RESUMO
The biofilm-induced "relatively immune-compromised zone" creates an immunosuppressive microenvironment that is a significant contributor to refractory infections in orthopedic endophytes. Consequently, the manipulation of immune cells to co-inhibit or co-activate signaling represents a crucial strategy for the management of biofilm. This study reports the incorporation of Mn2+ into mesoporous dopamine nanoparticles (Mnp) containing the stimulator of interferon genes (STING) pathway activator cGAMP (Mncp), and outer wrapping by M1-like macrophage cell membrane (m-Mncp). The cell membrane enhances the material's targeting ability for biofilm, allowing it to accumulate locally at the infectious focus. Furthermore, m-Mncp mechanically disrupts the biofilm through photothermal therapy and induces antigen exposure through photodynamic therapy-generated reactive oxygen species (ROS). Importantly, the modulation of immunosuppression and immune activation results in the augmentation of antigen-presenting cells (APCs) and the commencement of antigen presentation, thereby inducing biofilm-specific humoral immunity and memory responses. Additionally, this approach effectively suppresses the activation of myeloid-derived suppressor cells (MDSCs) while simultaneously boosting the activity of T cells. Our study showcases the efficacy of utilizing m-Mncp immunotherapy in conjunction with photothermal and photodynamic therapy to effectively mitigate residual and recurrent infections following the extraction of infected implants. As such, this research presents a viable alternative to traditional antibiotic treatments for biofilm that are challenging to manage.
Assuntos
Biofilmes , Indóis , Proteínas de Membrana , Polímeros , Biofilmes/efeitos dos fármacos , Polímeros/química , Animais , Indóis/química , Indóis/farmacologia , Camundongos , Proteínas de Membrana/metabolismo , Nanopartículas/química , Fotoquimioterapia/métodos , Porosidade , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Feminino , Transdução de Sinais/efeitos dos fármacos , Terapia Fototérmica , Células Supressoras Mieloides/metabolismo , Células Supressoras Mieloides/efeitos dos fármacos , Camundongos Endogâmicos C57BLRESUMO
Biofilm formation of Staphylococcus aureus in food processing environments raises significant safety concerns, necessitating the development of new antibiofilm approaches for controlling S. aureus contamination. This study aimed to elucidate the antibiofilm mechanism of 2R,3R-dihydromyricetin (DMY), a natural flavonoid, against S. aureus and evaluate its efficacy in reducing bacterial adhesion to eggshell. The results revealed that DMY was a potent inhibitor of S. aureus sortase A (SrtA) with an IC50 of 73.43 µM, preventing bacterial adhesion to fibrinogen and subsequent biofilm formation. Fluorescence quenching assay and surface plasmon resonance analysis confirmed that DMY could directly bind to S. aureus SrtA. Notably, circular dichroism spectra demonstrated a conformational change in SrtA from α-helical to ß-sheet structure upon DMY binding. Molecular dynamics simulation suggested that DMY bound to the catalytic pocket of S. aureus SrtA via hydrophobic interactions and hydrogen bonds. Furthermore, fluorescence microscopic observations further revealed that DMY attenuated the biofilm-related phenotype of SrtA by decreasing the anchoring of S. aureus protein A (SpA) onto cell wall. Importantly, pretreatment with 125 µg/mL DMY significantly reduced 1.14-1.75 log CFU/cm2 of S. aureus adhered on eggshells. Overall, these findings highlight how specific targeting of SrtA by DMY inhibits the attachment stages of biofilm development in S. aureus, making it a promising candidate for a novel disinfectant against this pathogen in the food industry.
Assuntos
Aminoaciltransferases , Aderência Bacteriana , Proteínas de Bactérias , Biofilmes , Cisteína Endopeptidases , Casca de Ovo , Flavonóis , Staphylococcus aureus , Staphylococcus aureus/efeitos dos fármacos , Cisteína Endopeptidases/metabolismo , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Aminoaciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Flavonóis/farmacologia , Animais , Aderência Bacteriana/efeitos dos fármacos , Casca de Ovo/microbiologia , Antibacterianos/farmacologia , Simulação de Dinâmica Molecular , GalinhasRESUMO
Bacteria-derived H2S plays multifunctional protective roles against antibiotics insult, and the H2S biogenesis pathway is emerging as a viable target for the antibacterial adjuvant design. However, the development of a pan-inhibitor against H2S-synthesizing enzymes is challenging and underdeveloped. Herein, we propose an alternative strategy to downregulate the H2S levels in H2S-producing bacteria, which depletes the bacteria-derived H2S chemically by H2S scavengers without acting on the synthesizing enzymes. After the screening of chemically diversified scaffolds and a structural optimization campaign, a potent and specific H2S scavenger is successfully identified, which displays efficient H2S depletion in several H2S-producing bacteria, potentiates both bactericidal agents and photodynamic therapy, enhances the bacterial clearance of macrophages and polymorphonuclear neutrophils, disrupts the formation of bacterial biofilm and increases the sensitivity of bacterial persister cells to antibiotics. Most importantly, such an H2S scavenger exhibits sensitizing effects with gentamicin in Pseudomonas aeruginosa -infected pneumonia and skin wound female mouse models. In aggregate, our results not only provide an effective strategy to deplete bacteria-derived H2S and establish the H2S biogenesis pathway as a viable target for persisters and drug-resistant bacteria, but also deliver a promising antibacterial adjuvant for potential clinical translation.
Assuntos
Antibacterianos , Sulfeto de Hidrogênio , Pseudomonas aeruginosa , Animais , Antibacterianos/farmacologia , Sulfeto de Hidrogênio/metabolismo , Sulfeto de Hidrogênio/farmacologia , Camundongos , Pseudomonas aeruginosa/efeitos dos fármacos , Feminino , Biofilmes/efeitos dos fármacos , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/imunologia , Humanos , Gentamicinas/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/metabolismo , Bactérias/efeitos dos fármacos , Células RAW 264.7 , Testes de Sensibilidade Microbiana , Modelos Animais de DoençasRESUMO
We present a patient with a post-pneumonectomy empyema refractory to surgical debridement and systemic antibiotics. The patient initially presented with a bronchopleural fistula and pneumothorax secondary to tuberculosis (TB) destroyed lung, which required a pneumonectomy with Eloesser flap. Ongoing pleural infection delayed the closure of the Eloesser flap, and thoracoscopic inspection of his chest cavity revealed a green, mucous biofilm-like structure lining the postpneumonectomy pleural cavity. Cultures identified pan-susceptible Pseudomonas aeruginosa. Despite debriding this biofilm-like structure and administering systemic antibiotics, the patient continued to show persistent signs of infection and regrowth of the film. We employed a novel approach to dissolve the biofilm-like structure using intrapleural dornase alfa followed by intrapleural antibiotic washes. After 3 weeks of daily washes, repeat inspection demonstrated the biofilm-like structure had completely resolved. Resolving the pseudomonas biofilm-like structure allowed permanent closure of his chest without further need for systemic antibiotics. At follow up 3 months later, he showed no sequalae. This treatment option can be an important adjunct to improve likelihood of chest closure in patients with post-pneumonectomy empyema that resists standard treatment options due to biofilm formation.
Assuntos
Antibacterianos , Biofilmes , Pneumonectomia , Infecções por Pseudomonas , Pseudomonas aeruginosa , Humanos , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Masculino , Pseudomonas aeruginosa/efeitos dos fármacos , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Antibacterianos/uso terapêutico , Antibacterianos/farmacologia , Empiema Pleural/microbiologia , Empiema Pleural/tratamento farmacológico , Empiema Pleural/cirurgia , Empiema Pleural/etiologia , Pessoa de Meia-Idade , Resultado do Tratamento , DesbridamentoRESUMO
Introduction. Peri-implantitis is a plaque-associated disease that leads to implant loss and arises from bacterial biofilms on the surface of the implant. Smoking is a risk factor for peri-implantitis and impedes treatment effectiveness. Additionally, aryl hydrocarbon receptor (AHR), IL-6, and IL-22 levels are related to peri-implantitis.Aim. We aimed to investigate the effects of nicotine on inflammatory response, bacterial growth and biofilm formation.Hypothesis/Gap Statement. We hypothesized that nicotine promoted pathogenic bacterial growth and biofilm formation, thereby aggravating inflammation.Methodology. The expression of AHR, IL-6 and IL-22 was measured in peri-implant sulci fluid using quantitative PCR and Western blot analyses. The cementum was incubated with bacterial suspension including Porphyromonas gingivalis, Streptococcus sanguinis and Fusobacterium nucleatum and treated with 100, 200, 250 and 300 µg ml-1 nicotine, and then, the absorbance and number of colony-forming units were detected. Biofilm formation was evaluated using the tissue culture plate method and safranin O staining. Carbohydrates and proteins were measured by the phenol-sulfuric acid method and the bicinchoninic acid method, respectively.Results. The results indicated that smoking increased the levels of AHR, IL-6 and IL-22. Functionally, nicotine promoted the growth of P. gingivalis, S. sanguinis and F. nucleatum. Additionally, it promoted the biofilm formation of these bacteria and increased the contents of carbohydrates and proteins.Conclusion. Nicotine promoted bacterial growth and biofilm build-up, suggesting that smoking may aggravate the progression of peri-implantitis.
Assuntos
Biofilmes , Nicotina , Peri-Implantite , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Nicotina/farmacologia , Humanos , Peri-Implantite/microbiologia , Fusobacterium nucleatum/efeitos dos fármacos , Fusobacterium nucleatum/crescimento & desenvolvimento , Fusobacterium nucleatum/fisiologia , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/crescimento & desenvolvimento , Masculino , Implantes Dentários/microbiologia , Feminino , Interleucina-6/metabolismo , Pessoa de Meia-Idade , Interleucinas/metabolismo , Streptococcus sanguis/efeitos dos fármacos , Streptococcus sanguis/crescimento & desenvolvimento , Bactérias/efeitos dos fármacos , Bactérias/classificação , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Bactérias/isolamento & purificação , Fumar/efeitos adversosRESUMO
Starmerella bombicola is a native yeast strain producing sophorolipids as secondary metabolites. This study explores the production, characterization, and biological activities of sophorolipids and investigates the antimicrobial, anti-biofilm, and antifungal properties of sophorolipids produced from oil refinery wastes by the yeast Starmerella bombicola. The present work demonstrated that S. bombicola MTCC 1910 when grown in oil refinery wastes namely palm fatty acid distillates and soy fatty acid distillates enhanced the rate of sophorolipids production drastically in comparison to vegetable oil, sunflower oil used as hydrophobic feedstock. Sophorolipid yields were 18.14, 37.21, and 46.1 g/L with sunflower oil, palm, and soy fatty acid distillates respectively. The crude biosurfactants were characterized using TLC, FTIR, and HPLC revealing to be acetylated sophorolipids containing both the acidic and lactonic isomeric forms. The surface lowering and emulsifying properties of the sophorolipids from refinery wastes were significantly higher than the sunflower oil-derived sophorolipids. Also, all the sophorolipids exhibited strong antibacterial properties (minimum inhibitory concentrations were between 50 and 200 µg mL-1) against Salmonella typhimurium, Bacillus cereus, and Staphylococcus epidermidis and were validated with morphological analysis by Scanning electron microscopy. All the sophorolipids were potent biofilm inhibitors and eradicators (minimum biofilm inhibitory and eradication concentrations were between 12.5 to 1000 µg mL-1) for all the tested organisms. Furthermore, antifungal activities were also found to exhibit about 16-56% inhibition at 1 mg mL-1 for fungal mycelial growth. Therefore, this endeavour of sophorolipids production using palm and soy fatty acid distillates not only opens up a window for the bioconversion of industrial wastes into productive biosurfactants but also concludes that sophorolipids from oil refinery wastes are potent antimicrobial, anti-biofilm, and antifungal agents, highlighting their potential in biotechnological and medical applications.
Assuntos
Antibacterianos , Antifúngicos , Biofilmes , Ácidos Graxos , Testes de Sensibilidade Microbiana , Óleos de Plantas , Biofilmes/efeitos dos fármacos , Antifúngicos/farmacologia , Antifúngicos/metabolismo , Antibacterianos/farmacologia , Antibacterianos/biossíntese , Óleos de Plantas/farmacologia , Óleos de Plantas/metabolismo , Óleos de Plantas/química , Bactérias/efeitos dos fármacos , Tensoativos/farmacologia , Tensoativos/metabolismo , Óleo de Palmeira/química , Óleo de Palmeira/farmacologia , Óleo de Girassol/química , Saccharomycetales/metabolismo , Fungos/efeitos dos fármacos , Óleo de Soja/metabolismo , Óleo de Soja/farmacologia , Ácidos OleicosRESUMO
Candida albicans (C. albicans) is a main cause of hospital-acquired fungal infections. Combination therapy is promising as a novel anti-C. albicans strategy because of its better efficacy. Theasaponins are pentacyclic triterpenes in the Camellia genus with multiple biological activities. Our previous studies prove that theasaponins display inhibitory activity against C. albicans. Ascorbic acid (VC) is a vitamin found in many plants that shows potential in combination therapy. However, whether VC enhances the activity of theasaponins remains unclear. In this study, the checkerboard micro-dilution method was used to assess the effect of VC (0-80 mmol/L) on the anti-C. albicans effect of theasaponins (0-1000 µg/mL). Then, the effects of theasaponins (31.25 µg/mL), VC (80 mmol/L), and theasaponins (31.25 µg/mL) + VC (80 mmol/L) on C. albicans planktonic cells and different stages of biofilm formation were assessed. Transcriptomic analysis was conducted to investigate the molecular mechanisms. According to the results, VC enhanced the anti-planktonic and anti-biofilm effect of theasaponins against C. albicans. The minimum inhibitory concentration of theasaponins was significantly decreased and the fungicidal efficiency was increased with the addition of VC. VC remarkably aggravated the suppression of theasaponins with regard to various virulence factors of C. albicans, including adhesion, early biofilm formation, mature biofilm, cell surface hydrophobicity, and phospholipase activity. Compared with the theasaponins or VC groups, the level of intracellular reactive oxygen species was higher, while the levels of mitochondrial membrane potential and adenosine triphosphate were lower in the combination group, suggesting more severe oxidative stress, mitochondrial injury, and energy deficiency. Transcriptomic analysis revealed that the combination predominantly suppressed the pathways of glycolysis, glycerophospholipid metabolism, glutathione metabolism, and cysteine and methionine metabolism. This implied that energy deficiency and redox imbalance were associated with the anti-C. albicans activity of the combination. These results prove that VC enhances the inhibitory effect of theasaponins against C. albicans and that the combination has the potential to be used as a topical antifungal therapy or disinfectant.
Assuntos
Antifúngicos , Ácido Ascórbico , Biofilmes , Candida albicans , Testes de Sensibilidade Microbiana , Saponinas , Candida albicans/efeitos dos fármacos , Ácido Ascórbico/farmacologia , Antifúngicos/farmacologia , Biofilmes/efeitos dos fármacos , Saponinas/farmacologia , Sinergismo FarmacológicoRESUMO
Quorum sensing (QS) is a bacterial cell-to-cell communication mechanism that plays an essential role in bacterial pathogenesis. QS governs bacterial behavior and controls biofilm formation, which in turn contributes to antibiotic resistance. Therefore, identifying and synthesizing novel compounds to overcome QS and inhibit biofilm formation are essential. Coumarins are important plant-derived natural products with wide-ranging bioactivities and extensive applications, including antibacterial, antifungal, anticoagulant, antioxidant, anticancer, and anti-inflammatory properties. Additionally, coumarins are capable of QS rewiring and biofilm formation inhibition, leading to higher susceptibility to antimicrobial agents and less antibiotic resistance. Therefore, in this review, we aim to provide an overview of QS and biofilm formation. This review also discusses the role of natural and synthesized coumarins in controlling QS, inhibiting biofilm formation, and inducing synergy in antibiotic-coumarin combinations. Hence, this review emphasizes the potential of coumarin compounds to act as antibacterial agents and demonstrates their ability to alleviate antibiotic resistance.
Assuntos
Antibacterianos , Biofilmes , Cumarínicos , Percepção de Quorum , Cumarínicos/farmacologia , Cumarínicos/química , Percepção de Quorum/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/química , Humanos , Bactérias/efeitos dos fármacosRESUMO
One of the major global health threats in the present era is antibiotic resistance. Biosynthesized iron oxide nanoparticles (FeNPs) can combat microbial infections and can be synthesized without harmful chemicals. In the present investigation, 16S rRNA gene sequencing was used to discover Streptomyces sp. SMGL39, an actinomycete isolate utilized to reduce ferrous sulfate heptahydrate (FeSO4.7H2O) to biosynthesize FeNPs, which were then characterized using UV-Vis, XRD, FTIR, and TEM analyses. Furthermore, in our current study, the biosynthesized FeNPs were tested for antimicrobial and antibiofilm characteristics against different Gram-negative, Gram-positive, and fungal strains. Additionally, our work examines the biosynthesized FeNPs' molecular docking and binding affinity to key enzymes, which contributed to bacterial infection cooperation via quorum sensing (QS) processes. A bright yellow to dark brown color shift indicated the production of FeNPs, which have polydispersed forms with particle sizes ranging from 80 to 180 nm and UV absorbance ranging from 220 to 280 nm. Biosynthesized FeNPs from actinobacteria significantly reduced the microbial growth of Fusarium oxysporum and L. monocytogenes, while they showed weak antimicrobial activity against P. aeruginosa and no activity against E. coli, MRSA, or Aspergillus niger. On the other hand, biosynthesized FeNPs showed strong antibiofilm activity against P. aeruginosa while showing mild and weak activity against B. subtilis and E. coli, respectively. The collaboration of biosynthesized FeNPs and key enzymes for bacterial infection exhibits hydrophobic and/or hydrogen bonding, according to this research. These results show that actinobacteria-biosynthesized FeNPs prevent biofilm development in bacteria.
Assuntos
Biofilmes , Nanopartículas Magnéticas de Óxido de Ferro , Testes de Sensibilidade Microbiana , Streptomyces , Streptomyces/metabolismo , Streptomyces/química , Biofilmes/efeitos dos fármacos , Nanopartículas Magnéticas de Óxido de Ferro/química , Simulação de Acoplamento Molecular , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/biossíntese , Fusarium/efeitos dos fármacos , Simulação por ComputadorRESUMO
Ventilator-associated pneumonia (VAP) remains one of the most common hospital-acquired infections (HAI). Considering the complicated diagnosis and the lack of effective treatment, prophylactic measures are suggested as the new standard to prevent the disease. Although VAP often manifests a polymicrobial nature, Pseudomonas aeruginosa remains one of the pathogens associated with the highest morbidity and mortality rates within these mechanically ventilated patients. In this paper, we report on the development of an antibacterial hydrogel coating using the polymyxin B (PMB) peptide to prevent bacterial adhesion to the polymeric substrate. We fully characterized the properties of the coating using atomic force microscopy (AFM), scanning electron microscopy (SEM), wettability analyses and Fourier-transform infrared (FTIR) and Raman spectroscopy. Furthermore, several biological assays confirmed the antibacterial and anti-biofilm effect of the tubing for at least 8 days against P. aeruginosa. On top of that, the produced coating is compliant with the requirements regarding cytocompatibility stated in the ISO (International Organization for Standardization) 10993 guidelines and an extended release of PMB over a period of at least 42 days was detected. In conclusion, this study serves as a foundation for peptide-releasing hydrogel formulas in the prevention of VAP.
Assuntos
Antibacterianos , Biofilmes , Hidrogéis , Pneumonia Associada à Ventilação Mecânica , Polimixina B , Pseudomonas aeruginosa , Pneumonia Associada à Ventilação Mecânica/prevenção & controle , Polimixina B/farmacologia , Polimixina B/química , Pseudomonas aeruginosa/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/química , Humanos , Hidrogéis/química , Biofilmes/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Aderência Bacteriana/efeitos dos fármacos , Peptídeos/química , Peptídeos/farmacologiaRESUMO
Multi-drug resistance is recognized as a significant worldwide public health concern in the current century. Biofilm formation further exacerbates bacterial resistance to antibacterial medications, host immunological responses, and phagocytosis, resulting in long-lasting chronic illnesses. Investigating natural resources is a very potent approach for developing alternative anti-infective medications to effectively control multi-drug resistant bacterial infections. In this study, a unique mushroom species namely Pleurotus platypus had been discovered from the Terai-Duars region of West Bengal, India. The myco-chemical profiling and preliminary chemical analysis of Pleurotus platypus methanolic extract determined the significant presence of metabolites belonging to several major chemical classes such as flavonoid, alkaloid, triterpenoid, polyphenol, benzoic acids, coumarin, flavone etc. Most intriguingly, the extract possessed effective antibacterial, antibiofilm and antivirulence properties against Staphylococcus aureus and Methicillin resistant Staphylococcus aureus, one of the most notable drug-resistant opportunistic and nosocomial pathogens. Mechanistically, the mushroom extract enhanced the production of Reactive Oxygen Species (ROS) inside the targeted bacteria, causing alterations in membrane potential, damage to the cellular membrane and further release of intracellular DNA, destined to cell death. Moreover, the methanolic extract reported the eradication of pre-existing biofilms from the urinary catheter surface, hinting towards its future application in the related field. To summarize, Pleurotus platypus methanolic extract could be an excellent alternative antibacterial and antibiofilm therapeutic candidate for the effective management of Staphylococcus infections with improved outcome.
Assuntos
Antibacterianos , Biofilmes , Membrana Celular , Testes de Sensibilidade Microbiana , Pleurotus , Espécies Reativas de Oxigênio , Staphylococcus aureus , Biofilmes/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Pleurotus/química , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Índia , Metanol/química , Virulência/efeitos dos fármacos , Animais , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologiaRESUMO
BACKGROUND: Considering that antimicrobial resistance among oral pathogens is a significant concern in dental practice, with broader implications for overall health due to the oral microbiota serving as a reservoir for antibiotic resistance genes (ARGs), research into natural products is crucial for addressing this issue. OBJECTIVE: This study aimed to evaluate tea tree oil (TTO) and chitosan (CH) performance against oral pathogens, including mixed-species biofilm, and its effects on bacteria growth, in addition to chemical characterization and cytotoxicity of TTO. METHODS: Tea Tree Oil and low molecular weight chitosan were used in this study. The chemical composition of TTO was analyzed using gas chromatography coupled with mass spectrometry (GC-MS). To evaluate TTO's antimicrobial properties, time-kill and cell viability assays were conducted. Additionally, minimum inhibitory concentration (MIC), minimum microbiocidal concentration (MMC), checkerboard, and biofilm assays were performed using TTO and CH alone and in combination. RESULTS: TTO chromatography peaks found consistent with the standard ISO4730:2017 and literature. TTO and CH exhibited inhibitory activity against all tested microorganisms. The predominantly microbiostatic activity of TTO is probably related to terpinen-4-ol associated with terpinene. The oil at MIC value was able to delay the log phase of Aggregatibacter actinomycetemcomitans growth. Fibroblasts (L929) viability remained above 70 % during 24 h for TTO concentrations ranging from 0.5 to 0.0625 mg/ml. TTO-CH combination showed a synergistic activity (FIC = 0.5) against A. actinomycetemcomitans and Streptococcus sanguinis, at a concentration of 0,25MIC for both species. The compounds at MIC concentration inhibited both monospecies and mixed-species biofilms studied bacteria to the same extent as the azithromycin control. CONCLUSION: TTO and CH demonstrated efficacy in combating oral pathogens and TTO-CH combination offers a promising approach to confront microbial resistance in the oral environment.
Assuntos
Antibacterianos , Biofilmes , Quitosana , Testes de Sensibilidade Microbiana , Óleo de Melaleuca , Óleo de Melaleuca/farmacologia , Óleo de Melaleuca/química , Quitosana/farmacologia , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Antibacterianos/farmacologia , Cromatografia Gasosa-Espectrometria de Massas , Boca/microbiologia , Humanos , Terpenos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Aggregatibacter actinomycetemcomitans/efeitos dos fármacos , Bactérias/efeitos dos fármacos , Farmacorresistência Bacteriana , Óleos Voláteis/farmacologia , Óleos Voláteis/química , Anti-Infecciosos/farmacologiaRESUMO
It is thought to be risk-free, environmentally benign, and safe for biological processes to produce zinc oxide nanoparticles from renewable resources. This study examined Cassia javanica's ability to create ZnONPs. The generated ZnONPs were analyzed using a variety of techniques, such as TEM, FTIR spectroscopy, UV-Vis spectroscopy, and XRD analysis. The antibacterial potential of ZnONPs has been investigated using both Agar well diffusion and microtitreplate (MTP) methods. One method used to evaluate ZnONPs' capacity to scavenge free radicals at different concentrations was the DPPH method. The permanent zinc oxide (ZnO) shape and the naturally occurring crystal structure of ZnONPs were validated by the XRD data. ZnONPs showed antibacterial activity with MICs of 31.7 µg/mL toward Bacillus subtilis, 62.5 µg/mL for Salmonella typhimurium, Escherichia coli while Clostridium sporogenes and Bacillus pumilus was 125µg/mL. Furthermore, ZnONPs demonstrated a range of antibiofilm activities toward Staphylococcus aureus (MRSA). ZnONPs showed an intriguing antioxidant capacity, achieving IC50 of 109.3 µg/ml µg/mL. Additionally, ZnONPs demonstrated low toxic effect on Vero cell with IC50 154.01 µg/mL as well as possible anticancer action when applied to the carcinoma cell lines HepG2 with IC50 of 47.48 µg/mL. Furthermore, ZnONPs at 62.5 µg/mL had a promising antiviral impact against HSV1 and COX B4, with antiviral activities of 75.4% and 65.8%, respectively.
Assuntos
Antibacterianos , Antineoplásicos , Antioxidantes , Antivirais , Biofilmes , Cassia , Testes de Sensibilidade Microbiana , Óxido de Zinco , Óxido de Zinco/farmacologia , Óxido de Zinco/química , Antibacterianos/farmacologia , Antibacterianos/química , Humanos , Biofilmes/efeitos dos fármacos , Antioxidantes/farmacologia , Antioxidantes/química , Antivirais/farmacologia , Antivirais/química , Animais , Cassia/química , Antineoplásicos/farmacologia , Antineoplásicos/química , Chlorocebus aethiops , Células Vero , Nanopartículas Metálicas/química , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Nanopartículas/químicaRESUMO
Periodontitis is a chronic inflammatory disease involving plaque biofilm as a pathogenic factor. Potassium ion plays an important role in cellular homeostasis; a large outflow of potassium may lead to local inflammation progression. In this work, the multifunctional short peptide molecule BmKTX-33 was designed by modifying the BmKTX, a Kv1.3 potassium channel inhibitor. This was to explore its antibacterial properties, capability of maintaining cell ion homeostasis, and bone-forming capacity. The results showed that BmKTX-33 had inhibitory effects on S. gordonii, F. nucleatum, and P. gingivalis. Moreover, BmKTX-33 also inhibited excessive potassium outflow in inflammatory environments. Finally, BmKTX-33 promoted MC3T3-E1 early osteogenesis while suppressing the NLRP3 inflammasome's production. In conclusion, BmKTX-33 not only has antibacterial properties, but also can inhibit the expression of NLRP3 inflammasome and play an anti-inflammatory role.
Assuntos
Proteína 3 que Contém Domínio de Pirina da Família NLR , Periodontite , Animais , Periodontite/tratamento farmacológico , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/uso terapêutico , Peptídeos/farmacologia , Peptídeos/química , Peptídeos/uso terapêutico , Porphyromonas gingivalis/efeitos dos fármacos , Potássio/metabolismo , Linhagem Celular , Osteogênese/efeitos dos fármacos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Anti-Inflamatórios/uso terapêutico , Biofilmes/efeitos dos fármacosRESUMO
A range of bacteria biofilm models exist for the testing of antibiotics. However, many of these are limited to a single experimental output, such as colony-forming units or metabolic activity. Furthermore, many biofilm models do not reflect the biological and physiochemical properties of the human host environment. This is an important issue in many conditions, but most noticeably in cystic fibrosis (CF). A large proportion of people with CF suffer from both chronic and intermittent infections, and in vitro, antibiotic susceptibility testing poorly correlates with patient treatment outcomes. Some biofilm models incorporate CF lung-relevant media, including synthetic sputum mimics, but do not consider the polymicrobial nature of the environment, which alters biofilm architecture, physiology, and the way microbes respond to treatment. The solid-air interface colony biofilm model described here is highly adaptable and incorporates both CF-relevant media and a polymicrobial context. This model can also be used for mid-throughput screening of antimicrobials and to study their effect on polymicrobial dynamics. Output measurements from the model can be colony-forming units, metabolic activity, and confocal microscopy analysis. The model can easily be adapted to different microorganisms, media, temperatures, and variable oxygen conditions and can be used to test a wide range of chemical, biological, and physical treatments.
Assuntos
Biofilmes , Fibrose Cística , Fibrose Cística/microbiologia , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Humanos , Antibacterianos/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/fisiologia , Modelos Biológicos , Testes de Sensibilidade Microbiana/métodosRESUMO
OBJECTIVES: Photodynamic inactivation (PDI) is a powerful technique for eradicating microorganisms, and our group previously demonstrated its effectiveness against planktonic cultures of Staphylococcus aureus bacteria using 5,10,15,20-tetrakis[4-(3-N,N-dimethylaminopropoxy)phenyl]porphyrin (TAPP) and visible light irradiation. However, biofilms exhibit a lower sensitivity to PDI, mainly due to limited penetration of the photosensitizer (PS). In the context of emerging antibacterial strategies, near-infrared treatments (NIRTs) have shown promise, especially for combating resistant strains. NIRT can act either through photon absorption by water, causing a thermal effect on bacteria, or by specific chromophores without a significant temperature increase. Our objective was to enhance biofilm sensitivity to TAPP-PDI by pretreatment with NIRT. This combined approach aims to disrupt biofilms and increase the efficacy of TAPP-PDI against bacterial biofilms. MATERIALS AND METHODS: In vitro biofilm models of S. aureus RN6390 were utilized. NIRTs involved a 980 nm laser (continuous mode, 7.5 W/cm2, 30 s, totaling 225 J/cm2) post-TAPP exposure to enhance photosensitizer accumulation. Subsequent visible light irradiation at 180 J/cm2 was employed to perform PDI. Colony-forming unit counts evaluated the synergistic effect on bacterial viability. Scanning electron microscopy visualized the architectural changes in the biofilm structure. TAPP was extracted from bacteria to estimate the impact of NIRT on biofilm penetration. RESULTS: Using in vitro biofilm models, NIRT application following biofilm exposure to TAPP increased PS accumulation per bacteria. Under these conditions, NIRT induced a transient increase in the temperature of PBS to 46.0 ± 2.6°C (ΔT = 21.5°C). Following exposure to visible light, a synergistic effect emerged, yielding a substantial 4.4 ± 0.1-log CFU reduction. In contrast, the PDI and NIRT treatments individually caused a decrease in viability of 0.9 ± 0.1 and 0.8 ± 0.2-log respectively. Interestingly, preheating TAPP-PBS to 46°C had no significant impact on TAPP-PDI efficacy, suggesting the involvement of thermal and nonthermal effects of NIR action. In addition to the enhanced TAPP penetration, NIRT dispersed the biofilms and induced clefts in the biofilm matrix. CONCLUSION: Our findings suggest that NIR irradiation serves as a complementary treatment to PDI. This combined strategy reduces bacterial numbers at lower PS concentrations than standalone PDI treatment, highlighting its potential as an effective and resource-efficient antibacterial approach.
Assuntos
Biofilmes , Fotoquimioterapia , Fármacos Fotossensibilizantes , Staphylococcus aureus , Biofilmes/efeitos dos fármacos , Biofilmes/efeitos da radiação , Staphylococcus aureus/efeitos dos fármacos , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Raios Infravermelhos , Porfirinas/farmacologiaRESUMO
BACKGROUND: Inorganic polyphosphate (polyP)-targeted polyphosphate kinase 1 (PPK1) has attracted much attention by virtue of its importance in bacterial pathogenicity and persistence, as well as its exclusive presence in microorganisms. However, only very few drugs have been found to be efficacious in inhibiting the Acinetobacter baumannii (A. baumannii) PPK1 protein. RESULTS: In this study, we identified Scutellarein (Scu), a potent PPK1 inhibitor that could significantly influence PPK1-regulated motility, biofilm formation, and bacterial persistence, which was further validated by the results of transcriptome analysis. Mechanistic explorations revealed that Scu achieved its enzyme inhibitory activity predominantly through direct engagement with the active center of PPK1. Moreover, the survival rate of Galleria mellonella larvae was increased by about 35% with 20 mg/kg of Scu treatment. The remarkable therapeutic benefits of Scu were also observed in the mouse pneumonia model, shown mainly by reduced bacterial colonization, pathological lesions, and inflammatory factors. CONCLUSION: Our results revealed that Scu could attenuate the pathogenicity and persistence of A. baumannii by interfering with its important kinase PPK1.
Assuntos
Infecções por Acinetobacter , Acinetobacter baumannii , Fosfotransferases (Aceptor do Grupo Fosfato) , Acinetobacter baumannii/efeitos dos fármacos , Animais , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo , Fosfotransferases (Aceptor do Grupo Fosfato)/antagonistas & inibidores , Camundongos , Infecções por Acinetobacter/tratamento farmacológico , Infecções por Acinetobacter/microbiologia , Biofilmes/efeitos dos fármacos , Antibacterianos/farmacologia , Mariposas/microbiologia , Feminino , Modelos Animais de DoençasRESUMO
This study aimed to identify the inhibitory effect of Agastache rugosa essential oil (AREO) on the cariogenic properties of Streptococcus mutans, which causes dental caries and dental plaque formation. After extracting the AREO, their effects on the growth and acid production of S. mutans were examined. Furthermore, S. mutans biofilm formation was observed on the resin teeth surface. The effect on the expression of biofilm-related genes of S. mutans was measured using real-time PCR. AREO components were analyzed using gas chromatography (GC) and GC-mass spectrometry (MS). The growth and acid production of S. mutans were significantly inhibited at concentrations of 0.02 mg/mL or higher of AREO. At 0.04 mg/mL, inhibition was similar to that of the positive control, 0.1% NaF. AREO suppressed the expression of virulence factors such as gtfB, gtfC, gtfD, gbpB, SpaP, brpA, relA, and vicR at concentrations of 0.02 mg/mL or higher. As a result of GC and GC-MS analyses, the main components of AREO included estragole, limonene, and ß-caryophyllene. These results suggest that A. rugosa may be a useful agent for inhibiting the cariogenic properties of S. mutans.
Assuntos
Agastache , Biofilmes , Óleos Voláteis , Streptococcus mutans , Biofilmes/efeitos dos fármacos , Óleos Voláteis/farmacologia , Óleos Voláteis/química , Streptococcus mutans/efeitos dos fármacos , Agastache/química , Cromatografia Gasosa-Espectrometria de Massas , Cárie Dentária/microbiologia , Cárie Dentária/prevenção & controle , Cárie Dentária/tratamento farmacológico , Derivados de Alilbenzenos/farmacologia , Anisóis/farmacologia , Anisóis/química , Fatores de Virulência , Antibacterianos/farmacologia , Antibacterianos/química , Humanos , Sesquiterpenos/farmacologia , Sesquiterpenos/química , Testes de Sensibilidade Microbiana , Limoneno/farmacologia , Limoneno/química , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacosRESUMO
BACKGROUND: Infections following shoulder surgery, particularly periprosthetic joint infection (PJI), are challenging to treat. Cutibacterium acnes is the causative pathogen in 39% to 76% of these cases. This study explores the efficacy of bacteriophage therapy as an alternative to conventional antibiotics for treating such infections. METHODS: Nine phages with lytic activity were isolated from the skin of humans using C. acnes ATCC 6919 as the indicator host. These phages were tested individually or in combination to assess host range and antibiofilm activity against clinical strains of C. acnes associated with PJIs. The phage cocktail was optimized for broad-spectrum activity and tested in vitro against biofilms formed on titanium discs to mimic the prosthetic environment. RESULTS: The isolated phages displayed lytic activity against a range of C. acnes clinical isolates. The phage cocktail significantly reduced the bacterial load of C. acnes strains 183, 184, and GG2A, as compared with untreated controls (p < 0.05). Individual phages, particularly CaJIE7 and CaJIE3, also demonstrated significant reductions in bacterial load with respect to specific strains. Moreover, phages notably disrupted the biofilm structure and reduced biofilm biomass, confirming the potential of phage therapy in targeting biofilm-associated infections. CONCLUSIONS: Our preclinical findings support the potential of phage therapy as a viable adjunct to traditional antibiotics for treating C. acnes infections in orthopedic device-related infections. The ability of phages to disrupt biofilms may be particularly beneficial for managing infections associated with prosthetic implants.
Assuntos
Bacteriófagos , Biofilmes , Infecções Relacionadas à Prótese , Biofilmes/efeitos dos fármacos , Humanos , Bacteriófagos/fisiologia , Bacteriófagos/isolamento & purificação , Infecções Relacionadas à Prótese/microbiologia , Infecções Relacionadas à Prótese/terapia , Terapia por Fagos/métodos , Infecções por Bactérias Gram-Positivas/terapia , Infecções por Bactérias Gram-Positivas/microbiologia , Especificidade de Hospedeiro , Propionibacteriaceae/fisiologia , Carga BacterianaRESUMO
Biofilms reduce antibiotic efficacy and lead to complications and mortality in human and equine patients with orthopedic infections. Equine bone marrow-derived mesenchymal stromal cells (MSC) kill planktonic bacteria and prevent biofilm formation, but their ability to disrupt established orthopedic biofilms is unknown. Our objective was to evaluate the ability of MSC to reduce established S. aureus or E. coli biofilms in vitro. We hypothesized that MSC would reduce biofilm matrix and colony-forming units (CFU) compared to no treatment and that MSC combined with the antibiotic, amikacin sulfate, would reduce these components more than MSC or amikacin alone. MSC were isolated from 5 adult Thoroughbred horses in antibiotic-free medium. 24-hour S. aureus or E. coli biofilms were co-cultured in triplicate for 24 or 48 hours in a transwell plate system: untreated (negative) control, 30 µg/mL amikacin, 1 x 106 passage 3 MSC, and MSC with 30 µg/mL amikacin. Treated biofilms were photographed and biofilm area quantified digitally. Biomass was quantified via crystal violet staining, and CFU quantified following enzymatic digestion. Data were analyzed using mixed model ANOVA with Tukey post-hoc comparisons (p < 0.05). MSC significantly reduced S. aureus biofilms at both timepoints and E. coli biofilm area at 48 hours compared to untreated controls. MSC with amikacin significantly reduced S. aureus biofilms versus amikacin and E. coli biofilms versus MSC at 48 hours. MSC significantly reduced S. aureus biomass at both timepoints and reduced S. aureus CFU at 48 hours versus untreated controls. MSC with amikacin significantly reduced S. aureus biomass versus amikacin at 24 hours and S. aureus and E. coli CFU versus MSC at both timepoints. MSC primarily disrupted the biofilm matrix but performed differently on S. aureus versus E. coli. Evaluation of biofilm-MSC interactions, MSC dose, and treatment time are warranted prior to testing in vivo.