Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.231
Filter
1.
Int Wound J ; 21(9): e70063, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39313239

ABSTRACT

PURPOSE: This study aims to assess the diagnostic accuracy of non-culture-based methodologies for detecting microorganisms in chronic wounds. METHODS: We systematically reviewed studies that evaluated the diagnostic accuracy of alternative tests in chronic wound samples, excluding studies focused on animal samples or unrelated conditions. The search encompassed PubMed, CINAHL, Scopus and Web of Science databases, employing the QUADAS-2 tool for risk of bias assessment. Our search included the PubMed, CINAHL, Scopus and Web of Science databases, and we assessed the risk of bias using the QUADAS-2 tool. A meta-analysis was conducted on polymerase chain reaction (PCR) and colorimetric methods to determine sensitivity, specificity, diagnostic odds ratio, and summary receiver-operating characteristic (sROC) curves using a random-effects model. For methods not suitable for quantitative synthesis, a narrative synthesis was performed. RESULTS: Nineteen studies involving various types of chronic wounds were analysed, revealing diverse diagnostic methods including fluorescence, PCR, colorimetry, voltammetry, electronic nose, biosensors, enzymatic methods, staining and microscopy. Combining fluorescence with clinical signs and symptoms (CSS) versus culture showed significant accuracy. Colorimetry demonstrated low sensitivity but high specificity, with a diagnostic odds ratio of 6.3. PCR generally exhibited good accuracy, although significant heterogeneity was noted, even in subgroup analyses. CONCLUSIONS: This study identified a broad spectrum of diagnostic approaches, highlighting the superior diagnostic accuracy achieved when microbiological analysis is combined with clinical assessments. However, the heterogeneity and methodological variations across studies present challenges in meta-analysis. Future research should aim for standardized and homogeneous study designs to enhance the assessment of diagnostic accuracy for alternative methods.


Subject(s)
Sensitivity and Specificity , Wound Infection , Humans , Wound Infection/diagnosis , Wound Infection/microbiology , Chronic Disease , Diagnostic Tests, Routine/methods , Diagnostic Tests, Routine/standards , Female , Male , Wounds and Injuries/diagnosis , Wounds and Injuries/microbiology , Adult , Middle Aged , Polymerase Chain Reaction/methods , Aged , Colorimetry/methods , Aged, 80 and over
2.
Narra J ; 4(2): e980, 2024 08.
Article in English | MEDLINE | ID: mdl-39280315

ABSTRACT

The emergence of multidrug-resistant (MDR) infections in wounds is a significant public health issue. The aim of this study was to investigate the prevalence and antimicrobial resistance profiles of MDR bacterial isolates in wound infections. Through a cross-sectional study, 1,035 bacterial isolates were collected from wound infection patients at Tugurejo Hospital in Semarang, Indonesia, over a three-year period (from January 2020 to December 2022). Initial identification involved Gram staining and colony morphology assessment, followed by biochemical assays and antimicrobial susceptibility testing using the VITEK®2 Compact system. Gram-negative bacteria constituted the majority of isolates (60.77%, n=629). The predominant strains included were Staphylococcus spp. (30.92%, n=320), Escherichia coli (18.45%, n=191), and Klebsiella pneumoniae (13.04%, n=135). Notably, Gram-negative bacteria exhibited a significantly higher likelihood of MDR development compared to their Gram-positive counterparts (p<0.001), with Gram-negative bacteria having a 2.05 times higher probability of acquiring MDR. These findings underscore the urgent need for comprehensive surveillance of antimicrobial resistance patterns and the implementation of tailored antimicrobial stewardship programs to address the pressing public health challenge of MDR wound infections. Further research is warranted to elucidate the complex interplay of factors contributing to MDR development in wound infections, thereby informing targeted intervention strategies and improving patient outcomes.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Multiple, Bacterial , Microbial Sensitivity Tests , Wound Infection , Humans , Indonesia/epidemiology , Cross-Sectional Studies , Wound Infection/microbiology , Wound Infection/epidemiology , Wound Infection/drug therapy , Prevalence , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Male , Female , Middle Aged , Adult , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/isolation & purification , Aged
3.
J Wound Care ; 33(9): 708-717, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39287030

ABSTRACT

OBJECTIVE: The aim of this in vitro experimental series was to explore the mode of action of a hydrocellular polyurethane foam dressing (HPFD) and how its advanced features support beneficial interactions with the wound bed to address common barriers to wound healing, thus supporting improved clinical outcomes. METHOD: Multiple in vitro microbiological tests were performed, assessing prevention of bacterial ingress, surface removal of bacteria, bacterial sequestration and retention into the dressing in a clinically relevant environment. Odour molecule concentrations were measured using gas chromatography and further assays explored matrix metalloproteinase (MMP)-9 retention in the dressing using enzyme linked immunosorbent assay. RESULTS: The HPFD demonstrated marked reductions in bioburden levels across multiple tests. These included prevention of bacterial ingress for seven days, removal of surface bacteria and absorption into the dressing. Further tests identified that most bacteria were sequestered into the hyperabsorbent layer (90.5% for Pseudomonas aeruginosa and 89.6% for meticillin-resistant Staphylococcus aureus). Moreover, the majority of bacteria (99.99% for both test organisms) were retained within the dressing, even upon compression. Additional tests demonstrated a marked reduction of odour molecules following incubation with HPFD and total retention of protease MMP-9 within the dressing. CONCLUSIONS: Proactive management of the wound environment with an appropriate advanced wound dressing, such as the HPFD examined in these in vitro investigations, can not only help to minimise the barriers to healing, as observed across this test series by direct interaction with the wound bed, but may, as a result, provide an ideal environment for wound progression with minimal disturbance.


Subject(s)
Polyurethanes , Wound Healing , Humans , Pseudomonas aeruginosa , Bandages , Wound Infection/microbiology , Odorants , Methicillin-Resistant Staphylococcus aureus
4.
J Wound Care ; 33(9): 702-707, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39287037

ABSTRACT

OBJECTIVE: The effect of continuous topical oxygen therapy (cTOT) on Pseudomonas aeruginosa biofilm gene transcription profiles following inoculation onto porcine skin, using a customised molecular assay was determined. METHOD: Sterilised porcine skin explants were inoculated with Pseudomonas aeruginosa in triplicate: 0 hours as negative control; 24 hours cTOT device on; 24 hours cTOT device off. The oxygen delivery system of the cTOT device was applied to the inoculated tissue and covered with a semi-occlusive dressing. All samples were incubated at 37±2°C for 24 hours, with the 0 hours negative control inoculated porcine skin samples recovered immediately. Planktonic suspensions and porcine skin biopsy samples were taken at 0 hours and 24 hours. Samples were processed and quantifiably assessed using gene specific reverse transcription-quantitative polymerase chain reaction assays for a panel of eight Pseudomonas aeruginosa genes (16S, pelA, pslA, rsaL, pcrV, pscQ, acpP, cbrA) associated with biofilm formation, quorum sensing, protein secretion/translocation and metabolism. RESULTS: Transcriptional upregulation of pelA, pcrV and acpP, responsible for intracellular adhesion, needletip protein production for type-3 secretion systems and fatty acid synthesis during proliferation, respectively, was observed when the cTOT device was switched on compared to when the device was switched off. Data suggest increased metabolic activity within bacterial cells following cTOT treatment. CONCLUSION: cTOT is an adjunctive therapy that supports faster healing and pain reduction in non-healing hypoxic wounds. Oxygen has previously been shown to increase susceptibility of biofilms to antibiotics through enhancing metabolism. Observed gene expression changes highlighted the impact of cTOT on biofilms, potentially influencing antimicrobial treatment success in wounds. Further in vitro and clinical investigations are warranted.


Subject(s)
Biofilms , Oxygen , Pseudomonas aeruginosa , Animals , Biofilms/drug effects , Pseudomonas aeruginosa/drug effects , Swine , Oxygen/metabolism , Disease Models, Animal , Pseudomonas Infections/therapy , Skin/metabolism , Skin/microbiology , Wound Infection/therapy , Administration, Topical , Wound Healing
5.
ACS Appl Mater Interfaces ; 16(38): 50321-50334, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39264683

ABSTRACT

The sophisticated environment of chronic wounds, characterized by prolonged exudation and recurrent bacterial infections, poses significant challenges to wound recovery. Recent advancements in multifunctional wound dressings fall short of providing comprehensive, accurate, and comfortable treatment. To address these issues, a battery-free and multifunctional microfluidic Janus wound dressing (MM-JWD) capable of three functions, including exudate management, antibacterial properties, and multiple indications of wound infection detection, has been developed. During the treatment, the fully soft microfluidic Janus membrane not only demonstrated stable unidirectional fluid transport capabilities under various skin deformations for a longer period but also provided antibacterial effects through surface treatment with chitosan quaternary ammonium salts and poly(vinyl alcohol). Furthermore, integrating multiple colorimetric sensors within the Janus membrane's microchannels and a dual-layer structure enabled simultaneous monitoring of the wound's pH, uric acid, and temperature. The monitoring was facilitated by smartphone recognition of color changes in the sensors. In vivo and in vitro tests confirmed the exudate management, antibacterial, and sensing capabilities of the MM-JWD, proving its efficacy in monitoring and promoting the healing of wounds. Overall, this study provides a valuable method for the design of multifunctional wound dressings for chronic wound care.


Subject(s)
Anti-Bacterial Agents , Bandages , Wound Healing , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Animals , Wound Healing/drug effects , Chitosan/chemistry , Chitosan/pharmacology , Polyvinyl Alcohol/chemistry , Staphylococcus aureus/drug effects , Humans , Wound Infection/drug therapy , Wound Infection/microbiology , Hydrogen-Ion Concentration , Mice , Uric Acid/chemistry , Colorimetry
6.
ACS Appl Mater Interfaces ; 16(38): 50160-50174, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39265036

ABSTRACT

Diabetic wounds are prone to recurrent infections, often leading to delayed healing. To address this challenge, we developed a chitin-copper sulfide (CuS@CH) composite sponge, which combines bacterial trapping with near-infrared (NIR) activated phototherapy for treating infected diabetic wounds. CuS nanoparticles were synthesized and incorporated in situ within the sponge using a chitin assisted biomineralization strategy. The positively charged chitin surface effectively adhered bacteria, while NIR irradiation of CuS generated reactive oxygen species (ROS) heat and Cu2+ to rapidly damage the trapped bacteria. This synergistic effect resulted in an exceptional antibacterial performance against E. coli (∼99.9%) and S. aureus (∼99.3%). The bactericidal mechanism involved NIR-induced glutathione oxidation, membrane lipid peroxidation, and increased membrane permeability. In diabetic mouse models, the CuS@CH sponge accelerated the wound healing of S. aureus infected wounds by facilitating collagen deposition and reducing inflammation. Furthermore, the sponge demonstrated good biocompatibility. This dual-functional platform integrating bacterial capture and NIR-triggered phototherapy shows promise as an antibacterial wound dressing to promote healing of infected diabetic wound.


Subject(s)
Anti-Bacterial Agents , Chitin , Copper , Diabetes Mellitus, Experimental , Escherichia coli , Infrared Rays , Staphylococcus aureus , Wound Healing , Animals , Wound Healing/drug effects , Staphylococcus aureus/drug effects , Mice , Copper/chemistry , Copper/pharmacology , Escherichia coli/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Chitin/chemistry , Chitin/pharmacology , Diabetes Mellitus, Experimental/pathology , Wound Infection/drug therapy , Wound Infection/microbiology , Wound Infection/pathology , Wound Infection/therapy , Reactive Oxygen Species/metabolism , Bandages , Staphylococcal Infections/drug therapy , Staphylococcal Infections/pathology
7.
Front Cell Infect Microbiol ; 14: 1428525, 2024.
Article in English | MEDLINE | ID: mdl-39310784

ABSTRACT

Introduction: Managing burn injuries is a challenge in healthcare. Due to the alarming increase in antibiotic resistance, new prophylactic and therapeutic strategies are being sought. This study aimed to evaluate the potential of live Lactic Acid Bacteria for managing burn infections, using Galleria mellonella larvae as an alternative preclinical animal model and comparing the outcomes with a common antibiotic. Methods: The antimicrobial activity of LAB isolated from human breast milk was assessed in vitro against Pseudomonas aeruginosa ATCC 27853. Additionally, the immunomodulatory effects of LAB were evaluated in vivo using the G. mellonella burn wound infection model. Results and discussion: In vitro results demonstrated the antimicrobial activity of Lactic Acid Bacteria against P. aeruginosa. In vivo results show that their prophylactic treatment improves, statistically significant, larval survival and modulates the expression of immunity-related genes, Gallerimycin and Relish/NF-κB, strain-dependently. These findings lay the foundation and suggest a promising alternative for burn wound prevention and management, reducing the risk of antibiotic resistance, enhancing immune modulation, and validating the potential G. mellonella as a skin burn wound model.


Subject(s)
Burns , Disease Models, Animal , Lactobacillales , Larva , Milk, Human , Pseudomonas aeruginosa , Animals , Burns/microbiology , Pseudomonas aeruginosa/drug effects , Humans , Larva/microbiology , Milk, Human/microbiology , Female , Pseudomonas Infections/microbiology , Pseudomonas Infections/drug therapy , Pseudomonas Infections/immunology , Moths/microbiology , Wound Infection/microbiology , Wound Infection/drug therapy , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests
8.
J Nanobiotechnology ; 22(1): 579, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304913

ABSTRACT

The high incidence and mortality rates associated with acute and chronic wound infections impose a significant burden on global healthcare systems. In terms of the management of wound infection, the reconstruction and regeneration of skin appendages are essential for the recovery of mechanical strength and physiological function in the regenerated skin tissue. Novel therapeutic approaches are a requisite for enhancing the healing of infected wounds and promoting the regeneration of skin appendages. Herein, a novel antimicrobial microneedle patch has been fabricated for the transdermal controlled delivery of adipose tissue-derived apoptotic vesicles (ApoEVs-AT@MNP) for the treatment of infected wounds, which is expected to achieve high-quality scarless healing of the wound skin while inhibiting the bacteria in the infected wound. The microneedle patch (MNP) system possesses adequate mechanical strength to penetrate the skin, allowing the tips to remain inside tissue for continuous active release of biomolecules, and subsequently degrades safely within the host body. In vivo transplantation demonstrates that ApoEVs-AT@MNP not only inhibits bacterial proliferation in infected wounds but also significantly promotes effective and rapid scarless wound healing. Particularly noteworthy is the ability of ApoEVs-AT@MNP to promote the rapid formation of mature, evenly arranged hair follicles in infected wounds, observed as early as 8 days following implantation, which is essential for the restoration of skin function. This rapid development of skin appendages has not been reported this early in previous studies. Therefore, ApoEVs-AT@MNP has emerged as an excellent, painless, non-invasive, and highly promising treatment for infected wounds.


Subject(s)
Adipose Tissue , Apoptosis , Needles , Wound Healing , Wound Healing/drug effects , Animals , Adipose Tissue/cytology , Mice , Apoptosis/drug effects , Skin/drug effects , Male , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Extracellular Vesicles/chemistry , Wound Infection/drug therapy , Anti-Infective Agents/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Mice, Inbred BALB C
9.
Narra J ; 4(2): e774, 2024 08.
Article in English | MEDLINE | ID: mdl-39280309

ABSTRACT

Infectious wounds on the skin surface are easily colonized by bacteria from pyogenic group that manifest as inflammation, such as Pseudomonas aeruginosa. P. aeruginosa is a Gram-negative bacterium and an opportunistic pathogen known for causing invasive state in critically ill and immunocompromised patients. The aim of this study was to detect the 16S rRNA and gyrB genes in P. aeruginosa using polymerase chain reaction (PCR) method. The sample in this study was pus isolate from a 5-year-old boy with leg wounds. The bacteria were isolated on brain heart infusion broth (BHIB) media and identified with molecular identification. Sequencing and BLAST analysis were carried out to determine the similarity of gene identity by comparing sample sequence with other isolate sequences on the Gene Bank. The results of molecular identification showed amplification DNA band of around 934 base pairs (bp) for 16S rRNA and 225 bp for gyrB gene. The BLAST program demonstrated that the sample had 99.89% similarity with P. aeruginosa strain XC4 (accession code ON795960.1) for the 16S rRNA gene. Meanwhile, the gyrB gene exhibited 99.10% similarity with the P. aeruginosa strain PSA-1.2 (accession code KP172300.1).


Subject(s)
DNA Gyrase , Polymerase Chain Reaction , Pseudomonas Infections , Pseudomonas aeruginosa , RNA, Ribosomal, 16S , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/isolation & purification , Male , Humans , RNA, Ribosomal, 16S/genetics , Polymerase Chain Reaction/methods , Child, Preschool , Pseudomonas Infections/microbiology , Pseudomonas Infections/diagnosis , DNA Gyrase/genetics , Indonesia , Wound Infection/microbiology , Wound Infection/diagnosis , Suppuration/microbiology
10.
Carbohydr Polym ; 346: 122554, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39245535

ABSTRACT

Frequent occurrence of wound infection caused by multiple-resistant bacteria (MRB) has posed a serious challenge to the current healthcare system relying on antibiotics. The development of novel antimicrobial materials with high safety and efficacy to heal wound infection is of great importance in combating this crisis. Herein, we prepared a promising antibacterial hydrogel by cross-linking ferrous ions (Fe2+) with the deprotonated carboxyl anion in sodium alginate (Na-ALG) to cure wound infections caused by methicillin-resistant Staphylococcus aureus (MRSA). Interestingly, ferrous-modified Na-ALG (Fe-ALG) hydrogel demonstrated better properties compared to the traditional Na-ALG-based hydrogels, including injectability, self-healing, appropriate fluidity, high-water retention, potent MRSA-killing efficacy, and excellent biocompatibility. Importantly, the addition of Fe2+ enhances the antibacterial efficacy of the Na-ALG hydrogel, enabling it to effectively eliminate MRSA and accelerate the healing of antibiotic-resistant bacterial-infected wounds in a remarkably short period (10 days). This modification not only facilitates wound closure and fur generation, but also mitigates systemic inflammation, thereby effectively impeding the spread of MRSA to the lungs. Taken together, Fe-ALG hydrogel is a promising therapeutic material for treating wound infections by Staphylococcus aureus, especially by antibiotic-resistant strains like MRSA.


Subject(s)
Alginates , Anti-Bacterial Agents , Ferrous Compounds , Hydrogels , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Wound Healing , Wound Infection , Alginates/chemistry , Alginates/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Ferrous Compounds/chemistry , Ferrous Compounds/pharmacology , Wound Healing/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Animals , Staphylococcal Infections/drug therapy , Wound Infection/drug therapy , Wound Infection/microbiology , Mice , Microbial Sensitivity Tests , Male
11.
Int J Nanomedicine ; 19: 8901-8927, 2024.
Article in English | MEDLINE | ID: mdl-39233743

ABSTRACT

Introduction: Graphene oxide (GO) nanoparticles have emerged as a compelling photothermal agent (PHTA) in the realm of photothermal antibacterial therapy, owing to their cost-effectiveness, facile synthesis, and remarkable photostability. Nevertheless, the therapeutic efficacy of GO nanoparticles is commonly hindered by their inherent drawback of low photothermal conversion efficiency (PCE). Methods: Herein, we engineer the Ag/GO-GelMA platform by growing the Ag on the surface of GO and encapsulating the Ag/GO nanoparticles into the GelMA hydrogels. Results: The resulting Ag/GO-GelMA platform demonstrates a significantly enhanced PCE (47.6%), surpassing that of pure GO (11.8%) by more than fourfold. As expected, the Ag/GO-GelMA platform, which was designed to integrate the benefits of Ag/GO nanoparticles (high PCE) and hydrogel (slowly releasing Ag+ to exert an inherent antibacterial effect), has been shown to exhibit exceptional antibacterial efficacy. Furthermore, transcriptome analyses demonstrated that the Ag/GO-GelMA platform could significantly down-regulate pathways linked to inflammation (the MAPK and PI3K-Akt pathways) and had the ability to promote cell migration. Discussion: Taken together, this study presents the design of a potent photothermal antibacterial platform (Ag/GO-GelMA) aimed at enhancing the healing of infectious wounds. The platform utilizes a handy method to enhance the PCE of GO, thereby making notable progress in the utilization of GO nano-PHTAs.


Subject(s)
Anti-Bacterial Agents , Graphite , Hydrogels , Silver , Wound Healing , Graphite/chemistry , Graphite/pharmacology , Wound Healing/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Silver/chemistry , Silver/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Humans , Mice , Photothermal Therapy/methods , Nanoparticles/chemistry , Wound Infection/drug therapy , Wound Infection/microbiology , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Cell Movement/drug effects
12.
J Med Microbiol ; 73(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-39268705

ABSTRACT

Introduction. As growing numbers of patients are at higher risk of infection, novel topical broad-spectrum antimicrobials are urgently required for wound infection management. Robust pre-clinical studies should support the development of such novel antimicrobials.Gap statement. To date, evidence of robust investigation of the cytotoxicity and antimicrobial spectrum of activity of antimicrobial peptides (AMP)s is lacking in published literature. Using a more clinical lens, we address this gap in experimental approach, building on our experience with poly-l-lysine (PLL)-based AMP polymers.Aim. To evaluate the in vitro bactericidal activity and cytotoxicity of a PLL-based 16-armed star AMP polymer, designated 16-PLL10, as a novel candidate antimicrobial.Methods. Antimicrobial susceptibilities of clinical isolates and reference strains of ESKAPE (Enterococcus spp., Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter spp.) pathogens, to 16-PLL10 were investigated. Human erythrocyte haemolysis and keratinocyte viability assays were used to assess toxicity. Modifications were made to 16-PLL10 and re-evaluated for improvement.Results. Minimum bactericidal concentration of 16-PLL10 ranged from 1.25 µM to ≥25 µM. At 2.5 µM, 16-PLL10 was broadly bactericidal against ESKAPE strains/wound isolates. Log-reduction in colony forming units (c.f.u.) per millilitre after 1 h, ranged from 0.3 (E. cloacae) to 5.6 (K. pneumoniae). At bactericidal concentrations, 16-PLL10 was toxic to human keratinocyte and erythrocytes. Conjugates of 16-PLL10, Trifluoroacetylated (TFA)-16-PLL10, and Poly-ethylene glycol (PEG)ylated 16-PLL10, synthesised to address toxicity, only moderately reduced cytotoxicity and haemolysis.Conclusions. Due to poor selectivity indices, further development of 16-PLL10 is unlikely warranted. However, considering the unmet need for novel topical antimicrobials, the ease of AMP polymer synthesises/modification is attractive. To support more rational development, prioritising clinically relevant pathogens and human cells, to establish selective toxicity profiles in vitro, is critical. Further characterisation and discovery utilising artificial intelligence and computational screening approaches can accelerate future AMP nanomaterial development.


Subject(s)
Antimicrobial Peptides , Microbial Sensitivity Tests , Polylysine , Humans , Polylysine/pharmacology , Polylysine/chemistry , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Erythrocytes/drug effects , Wound Infection/microbiology , Wound Infection/drug therapy , Klebsiella pneumoniae/drug effects , Hemolysis/drug effects , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects , Polymers/pharmacology , Polymers/chemistry , Acinetobacter baumannii/drug effects , Keratinocytes/drug effects , Bacteria/drug effects , Cell Survival/drug effects
13.
J Biomed Mater Res B Appl Biomater ; 112(8): e35458, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39122663

ABSTRACT

Bacterial infections already pose a significant threat to skin wounds, especially in diabetic patients who have difficulty healing wounds. However, wound or bacterial infections are known to produce excess reactive oxygen species (ROS), and hypoxia may further hinder wound healing and the development of chronic wounds. In this study, a multifunctional hydrogel for ROS scavenging and bacterial inhibition was developed by cross-linking polyvinyl alcohol (PVA) and sodium alginate (SA) with graphene oxide (GO) loaded with silver-platinum hybrid nanoparticles (GO@Ag-Pt). The PVA/SA hydrogel loaded with GO@Ag-Pt exhibited the ability to scavenge different types of ROS, generate O2, and kill a broad spectrum of bacteria in vitro. The silver-platinum hybrid nanoparticles significantly increased the antibacterial ability against Escherichia coli and Staphylococcus aureus compared with silver nanoparticles (AgNps). GO@Ag-Pt loaded hydrogel was effective in treating infections caused by S.aureus, thereby significantly promoting wound healing during the inflammatory phase. Hydrogel therapy significantly reduced the level of ROS and alleviated inflammation levels. Notably, our ROS-scavenging, antibacterial hydrogels can be used to effectively treat various types of wounds, including difficult-to-heal diabetic wounds with bacterial infections. Thus, this study proposes an effective strategy for various chronic wound healing based on ROS clearance and bacteriostatic hydrogels.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Hydrogels , Metal Nanoparticles , Reactive Oxygen Species , Silver , Staphylococcus aureus , Wound Healing , Reactive Oxygen Species/metabolism , Wound Healing/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Staphylococcus aureus/drug effects , Animals , Metal Nanoparticles/chemistry , Silver/chemistry , Silver/pharmacology , Escherichia coli/drug effects , Mice , Graphite/chemistry , Graphite/pharmacology , Inflammation/drug therapy , Polyvinyl Alcohol/chemistry , Polyvinyl Alcohol/pharmacology , Humans , Alginates/chemistry , Alginates/pharmacology , Wound Infection/drug therapy , Staphylococcal Infections/drug therapy , Male , Oxygen/chemistry , Free Radical Scavengers/pharmacology , Free Radical Scavengers/chemistry
14.
Wounds ; 36(7): 221-226, 2024 07.
Article in English | MEDLINE | ID: mdl-39110945

ABSTRACT

BACKGROUND: The Kahramanmaras earthquakes in Turkey on February 6, 2023, resulted in more than 100 000 injuries and 50 500 deaths. The main causes of morbidity and mortality in earthquake-affected patients include crush syndrome, trauma-related extremity injuries, and wound infections. OBJECTIVE: To investigate infective complications, causative microorganisms, treatments, and treatment responses in pediatric patients. METHODS: The case series involved 12 earthquake victims admitted to a tertiary treatment center between February 9 and 24, 2023. Wound sample cultures were obtained from patients with infected wounds via wound secretions and tissue samples collected during surgery. RESULTS: Nine patients were male (75%), and the mean age of patients was 12.6 ± 3.7 years. Seven patients (58.3%) experienced crush syndrome. Seven patients (58.3%) underwent fasciotomy operations. The main infectious complications were wound infections (58.3%) and urinary tract infections (25%). Nine different organisms were found to cause wound infection, with Enterococcus faecium (41.6%), Acinetobacter baumanii (33.3%), and Pseudomonas aeruginosa (16.6%) being the major bacterial isolates. All Acinetobacter strains were multidrug-resistant (MDR). CONCLUSION: Major disasters such as earthquakes are rare, and infections are the major complications that increase morbidity and mortality. Initial appropriate treatment contributes to improved outcomes, as MDR strains are common pathogens in these patients.


Subject(s)
Earthquakes , Wound Infection , Humans , Male , Female , Wound Infection/microbiology , Child , Adolescent , Turkey/epidemiology , Anti-Bacterial Agents/therapeutic use , Crush Syndrome
15.
Int J Nanomedicine ; 19: 8159-8174, 2024.
Article in English | MEDLINE | ID: mdl-39139505

ABSTRACT

Background: Wound healing has always been a focal point in clinical work. Bacterial infections and immune microenvironment disorders can both hinder normal wound healing. Current wound dressings only serve a covering function. Developing wound dressings with antibacterial and immunomodulatory functions is crucial for aiding wound healing. To address this issue, we have developed a hydrogel with antibacterial and immunomodulatory functions for managing infected wounds. Methods: The present study describes a photo-crosslinked antibacterial hydrogel composed of curcumin, silver nanoparticles-loaded reduced graphene oxide, and silk fibroin methacryloyl for the treatment of infected wounds. The study assessed its antibacterial properties and its capacity to induce macrophage M2 polarization through in vitro and in vivo experiments. Results: The hydrogel demonstrates robust antibacterial properties and enhances macrophage M2 polarization in both in vitro and in vivo settings. Moreover, it accelerates the healing of infected wounds in vivo by stimulating collagen deposition and angiogenesis. Conclusion: Overall, this hydrogel shows great potential in managing wound infections.


Subject(s)
Anti-Bacterial Agents , Graphite , Hydrogels , Metal Nanoparticles , Silver , Wound Healing , Wound Infection , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Wound Healing/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Metal Nanoparticles/chemistry , Silver/chemistry , Silver/pharmacology , Mice , Graphite/chemistry , Graphite/pharmacology , Wound Infection/drug therapy , Curcumin/pharmacology , Curcumin/chemistry , Macrophages/drug effects , Fibroins/chemistry , Fibroins/pharmacology , RAW 264.7 Cells , Humans , Immunomodulating Agents/pharmacology , Immunomodulating Agents/chemistry , Male
16.
J Wound Care ; 33(8): 554-559, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39149927

ABSTRACT

OBJECTIVE: The aim was to evaluate the effectiveness of a marine omega fatty acid-containing multimodal wound matrix (MWM) in reducing bacterial contamination and supporting wound area reduction (WAR) in patients with hard-to-heal wounds of varying aetiologies. METHOD: A prospective, single-site, pilot case series of patients with hard-to-heal wounds. All wounds were considered non-healing prior to inclusion as they had failed to achieve at least 50% WAR after at least four weeks of standard of care (SoC) treatments. Patients were seen once weekly for wound assessments, matrix application and dressing changes. Baseline and weekly fluorescence images, standard wound images and wound measurements were obtained. RESULTS: A total of three patients, two with venous leg ulcers (VLUs) and one with a diabetic foot ulcer (DFU) were enrolled in this pilot study. The mean baseline wound age prior to study enrolment was 24 weeks, with a mean baseline wound size of 8.61cm2. The two VLUs went on to complete closure. The DFU displayed a total WAR of 53% by six weeks, when the patient was lost to follow-up due to a geographical relocation. The mean percentage area reduction of all wounds combined was 82% upon study completion. CONCLUSION: The use of MWM proved to be effective and safe in this patient cohort. The wounds included in this case series failed to enter a healing trajectory with SoC wound therapies. The MWM supported wound closure and reduced bacterial loads in this patient cohort.


Subject(s)
Diabetic Foot , Varicose Ulcer , Wound Healing , Humans , Pilot Projects , Male , Female , Diabetic Foot/therapy , Diabetic Foot/microbiology , Prospective Studies , Aged , Middle Aged , Varicose Ulcer/therapy , Varicose Ulcer/microbiology , Leg Ulcer/microbiology , Leg Ulcer/therapy , Wound Infection/drug therapy , Wound Infection/microbiology , Aged, 80 and over , Treatment Outcome
17.
Nat Commun ; 15(1): 7128, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164298

ABSTRACT

Recent advances in mass spectrometry-based peptidomics have catalyzed the identification and quantification of thousands of endogenous peptides across diverse biological systems. However, the vast peptidomic landscape generated by proteolytic processing poses several challenges for downstream analyses and limits the comparability of clinical samples. Here, we present an algorithm that aggregates peptides into peptide clusters, reducing the dimensionality of peptidomics data, improving the definition of protease cut sites, enhancing inter-sample comparability, and enabling the implementation of large-scale data analysis methods akin to those employed in other omics fields. We showcase the algorithm by performing large-scale quantitative analysis of wound fluid peptidomes of highly defined porcine wound infections and human clinical non-healing wounds. This revealed signature phenotype-specific peptide regions and proteolytic activity at the earliest stages of bacterial colonization. We validated the method on the urinary peptidome of type 1 diabetics which revealed potential subgroups and improved classification accuracy.


Subject(s)
Algorithms , Mass Spectrometry , Peptides , Proteolysis , Proteomics , Animals , Humans , Peptides/metabolism , Swine , Proteomics/methods , Mass Spectrometry/methods , Diabetes Mellitus, Type 1/metabolism , Wound Infection/microbiology , Wound Infection/metabolism , Cluster Analysis
18.
Mil Med ; 189(Supplement_3): 179-183, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160845

ABSTRACT

INTRODUCTION: The material of a bandage plays an important role in wound management. Microorganisms can colonize the dressing and release toxins, which create dead cells in the wound. This allows the microorganisms to bind the dead cells and infect the wound. Thus, a dressing is needed that kills bacteria in the bandage. To combat health care-associated infections, antimicrobial treatment of medical textiles, such as gauze, uniforms, curtains, bed sheets, gowns, and masks, is required. Besides, antimicrobial resistance is another major problem of this century. Antibacterial overuse has contributed to drug-resistant bacteria. To combat these two problems, we synthesized new organo-selenium compounds that can be attached to the cotton of the dressing. We then used an in vivo wound model, which allowed us to measure the effectiveness of selenium attached to a cotton dressing, to prevent bacteria from infecting a wound. MATERIALS AND METHODS: Organo-selenium was attached to cotton fabric, resulting in a fabric with 0.1% selenium covalently attached to it. Staphylococcus aureus (as well as methicillin-resistant S. aureus [MRSA]), Stenotrophomonas maltophilia, Enterococcus faecalis, Staphylococcus epidermidis, and Pseudomonas aeruginosa were chosen for the wound infection study. All the bacteria were enumerated in the wound dressing and in the wound tissue under the dressing. Wounds were made on the backs of mice. The material was used as a bandage over the wound. Bacteria were injected into the wound under the bandage. The amount of bacteria in the wound after 5 days was determined. A similar study was performed using dressing material that was soaked in phosphate buffered saline at 37 °C for 3 months before use. RESULTS: Cotton dressing with selenium attached showed complete inhibition (7 logs, as compared with control dressing) of different bacterial strains, in both the dressing and "the tissue" of the wound. Similar results were obtained using selenium cotton dressing that was soaked for 3 months before use. Control cotton with no selenium showed complete infiltration of bacteria into the wound and the dressing. In addition, a study was performed under Food and Drug Administration standard methods to show the ability of the selenium to kill bacteria in the fabric, using material that was washed 5 times in detergent. This also showed complete killing of bacteria in the fabric. CONCLUSIONS: The results show that the selenium remains in the dressing after washing and is able to completely protect the wound from bacterial infection. In the selenium bandage, no bacteria were found in the bandage or the wound after 5 days.


Subject(s)
Bandages , Cotton Fiber , Selenium , Wound Infection , Animals , Selenium/pharmacology , Selenium/therapeutic use , Wound Infection/drug therapy , Wound Infection/prevention & control , Wound Infection/microbiology , Wound Infection/therapy , Mice , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Staphylococcus aureus/drug effects
19.
Front Cell Infect Microbiol ; 14: 1395577, 2024.
Article in English | MEDLINE | ID: mdl-39145303

ABSTRACT

Introduction: Biofilm-associated infections persist as a therapeutic challenge in contemporary medicine. The efficacy of antibiotic therapies is ineffective in numerous instances, necessitating a heightened focus on exploring novel anti-biofilm medical strategies. Among these, iminosugars emerge as a distinctive class of compounds displaying promising biofilm inhibition properties. Methods: This study employs an in vivo wound infection mouse model to evaluate the effectiveness of PDIA in treating biofilm-associated skin wound infections caused by Staphylococcus aureus and Pseudomonas aeruginosa. Dermic wounds in mice were infected with biofilm-forming strains, specifically S. aureus 48 and P. aeruginosa 5, which were isolated from patients with diabetic foot, and are well-known for their strong biofilm formation. The subsequent analysis included clinical, microbiological, and histopathological parameters. Furthermore, an exploration into the susceptibility of the infectious strains to hydrogen peroxide was conducted, acknowledging its potential presence during induced inflammation in mouse dermal wounds within an in vivo model. Results: The findings revealed the efficacy of PDIA iminosugar against the S. aureus strain, evidenced by a reduction in bacterial numbers within the wound and the inflammatory focus. Discussion: This study suggests that PDIA iminosugar emerges as an active and potentially effective antibiofilm agent, positioning it as a viable treatment option for staphylococcal infections.


Subject(s)
Anti-Bacterial Agents , Biofilms , Disease Models, Animal , Pseudomonas Infections , Pseudomonas aeruginosa , Staphylococcal Infections , Staphylococcus aureus , Animals , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects , Mice , Biofilms/drug effects , Biofilms/growth & development , Pseudomonas Infections/microbiology , Pseudomonas Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Wound Infection/microbiology , Wound Infection/drug therapy , Humans , Female
20.
Nat Commun ; 15(1): 7538, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39214978

ABSTRACT

Development of potent and broad-spectrum antimicrobial peptides (AMPs) could help overcome the antimicrobial resistance crisis. We develop a peptide language-based deep generative framework (deepAMP) for identifying potent, broad-spectrum AMPs. Using deepAMP to reduce antimicrobial resistance and enhance the membrane-disrupting abilities of AMPs, we identify, synthesize, and experimentally test 18 T1-AMP (Tier 1) and 11 T2-AMP (Tier 2) candidates in a two-round design and by employing cross-optimization-validation. More than 90% of the designed AMPs show a better inhibition than penetratin in both Gram-positive (i.e., S. aureus) and Gram-negative bacteria (i.e., K. pneumoniae and P. aeruginosa). T2-9 shows the strongest antibacterial activity, comparable to FDA-approved antibiotics. We show that three AMPs (T1-2, T1-5 and T2-10) significantly reduce resistance to S. aureus compared to ciprofloxacin and are effective against skin wound infection in a female wound mouse model infected with P. aeruginosa. In summary, deepAMP expedites discovery of effective, broad-spectrum AMPs against drug-resistant bacteria.


Subject(s)
Anti-Bacterial Agents , Antimicrobial Peptides , Microbial Sensitivity Tests , Animals , Mice , Female , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Drug Resistance, Bacterial/drug effects , Staphylococcus aureus/drug effects , Pseudomonas aeruginosa/drug effects , Disease Models, Animal , Wound Infection/drug therapy , Wound Infection/microbiology , Humans , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Gram-Negative Bacteria/drug effects , Antimicrobial Cationic Peptides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL