Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 308
1.
Carbohydr Polym ; 279: 118778, 2022 Mar 01.
Article En | MEDLINE | ID: mdl-34980345

Eliminating biofilms from infected tissue presents one of the most challenging issues in clinical treatment of chronic wounds. In biofilms, the extracellular polymeric substances (EPS) form gel structures by electrostatic forces between macromolecules. We hypothesized that cationic polymers could induce the gel-to-sol phase transition of the network, leading to biofilms disruptions. We first validated this assumption by using polyethyleneimine (PEI) as a model molecule, and further synthesized two cationic dextrans with high biodegradability for in vitro and in vivo evaluation. All the cationic polymers could destruct Pseudomonas aeruginosa (P. aeruginosa) biofilms. Treating biofilm with cationic dextrans significantly enhanced the bacterial antibiotic sensitivity. When tested in a biofilm-presenting mouse wound healing model, the cationic dextrans efficiently controlled infection, and accelerated the healing process. Our findings suggest that devising cationic polymers to trigger phase transition of biofilm is an effective, straightforward, and perhaps generic strategy for anti-bacterial therapies.


Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Dextrans/pharmacology , Hydrogels/pharmacology , Pseudomonas aeruginosa/drug effects , Alginates/chemistry , Animals , Cell Line , Cell Survival/drug effects , Cytokines/immunology , Dextrans/chemistry , Female , Hydrogels/chemistry , Mice, Inbred BALB C , Phase Transition , Polyethyleneimine/chemistry , Pseudomonas Infections/drug therapy , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/physiology , Skin/drug effects , Skin/immunology , Wound Infection/drug therapy , Wound Infection/immunology
2.
Int Immunopharmacol ; 100: 108094, 2021 Nov.
Article En | MEDLINE | ID: mdl-34508942

This work evaluated the immunomodulatory and anti-infective effects of Cratylia mollis lectin (Cramoll) in a model of wound infection induced by S. aureus. Swiss mice were divided into 3 groups (n = 12/group): non-inoculated (Control group); inoculated with S. aureus (Sa group); inoculated with S. aureus and treated with Cramoll (Sa + Cramoll group). In each animal, one lesion (64 mm2) was induced on the back and contaminated with S. aureus (~4.0 × 106 CFU/wound). The treatment with Cramoll (5 µg/animal/day) started 1-day post-infection (dpi) and extended for 10 days. Clinical parameters (wound size, inflammatory aspects, etc.) were daily recorded; while cytokines levels, bacterial load and histological aspects were determined in the cutaneous tissue at 4th dpi or 11th dpi. The mice infected with S. aureus exhibited a delay in wound contraction and the highest inflammatory scores. These effects were impaired by the treatment with Cramoll which reduced the release of key inflammatory mediators (TNF-α, NO, VEGF) and the bacterial load at wound tissue. Histological evaluations showed a restauration of skin structures in the animals treated with Cramoll. Taken together, these results provide more insights about the healing and immunomodulatory properties of Cramoll and suggest this lectin as a lead compound for treatment of wound infection.


Anti-Bacterial Agents/pharmacology , Fabaceae , Immunomodulating Agents/pharmacology , Plant Lectins/pharmacology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/drug effects , Wound Infection/prevention & control , Animals , Anti-Bacterial Agents/isolation & purification , Bacterial Load , Disease Models, Animal , Fabaceae/chemistry , Host-Pathogen Interactions , Immunomodulating Agents/isolation & purification , Mice , Nitric Oxide/metabolism , Plant Lectins/isolation & purification , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/growth & development , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/drug effects , Wound Infection/immunology , Wound Infection/metabolism , Wound Infection/microbiology
3.
Int J Mol Sci ; 22(15)2021 Jul 30.
Article En | MEDLINE | ID: mdl-34360986

Human Cytomegalovirus (HCMV) may cause severe infections in transplant recipients. HCMV-replication can be limited by HCMV-specific antibody responses. The impact of the antibody-dependent cellular phagocytosis (ADCP) on inhibition of HCMV-replication in natural infections has not been clarified. Therefore, we investigated the HCMV-specific ADCP response in a study cohort of lung-transplant recipients (LTRs) with different donor (D) and recipient (R) HCMV-serostatus. Follow-up plasma samples from 39 non/low-viremic and 36 highly viremic (>1000 HCMV copies/mL plasma) LTRs were collected for one (R+ LTRs) or two (D+/R- LTRs) years post-transplantation. The HCMV-specific ADCP responses were assessed by focal expansion assays (FEA) and flow-cytometry. In all LTRs, ADCP responses were detected against HCMV-infected cells and cell-free virions. When measured in fibroblasts as well as with cell-free virus, the HCMV-specific ADPC response was higher in LTRs than in HCMV-seropositive healthy controls. In D+/R- LTRs, a significant ADCP response developed over time after the receipt of an HCMV positive lung, and a level of <19 IE+ cells/focus in the FEA on fibroblasts was associated with further protection from high-level viremia. Taken together, a strong HCMV-specific ADCP response is elicited in transplant recipients, which may contribute to protection from high-level viremia in primary HCMV infection.


Cytomegalovirus Infections/immunology , Immunoglobulin G/immunology , Lung Transplantation/adverse effects , Phagocytosis , Wound Infection/immunology , Cells, Cultured , Cytomegalovirus/immunology , Cytomegalovirus Infections/etiology , Cytomegalovirus Infections/virology , Female , Humans , Male , Middle Aged , THP-1 Cells , Viral Load , Wound Infection/etiology , Wound Infection/virology
4.
Toxins (Basel) ; 13(3)2021 03 22.
Article En | MEDLINE | ID: mdl-33810194

Staphylococcus aureus is the most prevalent pathogen isolated from diabetic foot infections (DFIs). The purpose of this study was to evaluate its behavior in an in vitro model mimicking the conditions encountered in DFI. Four clinical S. aureus strains were cultivated for 16 weeks in a specific environment based on the wound-like medium biofilm model. The adaptation of isolates was evaluated as follows: by Caenorhabditis elegans model (to evaluate virulence); by quantitative Reverse Transcription-Polymerase Chain Reaction (qRT-PCR) (to evaluate expression of the main virulence genes); and by Biofilm Ring test® (to assess the biofilm formation). After 16 weeks, the four S. aureus had adapted their metabolism, with the development of small colony variants and the loss of ß-hemolysin expression. The in vivo nematode model suggested a decrease of virulence, confirmed by qRT-PCRs, showing a significant decrease of expression of the main staphylococcal virulence genes tested, notably the toxin-encoding genes. An increased expression of genes involved in adhesion and biofilm was noted. Our data based on an in vitro model confirm the impact of environment on the adaptation switch of S. aureus to prolonged stress environmental conditions. These results contribute to explore and characterize the virulence of S. aureus in chronic wounds.


Biofilms/growth & development , Diabetic Foot/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/growth & development , Wound Infection/microbiology , Diabetic Foot/immunology , Energy Metabolism , Gene Expression Regulation, Bacterial , Humans , Immune Evasion , Staphylococcal Infections/immunology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Time Factors , Virulence , Wound Infection/immunology
5.
Sci Rep ; 10(1): 21818, 2020 12 11.
Article En | MEDLINE | ID: mdl-33311597

Burn wounds are highly susceptible sites for colonization and infection by bacteria and fungi. Large wound surface, impaired local immunity, and broad-spectrum antibiotic therapy support growth of opportunistic fungi such as Candida albicans, which may lead to invasive candidiasis. Currently, it remains unknown whether depressed host defenses or fungal virulence drive the progression of burn wound candidiasis. Here we established an ex vivo burn wound model, where wounds were inflicted by applying preheated soldering iron to human skin explants, resulting in highly reproducible deep second-degree burn wounds. Eschar removal by debridement allowed for deeper C. albicans penetration into the burned tissue associated with prominent filamentation. Active migration of resident tissue neutrophils towards the damaged tissue and release of pro-inflammatory cytokine IL-1ß accompanied the burn. The neutrophil recruitment was further increased upon supplementation of the model with fresh immune cells. Wound area and depth decreased over time, indicating healing of the damaged tissue. Importantly, prominent neutrophil presence at the infected site correlated to the limited penetration of C. albicans into the burned tissue. Altogether, we established a reproducible burn wound model of candidiasis using ex vivo human skin explants, where immune responses actively control the progression of infection and promote tissue healing.


Burns/immunology , Candida albicans/immunology , Candidiasis/immunology , Neutrophils/immunology , Skin/immunology , Wound Infection/immunology , Adult , Burns/microbiology , Burns/pathology , Candidiasis/pathology , Female , Humans , Interleukin-1beta/immunology , Middle Aged , Neutrophils/pathology , Skin/microbiology , Skin/pathology , Wound Infection/microbiology , Wound Infection/pathology
6.
Sci Rep ; 10(1): 11494, 2020 07 13.
Article En | MEDLINE | ID: mdl-32661287

Immunocompromised patients are predisposed to chronically infected wounds. Especially ulcers in the dorsal region often experience secondary polymicrobial infections. However, current wound infection models mostly use single-strain bacteria. To mimic clinically occurring infections caused by fecal contamination in immunocompromised/immobile patients, which differ significantly from single-strain infections, the present study aimed at the establishment of a new mouse model using infection by fecal bacteria. Dorsal circular excision wounds in immunosuppressed mice were infected with fecal slurry solution in several dilutions up to 1:8,000. Impact of immunosuppressor, bacterial load and timing on development of wound infections was investigated. Wounds were analyzed by scoring, 3D imaging and swab analyses. Autofluorescence imaging was not successful. Dose-finding of cyclophosphamide-induced immunosuppression was necessary for establishment of bacterial wound infections. Infection with fecal slurry diluted 1:166 to 1:400 induced significantly delayed wound healing (p < 0.05) without systemic reactions. Swab analyses post-infection matched the initial polymicrobial suspension. The customized wound score confirmed significant differences between the groups (p < 0.05). Here we report the establishment of a simple, new mouse model for clinically occurring wound infections by fecal bacteria and the evaluation of appropriate wound analysis methods. In the future, this model will provide a suitable tool for the investigation of complex microbiological interactions and evaluation of new therapeutic approaches.


Coinfection/drug therapy , Feces/microbiology , Wound Infection/drug therapy , Wounds and Injuries/drug therapy , Animals , Anti-Bacterial Agents/pharmacology , Coinfection/immunology , Coinfection/microbiology , Coinfection/pathology , Disease Models, Animal , Humans , Immunocompromised Host/drug effects , Immunocompromised Host/immunology , Immunosuppression Therapy/adverse effects , Mice , Wound Healing/drug effects , Wound Healing/immunology , Wound Infection/immunology , Wound Infection/microbiology , Wound Infection/pathology , Wounds and Injuries/immunology , Wounds and Injuries/microbiology , Wounds and Injuries/pathology
7.
Surg Clin North Am ; 100(4): 681-693, 2020 Aug.
Article En | MEDLINE | ID: mdl-32681869

Chronic wounds present a unique therapeutic challenge to heal. Chronic wounds are colonized with bacteria and the presence of a biofilm that further inhibits the normal wound healing processes, and are locked into a very damaging proinflammatory response. The treatment of chronic wounds requires a coordinated approach, including debridement of devitalized tissue, minimizing bacteria and biofilm, control of inflammation, and the use of specialized dressings to address the specific aspects of the particular nonhealing ulcer.


Diabetic Angiopathies/physiopathology , Skin Ulcer/physiopathology , Wound Healing/physiology , Anti-Infective Agents/therapeutic use , Biofilms/drug effects , Chronic Disease , Cytokines/physiology , Diabetic Angiopathies/immunology , Diabetic Angiopathies/therapy , Drug Resistance, Bacterial/physiology , Drug Therapy, Combination , Humans , Immunity, Cellular/physiology , Peptide Hydrolases/physiology , Skin Ulcer/immunology , Skin Ulcer/therapy , Wound Healing/immunology , Wound Infection/immunology , Wound Infection/physiopathology , Wound Infection/therapy
8.
Adv Wound Care (New Rochelle) ; 9(2): 35-47, 2020 02 01.
Article En | MEDLINE | ID: mdl-31903297

Objective: Our goal was to develop a chronic wound model in mice that avoids implantation of foreign material or impaired immunity and to use this to characterize the local and systemic immune response associated with Pseudomonas aeruginosa infection. Approach: We generated bilateral full-thickness dermal wounds in healthy 10-12-week-old C57Bl6 mice. We waited 24 h to inoculate the developing wound eschar at these sites. We performed careful titration experiments with luminescent strains of P. aeruginosa to identify bacterial inoculation concentrations that consistently established stable infections in these animals. We performed flow cytometry-based immunophenotyping of immune cell infiltrates at the wound site, spleen, and draining lymph nodes over time. Finally, we compared inflammatory responses seen in wound inoculation with planktonic bacteria, preformed biofilm, and heat-killed (HK) P. aeruginosa. Results: Using this delayed inoculation model and 7.5 ± 2.5 × 102 CFU/mL of PAO1 we consistently established stable infections that lasted at 10 days in duration. During early infection, we detected a strong upregulation of inflammatory cytokines and neutrophil infiltration at the wound site, while natural killer (NK) cells and dendritic cells (DCs) were reduced. At the systemic level, only plasmacytoid DCs were increased early in infection. During later stages, there was systemic upregulation of B cells, T cells, and macrophages, whereas NK cells and interferon killer DCs were reduced. Infections with P. aeruginosa biofilms were not more virulent than infections with planktonic P. aeruginosa, whereas treatment with HK P. aeruginosa only induces a short-term inflammatory state. Innovation: We describe a versatile wound model of chronic P. aeruginosa infection that lasts 10 days without causing sepsis or other excessive morbidity. Conclusion: This model may facilitate the study of chronic wound infections in immunocompetent mice. Our findings also highlight the induction of early innate immune cell populations during P. aeruginosa infection.


Biofilms/growth & development , Cytokines/immunology , Pseudomonas Infections/immunology , Wound Infection/immunology , Wound Infection/microbiology , Animals , Disease Models, Animal , Immunity , Immunocompetence , Male , Mice , Mice, Inbred C57BL , Pseudomonas aeruginosa , Wound Infection/pathology
9.
Pathog Dis ; 78(5)2020 07 01.
Article En | MEDLINE | ID: mdl-31116394

The majority of chronic wounds are associated with bacterial biofilms recalcitrant to antibiotics and host responses. Immunomodulatory S100A8/A9 is suppressed in Pseudomonas aeruginosa biofilm infected wounds. We aimed at investigating a possible additive effect between S100A8/A9 and ciprofloxacin against biofilms. MATERIALS/METHODS: Thirty-two mice were injected with alginate-embedded P. aeruginosa following a third-degree burn. The mice were randomized into four groups receiving combination ciprofloxacin and S100A8/A9 or monotherapy ciprofloxacin, S100A8/A9 or a placebo and evaluated by host responses and quantitative bacteriology in wounds. In addition, in vitro checkerboard analysis was performed, with P. aeruginosa and ascending S100A8/A9 and ciprofloxacin concentrations. RESULTS: S100A8/A9 augmented the effect of ciprofloxacin in vivo by lowering the bacterial quantity compared to the placebo arm and the two monointervention groups (P < 0.0001). S100A8 and 100A9 were increased in the double-treated group as compared to the monointervention groups (P = 0.032, P = 0.0023). Tissue inhibitor of metalloproteinases-1 and keratinocyte\chemokine chemoattractant-1 were increased in the double-intervention group compared to the S100A8/A9 group (P = 0.050, P = 0.050). No in vitro synergism was detected. CONCLUSION: The observed ciprofloxacin-augmenting effect of S100A8/A9 in vivo was not confirmed by checkerboard analysis, indicating dependence on host cells for the S100A8/A9 effect. S100A8/A9 and ciprofloxacin is a promising therapy for optimizing chronic wound treatment.


Biofilms/drug effects , Calgranulin A/physiology , Ciprofloxacin/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Wound Infection/immunology , Wound Infection/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Chronic Disease , Colony Count, Microbial , Cytokines/metabolism , Disease Models, Animal , Drug Synergism , Female , Host-Pathogen Interactions , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred BALB C , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology
10.
Methods Mol Biol ; 2069: 47-58, 2020.
Article En | MEDLINE | ID: mdl-31523764

Immunofluorescence microscopy is a widely used laboratory method which allows detection and visualization of specific antigens. The method employs the specificity of antibodies to deliver fluorophore to a specific target and then visualize it with a microscope. The power of the technique is that it requires relatively little manipulation and relatively few bacterial cells, enabling the detection of antigen expression where other methods cannot, such as during an actual infection in an animal. Here, we apply the method to follow antigen expression on the surface of MRSA cells over time in in vivo infection models.


Antigens, Bacterial/immunology , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Wound Infection , Animals , Disease Models, Animal , Humans , Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Microscopy, Fluorescence , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Wound Infection/immunology , Wound Infection/microbiology , Wound Infection/pathology
11.
Proc Natl Acad Sci U S A ; 116(41): 20500-20504, 2019 10 08.
Article En | MEDLINE | ID: mdl-31548430

Skin wound infections are a significant health problem, and antibiotic resistance is on the rise. Mast cells (MCs) have been shown to contribute to host-defense responses in certain bacterial infections, but their role in skin wound superinfection is unknown. We subjected 2 MC-deficient mouse strains to Pseudomonas aeruginosa skin wound infection and found significantly delayed wound closure in infected skin wounds. This delay was associated with impaired bacterial clearance in the absence of MCs. Engraftment of MCs restored both bacterial clearance and wound closure. Bacterial killing was dependent on IL-6 released from MCs, and engraftment with IL-6-deficient MCs failed to control wound infection. Treatment with recombinant IL-6 enhanced bacterial killing and resulted in the control of wound infection and normal wound healing in vivo. Taken together, our results demonstrate a defense mechanism for boosting host innate immune responses, namely effects of MC-derived IL-6 on antimicrobial functions of keratinocytes.


Keratinocytes/immunology , Mast Cells/immunology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/immunology , Skin/immunology , Wound Healing/immunology , Wound Infection/prevention & control , Animals , Anti-Bacterial Agents/pharmacology , Cells, Cultured , Humans , Interleukin-6/pharmacology , Keratinocytes/drug effects , Mast Cells/cytology , Mice , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Skin/drug effects , Wound Healing/drug effects , Wound Infection/immunology , Wound Infection/microbiology
12.
J Trauma Acute Care Surg ; 87(2): 337-341, 2019 08.
Article En | MEDLINE | ID: mdl-31008865

BACKGROUND: Trauma induces a complex immune response, requiring a systems biology approach to capture multicellular changes. Using mass cytometry by time-of-flight (CyTOF), we evaluated time-dependent changes in peripheral blood in trauma patients to identify changes correlated with infection. METHODS: Total leukocytes were prepared via red blood cell lysis using peripheral blood samples from trauma patients with an Injury Severity Score greater than 20 at Days 1, 3, and 5 after injury, and from age- and sex-matched uninjured controls. Cells were stained using a 33-marker immunophenotyping CyTOF panel. Statistics were calculated using one-way analysis of variance with multiple comparisons. RESULTS: The CyTOF staining demonstrated changes in many cell subsets. The mean expression intensity of CD86 on monocytes decreased significantly at all time points after injury. When the patients were stratified based on development of infection, there was a trend to decreased CD86 expression on monocytes of those patients that developed subsequent infection. Based on stratification, we identified significantly increased expression of CD39 on NK cells only in patients that developed an infection. CONCLUSION: This study used a systems biology approach to identify novel changes in circulating immune cell subsets in trauma patients correlating with post-traumatic infection. Decreased expression of CD86, a costimulatory molecule, on monocytes demonstrates that trauma affects the innate system's ability to control T-cell immunity. We also found that CD39 expression on NK cells increased significantly in patients with subsequent infection. CD39 is a protein that generates adenosine, which has immunosuppressive effects on several immune cell types including NK cells. In summary, our results point to pathways that may be central to second-hit infections and further study to delineate these pathways could be key to generating clinical biomarkers or targeted immune therapies for trauma patients. LEVEL OF EVIDENCE: Prognostic study, level II.


Killer Cells, Natural , Monocytes , Wound Infection/etiology , Wounds and Injuries/complications , Case-Control Studies , Female , Flow Cytometry , Humans , Immunophenotyping , Killer Cells, Natural/physiology , Male , Middle Aged , Monocytes/physiology , Phenotype , Wound Infection/immunology , Wounds and Injuries/immunology
13.
Science ; 363(6434)2019 03 29.
Article En | MEDLINE | ID: mdl-30923196

Bacteriophage are abundant at sites of bacterial infection, but their effects on mammalian hosts are unclear. We have identified pathogenic roles for filamentous Pf bacteriophage produced by Pseudomonas aeruginosa (Pa) in suppression of immunity against bacterial infection. Pf promote Pa wound infection in mice and are associated with chronic human Pa wound infections. Murine and human leukocytes endocytose Pf, and internalization of this single-stranded DNA virus results in phage RNA production. This triggers Toll-like receptor 3 (TLR3)- and TIR domain-containing adapter-inducing interferon-ß (TRIF)-dependent type I interferon production, inhibition of tumor necrosis factor (TNF), and the suppression of phagocytosis. Conversely, immunization of mice against Pf prevents Pa wound infection. Thus, Pf triggers maladaptive innate viral pattern-recognition responses, which impair bacterial clearance. Vaccination against phage virions represents a potential strategy to prevent bacterial infection.


Immune Tolerance , Phagocytosis/immunology , Pseudomonas Infections/immunology , Pseudomonas Phages/physiology , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/virology , Wound Infection/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Animals , Antibodies, Viral/immunology , Humans , Interferons/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Pseudomonas Phages/immunology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Tumor Necrosis Factor-alpha/metabolism
14.
Drug Deliv Transl Res ; 9(4): 748-763, 2019 08.
Article En | MEDLINE | ID: mdl-30652257

Staphylococcus aureus (SA) and methicillin-resistant Staphylococcus aureus (MRSA) have been a major cause of morbidity in thermally injured patients. The skin barrier gets disrupted and loss of immunity further makes burn sites an easy target for bacterial colonization. In the current study, combined potential of lipid-polymer hybrid nanoparticles (LPHNs) with fusidic acid was explored as a promising strategy toward combating resistant bacteria in burn wound infection sites. The developed systems exhibited particle size (310.56 ± 5.22 nm), zeta potential (24.3 ± 4.18 mV) and entrapment efficiency (78.56 ± 3.56%) with a spherical shape. The hybrid nanoparticles were further gelled into carbopol and demonstrated better permeation (76.53 ± 1.55%) and retention characteristics (56.41 ± 4.67%) as compared to the conventional formulation. The topical delivery of FA into the skin layers by FA-LPHN gel was found to be significantly higher (p < 0.05) compared to FA-CC. The in vivo potential was further assessed in murine burn wound model inflicted with MRSA 33591 bacterium with the determination of parameters like bacterial burden, wound contraction, morphological and histopathological examination of wounds. The bacterial count decreased drastically in FA-LPHN gel group (5.22 log CFU/mL) on day 3 with significant difference in comparison to FA-CC. The wound size reduction in FA-LPHN gel (68.70 ± 3.65%) was higher as compared to FA-CC (73.30 ± 4.23%) and control groups (83.30 ± 4.40%) on day 5. The current study presents a safe and effective formulation strategy for the treatment of MRSA-infected burn wounds by providing moist environment and prevention from bacterial infection.


Burns/drug therapy , Drug Carriers/administration & dosage , Fusidic Acid/administration & dosage , Methicillin-Resistant Staphylococcus aureus , Nanoparticles/administration & dosage , Skin/metabolism , Staphylococcal Infections/drug therapy , Wound Infection/drug therapy , Animals , Burns/immunology , Burns/microbiology , Cytokines/immunology , Drug Carriers/pharmacokinetics , Female , Fusidic Acid/pharmacokinetics , Gels , Lipids/administration & dosage , Lipids/pharmacokinetics , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Mice, Inbred BALB C , Polymers/administration & dosage , Polymers/pharmacokinetics , Rats, Wistar , Skin/drug effects , Skin/immunology , Skin Absorption , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Wound Infection/immunology , Wound Infection/microbiology
15.
Article En | MEDLINE | ID: mdl-31998665

Burn wound injury affects soldiers and civilians alike, often resulting in a dynamic, but un-orchestrated, host response that can lead to infection, scarring, and potentially death. To mitigate these factors, it is important to have a clinically relevant model of burn wound infection that can be utilized for advancing burn wound treatments. Our previous reports have demonstrated the ability of Pseudomonas aeruginosa to generate a biofilm infection within a modified Walker-Mason rat burn model of deep-partial (DPT) and full-thickness (FT) burn wounds (10% total body surface area) in male Sprague-Dawley rats (350-450 g). Here, we further define this model with respect to the host response when challenged with P. aeruginosa infection between the two burn types. Following burn injury and immediate surface exposure to P. aeruginosa, inflammation at the local and systemic levels were monitored for an 11 days period. Compared to burn-only groups, infection with P. aeruginosa further promoted local inflammation in both DPT and FT burn wounds, which was evident by enhanced cellular influx (including neutrophils and monocytes), increased levels of several pro-inflammatory cytokines (IL-1ß, IL-6, GRO/KC, andMIP-1α), and reduced IL-10. Systemically, only minor changes were seen in circulating white blood cells and cytokines; however, increases in high mobility group box-1 (HMGB-1) and hyaluronan, as well as decreases in fibronectin were noted particularly in FT burns. Compared to the burn-only group, P. aeruginosa infection resulted in sustained and/or higher levels of HMGB-1 and hyaluronan. Combined with our previous work that defined the burn depth and development of P. aeruginosa biofilms within the wound, this study further establishes this model by defining the host response to the burn and biofilm-infection. Furthermore, this characterization shows several similarities to what is clinically seen and establishes this model for future use in the development and testing of novel therapeutics for burn wound treatment at home and on the battlefield.


Burns/immunology , Burns/microbiology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Wound Infection/immunology , Animals , Biofilms/growth & development , Chemokine CCL3/metabolism , Chemokines/blood , Chemokines/metabolism , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Inflammation , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Leukocyte Count , Male , Monocytes , Neutrophils , Rats , Rats, Sprague-Dawley
16.
Wounds ; 30(11): 345-352, 2018 Nov.
Article En | MEDLINE | ID: mdl-30418163

Antimicrobial approaches (eg, antibiotics and antiseptics) have been used for decades in the treatment of infected wounds, ulcers, and burns. However, an increasing number of meta-analyses have raised questions regarding the therapeutic value of these approaches. Newer findings show that the body actively hosts an ecosystem of bacteria, fungi, viruses, and mites on its outer surfaces, known as the microbiome, as part of its defense against pathogens. Antimicrobials would disrupt this system and thereby work against the strategy the body has chosen. Recently, a new technology, micropore particle technology (MPPT), has been identified; it is not an antimicrobial but instead acts as a passive immunotherapy that disrupts the weaponry bacteria and fungi use to inhibit the immune system, allowing the immune system to recover. Clinical findings show MPPT removes wound infections 60% quicker than antibiotics and antiseptics and promotes the healing of chronic wounds that have not responded to antimicrobials. These effects are achieved without antimicrobial action and, considering the limited therapeutic benefits of antibiotics and antiseptics for wound infections, it is valid to question the use of antimicrobial approaches in wound care and the dogma that a reduction in microbial burden will lead to a reduction in infection. Instead, it may be time to consider a paradigm shift in wound healing away from antimicrobials and towards therapies that support the immune system and the microbiome. This review covers the increasing evidence that infections on external surfaces have to be treated fundamentally differently to internal infections.


Biomedical Technology/trends , Microbiota/physiology , Re-Epithelialization/immunology , Skin Physiological Phenomena/immunology , Wound Healing/physiology , Wound Infection/immunology , Anti-Infective Agents , Humans , Immunotherapy/trends , Wound Healing/immunology , Wound Infection/therapy
17.
J Surg Res ; 231: 448-452, 2018 11.
Article En | MEDLINE | ID: mdl-30278967

BACKGROUND: Systemic inflammatory response syndrome (SIRS) is associated with organ failure and infectious complications after major burn injury. Recent evidence has linked melanocortin signaling to anti-inflammatory and wound-repair functions, with mutations in the melanocortin 1 receptor (MC1R) gene leading to increased inflammatory responses. Our group has previously demonstrated that MC1R gene polymorphisms are associated with postburn hypertrophic scarring. Thus, we hypothesized that MC1R single nucleotide polymorphisms (SNPs) would be associated with increased burn-induced SIRS and increased infectious complications. METHODS: We performed a retrospective cohort study of adults (>18 y of age) admitted to our burn center with >20% total body surface area (TBSA) partial/full thickness burns between 2006 and 2013. We screened for five MC1R SNPs (V60L, V92M, R151C, R163Q, T314T) by polymerase chain reaction from genomic DNA isolated from blood samples. We performed a detailed review of each patient chart to identify age, sex, race, ethnicity, %TBSA burned, burn wound infections (BWIs), and 72-hr intravenous fluid volume, the latter a surrogate for a dysfunctional inflammatory response to injury. Association testing was based on multivariable regression. RESULTS: Of 106 subjects enrolled, 82 had complete data for analysis. Of these, 64 (78%) were male, with a median age of 39 and median burn size of 30% TBSA. A total of 36 (44%) subjects developed BWIs. The median total administered IV crystalloid in first 72h was 24.6 L. In multivariate analysis, the R151C variant allele was a significant independent risk factor for BWI (adjusted prevalence ratio 2.03; 95% CI: 1.21-3.39; P = 0.007), and the V60L variant allele was independently associated with increased resuscitation fluid volume (P = 0.021). CONCLUSIONS: This is the first study to demonstrate a significant association between genetic polymorphisms and a nonfatal burn-induced SIRS complication. Our findings suggest that MC1R polymorphisms contribute to dysfunctional responses to burn injury that may predict infectious and inflammatory complications.


Burns/complications , Polymorphism, Single Nucleotide , Receptor, Melanocortin, Type 1/genetics , Systemic Inflammatory Response Syndrome/genetics , Wound Infection/genetics , Adolescent , Adult , Aged , Burns/genetics , Burns/immunology , Female , Genetic Markers , Genotyping Techniques , Humans , Linear Models , Male , Middle Aged , Multivariate Analysis , Receptor, Melanocortin, Type 1/immunology , Retrospective Studies , Systemic Inflammatory Response Syndrome/immunology , Wound Infection/immunology , Young Adult
18.
Biomater Sci ; 6(9): 2472-2486, 2018 Aug 21.
Article En | MEDLINE | ID: mdl-30066700

Burn wounds are associated with a series of risks, such as infection and pathologic scar tissue formation, which significantly delay wound healing and lead to complications. In this study, we successfully fabricated a dextran-hyaluronic acid (Dex-HA) hydrogel enriched with sanguinarine (SA) incorporated into gelatin microspheres (GMs), which had high porosity, good swelling ratio, enhanced NIH-3T3 fibroblast cell proliferation, and sustained SA release profile. The in vitro degradation results indicate that the SA/GMs/Dex-HA hydrogel can be degraded. The in vitro antibacterial tests showed that the SA/GMs/Dex-HA hydrogel can inhibit methicillin-resistant Staphylococcus aureus (MRSA) and Escherichia coli (E. coli). We evaluated the wound-healing effects and antibacterial properties of SA/GMs/Dex-HA hydrogels in a rat full-thickness burn infection model. The hematoxylin-eosin (H&E) and Masson's trichrome staining results of the SA/GMs/Dex-HA hydrogel showed that it improved re-epithelialization and enhanced extracellular matrix remodeling, and immunohistochemistry results showed that the expression of TGF-ß1 and TNF-α was decreased, while the TGF-ß3 expression was increased. Our findings suggest that the SA/GMs/Dex-HA hydrogel provides a potential way for infected burn treatment with high-quality and efficient scar inhibition.


Anti-Bacterial Agents/administration & dosage , Benzophenanthridines/administration & dosage , Dextrans/administration & dosage , Hyaluronic Acid/administration & dosage , Hydrogels/administration & dosage , Isoquinolines/administration & dosage , Staphylococcal Infections/drug therapy , Wound Infection/drug therapy , Animals , Anti-Bacterial Agents/chemistry , Benzophenanthridines/chemistry , Burns/drug therapy , Cell Survival/drug effects , Dextrans/chemistry , Escherichia coli/drug effects , Escherichia coli/growth & development , Gelatin/administration & dosage , Gelatin/chemistry , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Isoquinolines/chemistry , Male , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Mice , NIH 3T3 Cells , Rats, Sprague-Dawley , Skin/drug effects , Skin/immunology , Skin/pathology , Staphylococcal Infections/immunology , Staphylococcal Infections/pathology , Transforming Growth Factor beta/immunology , Tumor Necrosis Factor-alpha/immunology , Wound Healing/drug effects , Wound Infection/immunology , Wound Infection/pathology
19.
Microb Pathog ; 123: 426-432, 2018 Oct.
Article En | MEDLINE | ID: mdl-30075242

Considering the increased antibiotic resistance of Pseudomonas aeruginosa, the evaluation of immune response against the antigens of this bacterium seems necessary. In this study, the protective efficacy and immunological properties of P. aeruginosa recombinant PilQ (r-PilQ) and type b-flagellin (FLB) proteins was evaluated in the burn mouse model of infection. The inbred BALB/c mice were immunized with r-PilQ and FLB antigens. To investigate the type of induced immune response, sera were analyzed by ELISA for total IgG, IgG1, and IgG2a isotypes. After the final immunization, the IL-4, IFN-γ, and IL-17 cytokines level were examined in the spleen of non-challenged mice. Fifty days after lethal challenge, the survival rate and bacterial burden in the skin and other internal organs of experimental mice were assessed. The in vivo administration of r-PilQ, FLB and combined antigen resulted in a significant increase in the survival of mice (66%, 75%, and 83%, respectively) infected by the PAO1 strain of P. aeruginosa in the burn model of infection. Immunization of mice with r-PilQ and FLB mixture induced high titers of IL-4 and IL-17 cytokines compared to control groups (P < 0.05). The high titer of antisera raised against combined antigen was able to inhibit the systemic spread of the PAO1 strain from the site of infection to the internal organs. We concluded that the parallel role of IL-4 and IL-17 is necessary for elimination of the bacteria and promotion of survival in the immunized burn mice.


Bacterial Vaccines/immunology , Burns/immunology , Fimbriae Proteins/immunology , Flagellin/immunology , Pseudomonas Infections/immunology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/immunology , Wound Infection/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Burns/microbiology , Cytokines/metabolism , Disease Models, Animal , Female , Fimbriae Proteins/administration & dosage , Fimbriae Proteins/genetics , Flagellin/administration & dosage , Flagellin/genetics , Immunity, Humoral , Immunization , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunologic Factors/metabolism , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred BALB C , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Recombinant Proteins , Spleen/immunology , Survival Rate , Wound Infection/microbiology , Wound Infection/prevention & control
20.
Sci Rep ; 8(1): 10738, 2018 Jul 16.
Article En | MEDLINE | ID: mdl-30013112

Chronic infections are often associated with the presence of a biofilm, a community of microorganisms coexisting within a protective matrix of extracellular polymeric substance. Living within a biofilm can make resident microbes significantly more tolerant to antibiotics in comparison to planktonic, free-floating cells. Thus, agents that can degrade biofilms are being pursued for clinical applications. While biofilm degrading and dispersing agents may represent attractive adjunctive therapies for biofilm-associated chronic infections, very little is known about how the host responds to the sudden dispersal of biofilm cells. In this study, we found that large-scale, in vivo dispersal of motile biofilm bacteria by glycoside hydrolases caused lethal septicemia in the absence of antibiotic therapy in a mouse wound model. However, when administered prudently, biofilm degrading enzymes had the potential to potentiate the efficacy of antibiotics and help resolve biofilm-associated wound infections.


Biofilms/drug effects , Glycoside Hydrolases/adverse effects , Pseudomonas aeruginosa/pathogenicity , Sepsis/chemically induced , Wound Infection/drug therapy , Animals , Anti-Bacterial Agents/administration & dosage , Biofilms/growth & development , Disease Models, Animal , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/methods , Glycoside Hydrolases/administration & dosage , Humans , Mice , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/immunology , Sepsis/immunology , Sepsis/mortality , Skin/injuries , Skin/microbiology , Wound Infection/immunology , Wound Infection/microbiology
...