after cell lysis are found to hinder wound healing. To address these challenges, encapsulating LL37 in microspheres (MS) and loading the MS onto activated carbon (AC)-chitosan (CS) hydrogel. This advanced wound dressing not only protects LL37 from degradation but also targets bacterial toxins, aiding in the healing of chronic wound infections. First, LL37 MS and LL37-AC-CS hydrogel were prepared and characterised in terms of physicochemical properties, drug release, and peptide-polymer compatibility. Antibacterial and antibiofilm activity, bacterial toxin elimination, cell migration, and cell cytotoxicity activities were investigated. LL37-AC-CS hydrogel was effective against Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus. LL37-AC-CS hydrogel bound more endotoxin than AC with CS hydrogel alone. The hydrogel also induced cell migration after 72 h and showed no cytotoxicity towards NHDF after 72 h of treatment. In conclusion, the LL37-AC-CS hydrogel was shown to be a stable, non-toxic advanced wound dressing method with enhanced antimicrobial and antitoxin activity, and it can potentially be applied to chronic wound infections to accelerate wound healing.


Anti-Bacterial Agents , Bandages , Chitosan , Escherichia coli , Hydrogels , Microspheres , Pseudomonas aeruginosa , Staphylococcus aureus , Chitosan/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Staphylococcus aureus/drug effects , Humans , Pseudomonas aeruginosa/drug effects , Escherichia coli/drug effects , Wound Healing/drug effects , Wound Infection/drug therapy , Wound Infection/microbiology , Wound Infection/prevention & control , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/administration & dosage , Cathelicidins , Microbial Sensitivity Tests/methods , Bacterial Toxins , Drug Liberation , Cell Movement/drug effects , Carbon/chemistry , Biofilms/drug effects
9.
Elife ; 132024 May 20.
Article En | MEDLINE | ID: mdl-38767331

Wound infections are highly prevalent and can lead to delayed or failed healing, causing significant morbidity and adverse economic impacts. These infections occur in various contexts, including diabetic foot ulcers, burns, and surgical sites. Enterococcus faecalis is often found in persistent non-healing wounds, but its contribution to chronic wounds remains understudied. To address this, we employed single-cell RNA sequencing (scRNA-seq) on infected wounds in comparison to uninfected wounds in a mouse model. Examining over 23,000 cells, we created a comprehensive single-cell atlas that captures the cellular and transcriptomic landscape of these wounds. Our analysis revealed unique transcriptional and metabolic alterations in infected wounds, elucidating the distinct molecular changes associated with bacterial infection compared to the normal wound healing process. We identified dysregulated keratinocyte and fibroblast transcriptomes in response to infection, jointly contributing to an anti-inflammatory environment. Notably, E. faecalis infection prompted a premature, incomplete epithelial-mesenchymal transition in keratinocytes. Additionally, E. faecalis infection modulated M2-like macrophage polarization by inhibiting pro-inflammatory resolution in vitro, in vivo, and in our scRNA-seq atlas. Furthermore, we discovered macrophage crosstalk with neutrophils, which regulates chemokine signaling pathways, while promoting anti-inflammatory interactions with endothelial cells. Overall, our findings offer new insights into the immunosuppressive role of E. faecalis in wound infections.


If wounds get infected, they heal much more slowly, sometimes leading to skin damage and other complications, including disseminated infections or even amputation. Infections can happen in many types of wounds, ranging from ulcers in patients with diabetes to severe burns. If infections are not cleared quickly, the wounds can become 'chronic' and are unable to heal without intervention. Enterococcus faecalis is a type of bacteria that normally lives in the gut. Within that environment, in healthy people, it is not harmful. However, if it comes into contact with wounds ­ particularly diabetic ulcers or the site of a surgery ­ it can cause persistent infections and prevent healing. Although researchers are beginning to understand how E. faecalis initially colonises wounds, the biological mechanisms that transform these infections into chronic wounds are still largely unknown. Celik et al. therefore set out to investigate exactly how E. faecalis interferes with wound healing. To do this, Celik et al. looked at E. faecalis-infected wounds in mice and compared them to uninfected ones. Using a genetic technique called single-cell RNA sequencing, Celik et al. were able to determine which genes were switched on in individual skin and immune cells at the site of the wounds. This in turn allowed the researchers to determine how those cells were behaving in both infected and uninfected conditions. The experiments revealed that when E. faecalis was present in wounds, several important cell types in the wounds did not behave normally. For example, although the infected skin cells still underwent a change in behaviour required for healing (called an epithelial-mesenchymal transition), the change was both premature and incomplete. In other words, the skin cells in infected wounds started changing too early and did not finish the healing process properly. E. faecalis also changed the way macrophages and neutrophils worked within the wounds. These are cells in our immune system that normally promote inflammation, a process involved in both uninfected wounds or during infections and is a key part of wound healing when properly controlled. In the E. faecalis-infected wounds, these cells' inflammatory properties were suppressed, making them less helpful for healing. These results shed new light on how E. faecalis interacts with skin cells and the immune system to disrupt wound healing. Celik et al. hope that this knowledge will allow us to find new ways to target E. faecalis infections, and ultimately develop treatments to help chronic wounds heal better and faster.


Enterococcus faecalis , Gram-Positive Bacterial Infections , Keratinocytes , Wound Healing , Enterococcus faecalis/physiology , Enterococcus faecalis/genetics , Animals , Mice , Gram-Positive Bacterial Infections/microbiology , Keratinocytes/microbiology , Keratinocytes/metabolism , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Disease Models, Animal , Wound Infection/microbiology , Transcriptome , Mice, Inbred C57BL , Single-Cell Analysis , Epithelial-Mesenchymal Transition/genetics , Male , Fibroblasts/microbiology , Fibroblasts/metabolism
10.
ACS Appl Mater Interfaces ; 16(20): 25757-25772, 2024 May 22.
Article En | MEDLINE | ID: mdl-38738757

The development of therapeutics with high antimicrobial activity and immunomodulatory effects is urgently needed for the treatment of infected wounds due to the increasing danger posed by recalcitrant-infected wounds. In this study, we developed light-controlled antibacterial, photothermal, and immunomodulatory biomimetic N/hPDA@M nanoparticles (NPs). This nanoplatform was developed by loading flavonoid naringenin onto hollow mesoporous polydopamine NPs in a π-π-stacked configuration and encasing them with macrophage membranes. First, our N/hPDA@M NPs efficiently neutralized inflammatory factors present within the wound microenvironment by the integration of macrophage membranes. Afterward, the N/hPDA@M NPs effectively dismantled bacterial biofilms through a combination of the photothermal properties of PDA and the quorum sensing inhibitory effects of naringenin. It is worth noting that N/hPDA@M NPs near-infrared-enhanced release of naringenin exhibited specificity toward the NF-κB-signaling pathway, effectively mitigating the inflammatory response. This innovative design not only conferred remarkable antibacterial properties upon the N/hPDA@M NPs but also endowed them with the capacity to modulate inflammatory responses, curbing excessive inflammation and steering macrophage polarization toward the M2 phenotype. As a result, this multifaceted approach significantly contributes to expediting the healing process of infected skin wounds.


Anti-Bacterial Agents , Biofilms , Indoles , NF-kappa B , Nanoparticles , Quorum Sensing , Wound Healing , Biofilms/drug effects , Nanoparticles/chemistry , Mice , NF-kappa B/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Wound Healing/drug effects , Animals , Quorum Sensing/drug effects , Indoles/chemistry , Indoles/pharmacology , Signal Transduction/drug effects , Flavanones/chemistry , Flavanones/pharmacology , RAW 264.7 Cells , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Polymers/chemistry , Polymers/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Wound Infection/drug therapy , Wound Infection/microbiology , Wound Infection/pathology , Immunomodulating Agents/chemistry , Immunomodulating Agents/pharmacology , Humans
11.
Int J Mol Sci ; 25(10)2024 May 17.
Article En | MEDLINE | ID: mdl-38791502

Wound infection is one of the most important factors affecting wound healing, so its effective control is critical to promote the process of wound healing. However, with the increasing prevalence of multi-drug-resistant (MDR) bacterial strains, the prevention and treatment of wound infections are now more challenging, imposing heavy medical and financial burdens on patients. Furthermore, the diminishing effectiveness of conventional antimicrobials and the declining research on new antibiotics necessitate the urgent exploration of alternative treatments for wound infections. Recently, phage therapy has been revitalized as a promising strategy to address the challenges posed by bacterial infections in the era of antibiotic resistance. The use of phage therapy in treating infectious diseases has demonstrated positive results. This review provides an overview of the mechanisms, characteristics, and delivery methods of phage therapy for combating pathogenic bacteria. Then, we focus on the clinical application of various phage therapies in managing refractory wound infections, such as diabetic foot infections, as well as traumatic, surgical, and burn wound infections. Additionally, an analysis of the potential obstacles and challenges of phage therapy in clinical practice is presented, along with corresponding strategies for addressing these issues. This review serves to enhance our understanding of phage therapy and provides innovative avenues for addressing refractory infections in wound healing.


Phage Therapy , Wound Infection , Phage Therapy/methods , Humans , Wound Infection/therapy , Wound Infection/microbiology , Wound Healing , Bacterial Infections/therapy , Bacterial Infections/microbiology , Bacteriophages/physiology , Animals , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Multiple, Bacterial
13.
Front Cell Infect Microbiol ; 14: 1386483, 2024.
Article En | MEDLINE | ID: mdl-38756229

Background: Ducrosia anethifolia is an aromatic desert plant used in Saudi folk medicine to treat skin infections. It is widely found in Middle Eastern countries. Methods: A methanolic extract of the plant was prepared, and its phytoconstituents were determined using LC-MS. In-vitro and in-vivo antibacterial and antibiofilm activities of the methanolic extract were evaluated against multidrug-resistant bacteria. The cytotoxic effect was assessed using HaCaT cell lines in-vitro. Diabetic mice were used to study the in-vivo antibiofilm and wound healing activity using the excision wound method. Results: More than 50 phytoconstituents were found in the extract after LC-MS analysis. The extract exhibited antibacterial activity against both the tested pathogens. The extract was free of irritant effects on mice skin, and no cytotoxicity was observed on HaCaT cells with an IC50 value of 1381 µg/ml. The ointment formulation of the extract increased the healing of diabetic wounds. The microbial load of both pathogens in the wounded tissue was also reduced after the treatment. The extract was more effective against methicillin-resistant Staphylococcus aureus (MRSA) than MDR-P. aeruginosa in both in vitro and in vivo experiments. Further, skin regeneration was also observed in histological studies. Conclusions: The results showed that D. anethifolia methanol extract supports wound healing in infected wounds in diabetic mice through antibacterial, antibiofilm, and wound healing activities.


Anti-Bacterial Agents , Biofilms , Diabetes Mellitus, Experimental , Methicillin-Resistant Staphylococcus aureus , Plant Extracts , Pseudomonas aeruginosa , Wound Healing , Animals , Biofilms/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Plant Extracts/pharmacology , Plant Extracts/chemistry , Mice , Anti-Bacterial Agents/pharmacology , Wound Healing/drug effects , Pseudomonas aeruginosa/drug effects , Humans , Diabetes Mellitus, Experimental/drug therapy , Microbial Sensitivity Tests , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Cell Line , HaCaT Cells , Male , Wound Infection/drug therapy , Wound Infection/microbiology , Disease Models, Animal , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology
14.
Emerg Infect Dis ; 30(5): 1055-1057, 2024 May.
Article En | MEDLINE | ID: mdl-38666739

We report a clinical isolate of Burkholderia thailandensis 2022DZh obtained from a patient with an infected wound in southwest China. Genomic analysis indicates that this isolate clusters with B. thailandensis BPM, a human isolate from Chongqing, China. We recommend enhancing monitoring and surveillance for B. thailandensis infection in both humans and livestock.


Burkholderia Infections , Burkholderia , Phylogeny , Wound Infection , Humans , Male , Burkholderia/genetics , Burkholderia/isolation & purification , Burkholderia/classification , Burkholderia Infections/microbiology , Burkholderia Infections/diagnosis , China/epidemiology , Genome, Bacterial , Wound Infection/microbiology , Middle Aged
15.
Wounds ; 36(3): 90-94, 2024 03.
Article En | MEDLINE | ID: mdl-38684124

BACKGROUND: NPWT has been used to treat various wounds. Scant evidence exists on the use of custom-made NPWT for infected wounds. NPWT dressings promote wound healing by increasing local blood flow and antibiotic concentration, and by removing exudates from the wound. OBJECTIVE: To report the use of custom-made NPWT dressings to manage complex infected wounds of the lower limb. MATERIALS AND METHODS: The authors retrospectively reviewed the records of 43 patients with complex infected wounds of the lower limb treated with debridement and low-cost, custom-made NPWT dressing connected to wall suction from January 1, 2018 to December 31, 2020, at PSG Medical College Hospital, Coimbatore, India. RESULTS: A total of 43 patients with infected wounds of the lower limb were treated with the custom-made NPWT dressings. Second-look debridement was required in 5 patients. An average of 5 dressing changes were required for optimal wound granulation, with 23% of patients (n = 10) requiring secondary suturing and 62% (n = 27) requiring STSG for definitive coverage of the wound. Healing by secondary intention was achieved in 6 patients. The average duration from the start of therapy until the wound was ready for coverage (STSG or secondary suturing) was 2.5 weeks (range, 1-5 weeks), with an average time to complete wound healing of 5 weeks (range, 3-7 weeks). The most common wound isolate was Staphylococcus aureus (60%). No complications occurred. CONCLUSIONS: Custom-made NPWT dressings are safe to use in complex infected lower limb wounds. These dressings keep the wound dry and promote healing. Wound debridement followed by NPWT combined with antibiotic therapy can act synergistically to promote wound healing and control infection.


Debridement , Negative-Pressure Wound Therapy , Wound Healing , Wound Infection , Humans , Negative-Pressure Wound Therapy/methods , Retrospective Studies , Male , Female , Wound Infection/therapy , Wound Infection/microbiology , Middle Aged , Debridement/methods , Adult , Bandages , Treatment Outcome , Aged , Anti-Bacterial Agents/therapeutic use
16.
Int J Biol Macromol ; 269(Pt 2): 131795, 2024 Jun.
Article En | MEDLINE | ID: mdl-38670175

Bacterial infections during wound healing impede the healing process and trigger local or systemic inflammatory reactions. Consequently, there is an urgent need to develop a new material with antimicrobial and antioxidant properties to promote infected wound healing. A synergistically antimicrobial and antioxidant hyaluronic acid hydrogel (HMn) is prepared by employing MnO2 nanosheets into 4ARM-PEG5000-SH crosslinked methacrylated hyaluronic acid (HAMA) network. The coordination between sulfhydryl groups of 4ARM-PEG5000-SH and MnO2 nanosheets ensures entrapment of the nanosheets within the hydrogel, while the interaction between 4ARM-PEG5000-SH and HAMA results in facile gelation through thiol-ene click reaction. MnO2 nanosheets exhibit strong photothermal properties and reactive oxygen species (ROS) scavenging abilities, while hyaluronic acid promotes wound healing. When subjected to near-infrared (NIR) irradiation, the HMn achieves a bactericidal rate of 95.24 % for Staphylococcus aureus and nearly 100 % for Escherichia coli. In animal experiments, treatment with the HMn under NIR irradiation results in the best wound healing outcomes. Both in vitro and vivo biocompatible assays demonstrate that the HMn has rarely cell cytotoxicity and tissue damage. The HMn is easy to prepare and has good biocompatibility as well as efficient antibacterial and antioxidant properties, providing a novel method for the treatment of infected wounds.


Antioxidants , Escherichia coli , Hyaluronic Acid , Hydrogels , Staphylococcus aureus , Wound Healing , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Antioxidants/pharmacology , Antioxidants/chemistry , Animals , Wound Healing/drug effects , Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Wound Infection/drug therapy , Wound Infection/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Mice , Reactive Oxygen Species/metabolism , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Humans , Drug Synergism
17.
J Mater Chem B ; 12(21): 5111-5127, 2024 May 29.
Article En | MEDLINE | ID: mdl-38687208

Recently, bacterial infections have become a global crisis, greatly threatening the health of human beings. The development of a non-antibiotic biomaterial is recognized as an alternative way for the effective treatment of bacterial infections. In the present work, a multifunctional copper peroxide (CP) nanodot-decorated gold nanostar (GNS)/silica nanorod (SiNR) Janus nanostructure (GNS@CP/SiNR) with excellent antibacterial activity was reported. Due to the formation of the Janus nanostructure, GNS@CP/SiNR displayed strong plasmonic resonance absorbance in the near infrared (NIR)-II region that enabled the nanosystem to achieve mild photothermal therapy (MPTT). In acidic conditions, CP decorated on GNS@CP/SiNR dissociated rapidly by releasing Cu2+ and H2O2, which subsequently transformed to ˙OH via the Fenton-like reaction for chemodynamic therapy (CDT). As a result, GNS@CP/SiNR could effectively inhibit both Gram-negative Escherichia coli (E. coli) and Gram-positive Staphylococcus aureus (S. aureus), and eradicate the associated bacterial biofilms by exerting the synergistic MPTT/CDT antibacterial effect. Moreover, GNS@CP/SiNR was also demonstrated to be effective in treating wound infections, as verified on the S. aureus-infected full thickness excision wound rat model. Our mechanism study revealed that the synergistic MPTT/CDT effect of GNS@CP/SiNR firstly caused bacterial membrane damage, followed by boosting intracellular ROS via the severe oxidative stress effect, which subsequently caused the depletion of intracellular GSH and DNA damage, finally leading to the death of bacteria.


Anti-Bacterial Agents , Copper , Escherichia coli , Gold , Hydroxyl Radical , Nanotubes , Silicon Dioxide , Staphylococcus aureus , Gold/chemistry , Gold/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Animals , Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Nanotubes/chemistry , Hydroxyl Radical/metabolism , Hydroxyl Radical/chemistry , Copper/chemistry , Copper/pharmacology , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology , Rats , Wound Infection/drug therapy , Wound Infection/microbiology , Photothermal Therapy , Infrared Rays , Microbial Sensitivity Tests , Metal Nanoparticles/chemistry , Rats, Sprague-Dawley , Biofilms/drug effects
18.
Int J Biol Macromol ; 268(Pt 2): 131637, 2024 May.
Article En | MEDLINE | ID: mdl-38636748

Bacterial-infected wound repair has become a significant public health concern. This study developed a novel 3D-printed piezocatalytic SF-MA/PEGDA/Ag@BT (SPAB) hydrogels were fabricated by using digital light processing. These hydrogels exhibited high consistency, mechanical properties and good biocompatibility. Besides, the SPAB hydrogels exhibited excellent piezocatalytic performance and thus could induce piezoelectric polarization under ultrasound to generate reactive oxygen species (ROS). The SPAB hydrogels possessed an antibacterial rate of 99.23% and 99.96% for Escherichia coli and Staphylococcus aureus, respectively, under 5 min of ultrasonic stimulation (US) in vitro. The US-triggered piezocatalytic performance could increase antibacterial activity and improve the healing process of the infected wound. Therefore, the 3D printed piezocatalytic SPAB hydrogels could be unutilized as wound dressing in the field of bacterial-infected wound repair.


Anti-Bacterial Agents , Escherichia coli , Hydrogels , Printing, Three-Dimensional , Staphylococcus aureus , Wound Healing , Hydrogels/chemistry , Hydrogels/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Wound Healing/drug effects , Wound Infection/drug therapy , Wound Infection/microbiology , Catalysis , Animals , Reactive Oxygen Species/metabolism , Humans , Polyethylene Glycols/chemistry , Ultrasonic Waves
19.
J Wound Care ; 33(Sup4a): xcix-cx, 2024 Apr 02.
Article En | MEDLINE | ID: mdl-38588056

Metal-based nanoparticles (MNPs) are promoted as effective compounds in the treatment of bacterial infections and as possible alternatives to antibiotics. These MNPs are known to affect a broad spectrum of microorganisms using a multitude of strategies, including the induction of reactive oxygen species and interaction with the inner structures of the bacterial cells. The aim of this review was to summarise the latest studies about the effect of metal-based nanoparticles on pathogenic bacterial biofilm formed in wounds, using the examples of Gram-positive bacterium Staphylococcus aureus and Gram-negative bacterium Pseudomonas aeruginosa, as well as provide an overview of possible clinical applications.


Nanoparticles , Staphylococcal Infections , Wound Infection , Humans , Biofilms , Staphylococcus aureus , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Pseudomonas aeruginosa , Nanoparticles/therapeutic use , Wound Infection/drug therapy , Wound Infection/microbiology
20.
J Wound Care ; 33(3): 180-188, 2024 Mar 02.
Article En | MEDLINE | ID: mdl-38451790

OBJECTIVE: The incidence of hard-to-heal wound infection, especially as a result of multidrug-resistant Gram-negative organisms, has increased in recent years. The reason for the increase is multifactorial and the ability of these pathogenic isolates to form biofilms is one of the important risk factors in wound infection. This study aimed to evaluate the risk factors associated with such cases. METHOD: This prospective analytical study, conducted over a period of two months, included pus or tissue samples from hospital inpatients with Gram-negative hard-to-heal wound infection. The samples were processed with conventional microbiological techniques. Patient demographic details and the presence of various risk factors were recorded. Biofilm production was detected by tissue culture plate method in the laboratory. The data were analysed using SPSS version 21 (IBM Ltd., US). RESULTS: The experimental cohort comprised 200 patients. Klebsiella spp. was the most common identified organism, followed by Escherichia coli and Pseudomonas spp. Carbapenem resistance was observed in 106 (53%) strains. Almost 66% of the strains showed biofilm formation. On evaluation of associated risk factors, age (p=0.043), presence of biofilms (p=0.0001), diabetes (p=0.002), hypertension (p=0.02) and medical device use (p=0.008) had significant association, whereas sex, previous surgery and prior antibiotic use had no significant impact on the chronicity of the wound. CONCLUSION: In this study, chronicity of wounds was observed to be associated with multiple risk factors, especially the biofilm-forming ability of the strain. Biofilms are difficult to eradicate and additional measures, such as physical debridement, are important for resolving chronicity. Knowledge about specific risk factors would also allow clinicians a better understanding of the healing process and drive appropriate wound care interventions. DECLARATION OF INTEREST: A grant was received from the Indian Council of Medical Research (ICMR) for this work (grant ID: 2017-02686). The authors have no conflicts of interest to declare.


Wound Healing , Wound Infection , Humans , Tertiary Care Centers , Prospective Studies , Wound Infection/epidemiology , Wound Infection/microbiology , Risk Factors , Biofilms
...