Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 12.844
1.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article En | MEDLINE | ID: mdl-38731983

Acne vulgaris is a prevalent skin disorder affecting many young individuals, marked by keratinization, inflammation, seborrhea, and colonization by Cutibacterium acnes (C. acnes). Ellagitannins, known for their antibacterial and anti-inflammatory properties, have not been widely studied for their anti-acne effects. Chestnut (Castanea sativa Mill., C. sativa), a rich ellagitannin source, including castalagin whose acne-related bioactivity was previously unexplored, was investigated in this study. The research assessed the effect of C. sativa leaf extract and castalagin on human keratinocytes (HaCaT) infected with C. acnes, finding that both inhibited IL-8 and IL-6 release at concentrations below 25 µg/mL. The action mechanism was linked to NF-κB inhibition, without AP-1 involvement. Furthermore, the extract displayed anti-biofilm properties and reduced CK-10 expression, indicating a potential role in mitigating inflammation, bacterial colonization, and keratosis. Castalagin's bioactivity mirrored the extract's effects, notably in IL-8 inhibition, NF-κB inhibition, and biofilm formation at low µM levels. Other polyphenols, such as flavonol glycosides identified via LC-MS, might also contribute to the extract's biological activities. This study is the first to explore ellagitannins' potential in treating acne, offering insights for developing chestnut-based anti-acne treatments pending future in vivo studies.


Acne Vulgaris , Fagaceae , Hydrolyzable Tannins , Plant Extracts , Plant Leaves , Humans , Hydrolyzable Tannins/pharmacology , Fagaceae/chemistry , Acne Vulgaris/microbiology , Acne Vulgaris/drug therapy , Plant Extracts/pharmacology , Plant Extracts/chemistry , Plant Leaves/chemistry , Keratinocytes/drug effects , Keratinocytes/metabolism , NF-kappa B/metabolism , HaCaT Cells , Propionibacterium acnes/drug effects , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Interleukin-8/metabolism
2.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article En | MEDLINE | ID: mdl-38732057

Implant therapy is a common treatment option in dentistry and orthopedics, but its application is often associated with an increased risk of microbial contamination of the implant surfaces that cause bone tissue impairment. This study aims to develop two silver-enriched platelet-rich plasma (PRP) multifunctional scaffolds active at the same time in preventing implant-associated infections and stimulating bone regeneration. Commercial silver lactate (L) and newly synthesized silver deoxycholate:ß-Cyclodextrin (B), were studied in vitro. Initially, the antimicrobial activity of the two silver soluble forms and the PRP enriched with the two silver forms has been studied on microbial planktonic cells. At the same time, the biocompatibility of silver-enriched PRPs has been assessed by an MTT test on human primary osteoblasts (hOBs). Afterwards, an investigation was conducted to evaluate the activity of selected concentrations and forms of silver-enriched PRPs in inhibiting microbial biofilm formation and stimulating hOB differentiation. PRP-L (0.3 µg/mm2) and PRP-B (0.2 µg/mm2) counteract Staphylococcus aureus, Staphylococcus epidermidis and Candida albicans planktonic cell growth and biofilm formation, preserving hOB viability without interfering with their differentiation capability. Overall, the results obtained suggest that L- and B-enriched PRPs represent a promising preventive strategy against biofilm-related implant infections and demonstrate a new silver formulation that, together with increasing fibrin binding protecting silver in truncated cone-shaped cyclic oligosaccharides, achieved comparable inhibitory results on prokaryotic cells at a lower concentration.


Biofilms , Osteoblasts , Platelet-Rich Plasma , Silver , Humans , Biofilms/drug effects , Silver/chemistry , Silver/pharmacology , Osteoblasts/drug effects , Osteoblasts/cytology , Staphylococcus aureus/drug effects , Candida albicans/drug effects , Prosthesis-Related Infections/prevention & control , Prosthesis-Related Infections/microbiology , Staphylococcus epidermidis/drug effects
3.
J Photochem Photobiol B ; 255: 112905, 2024 Jun.
Article En | MEDLINE | ID: mdl-38703452

Bacterial antibiotic resistance is one of the most significant challenges for public health. The increase in bacterial resistance, mainly due to microorganisms harmful to health, and the need to search for alternative treatments to contain infections that cannot be treated by conventional antibiotic therapy has been aroused. An alternative widely studied in recent decades is antimicrobial photodynamic therapy (aPDT), a treatment that can eliminate microorganisms through oxidative stress. Although this therapy has shown satisfactory results in infection control, it is still controversial in the scientific community whether bacteria manage to develop resistance after successive applications of aPDT. Thus, this work provides an overview of the articles that performed successive aPDT applications in models using bacteria published since 2010, focusing on sublethal dose cycles, highlighting the main PSs tested, and addressing the possible mechanisms for developing tolerance or resistance to aPDT, such as efflux pumps, biofilm formation, OxyR and SoxRS systems, catalase and superoxide dismutase enzymes and quorum sensing.


Biofilms , Drug Resistance, Bacterial , Photochemotherapy , Photosensitizing Agents , Drug Resistance, Bacterial/drug effects , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Biofilms/drug effects , Bacteria/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/chemistry , Quorum Sensing/drug effects , Humans , Catalase/metabolism , Oxidative Stress/drug effects
4.
AAPS PharmSciTech ; 25(5): 106, 2024 May 09.
Article En | MEDLINE | ID: mdl-38724834

The primary factor underlying the virulence of Candida albicans is its capacity to form biofilms, which in turn leads to recurrent complications. Over-the-counter antifungal treatments have proven ineffective in eliminating fungal biofilms and the inflammatory cytokines produced during fungal infections. Chitosan nanoparticles offer broad and versatile therapeutic potential as both antifungal agents and carriers for antifungal drugs to combat biofilm-associated Candida infections. In our study, we endeavoured to develop chitosan nanoparticles utilising chitosan and the antifungal crosslinker phytic acid targeting C. albicans. Phytic acid, known for its potent antifungal and anti-inflammatory properties, efficiently crosslinks with chitosan. The nanoparticles were synthesised using the ionic gelation technique and subjected to analyses including Fourier transform infrared spectroscopy, dynamic light scattering, and zeta potential analysis. The synthesised nanoparticles exhibited dimensions with a diameter (Dh) of 103 ± 3.9 nm, polydispersity index (PDI) of 0.33, and zeta potential (ZP) of 37 ± 2.5 mV. These nanoparticles demonstrated an antifungal effect with a minimum inhibitory concentration (MIC) of 140 ± 2.2 µg/mL, maintaining cell viability at approximately 90% of the MIC value and reducing cytokine levels. Additionally, the nanoparticles reduced ergosterol content and exhibited a 62% ± 1.2 reduction in biofilm susceptibility, as supported by colony-forming unit (CFU) and XTT assays-furthermore, treatment with nanoparticles reduced exopolysaccharide production and decreased secretion of aspartyl protease by C. albicans. Our findings suggest that the synthesised nanoparticles effectively combat Candida albicans infections. In vivo studies conducted on a mouse model of vaginal candidiasis confirmed the efficacy of the nanoparticles in combating fungal infections in vivo.


Antifungal Agents , Biofilms , Candida albicans , Chitosan , Microbial Sensitivity Tests , Nanoparticles , Phytic Acid , Chitosan/chemistry , Biofilms/drug effects , Nanoparticles/chemistry , Antifungal Agents/pharmacology , Antifungal Agents/administration & dosage , Animals , Candida albicans/drug effects , Mice , Microbial Sensitivity Tests/methods , Phytic Acid/pharmacology , Phytic Acid/administration & dosage , Phytic Acid/chemistry , Female , Candidiasis/drug therapy , Particle Size , Drug Carriers/chemistry , Cross-Linking Reagents/chemistry , Cytokines/metabolism
5.
PLoS One ; 19(5): e0303039, 2024.
Article En | MEDLINE | ID: mdl-38701045

The complexity of chronic wounds creates difficulty in effective treatments, leading to prolonged care and significant morbidity. Additionally, these wounds are incredibly prone to bacterial biofilm development, further complicating treatment. The current standard treatment of colonized superficial wounds, debridement with intermittent systemic antibiotics, can lead to systemic side-effects and often fails to directly target the bacterial biofilm. Furthermore, standard of care dressings do not directly provide adequate antimicrobial properties. This study aims to assess the capacity of human-derived collagen hydrogel to provide sustained antibiotic release to disrupt bacterial biofilms and decrease bacterial load while maintaining host cell viability and scaffold integrity. Human collagen harvested from flexor tendons underwent processing to yield a gellable liquid, and subsequently was combined with varying concentrations of gentamicin (50-500 mg/L) or clindamycin (10-100 mg/L). The elution kinetics of antibiotics from the hydrogel were analyzed using liquid chromatography-mass spectrometry. The gel was used to topically treat Methicillin-resistant Staphylococcus aureus (MRSA) and Clostridium perfringens in established Kirby-Bauer and Crystal Violet models to assess the efficacy of bacterial inhibition. 2D mammalian cell monolayers were topically treated, and cell death was quantified to assess cytotoxicity. Bacteria-enhanced in vitro scratch assays were treated with antibiotic-embedded hydrogel and imaged over time to assess cell death and mobility. Collagen hydrogel embedded with antibiotics (cHG+abx) demonstrated sustained antibiotic release for up to 48 hours with successful inhibition of both MRSA and C. perfringens biofilms, while remaining bioactive up to 72 hours. Administration of cHG+abx with antibiotic concentrations up to 100X minimum inhibitory concentration was found to be non-toxic and facilitated mammalian cell migration in an in vitro scratch model. Collagen hydrogel is a promising pharmaceutical delivery vehicle that allows for safe, precise bacterial targeting for effective bacterial inhibition in a pro-regenerative scaffold.


Anti-Bacterial Agents , Biofilms , Collagen , Hydrogels , Methicillin-Resistant Staphylococcus aureus , Biofilms/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Humans , Collagen/chemistry , Hydrogels/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Clindamycin/pharmacology , Clindamycin/administration & dosage , Microbial Sensitivity Tests , Administration, Topical , Gentamicins/pharmacology , Gentamicins/administration & dosage
6.
Pathog Dis ; 822024 Feb 07.
Article En | MEDLINE | ID: mdl-38730561

Antibiotic resistance (ATBR) is increasing every year as the overuse of antibiotics (ATBs) and the lack of newly emerging antimicrobial agents lead to an efficient pathogen escape from ATBs action. This trend is alarming and the World Health Organization warned in 2021 that ATBR could become the leading cause of death worldwide by 2050. The development of novel ATBs is not fast enough considering the situation, and alternative strategies are therefore urgently required. One such alternative may be the use of non-thermal plasma (NTP), a well-established antimicrobial agent actively used in a growing number of medical fields. Despite its efficiency, NTP alone is not always sufficient to completely eliminate pathogens. However, NTP combined with ATBs is more potent and evidence has been emerging over the last few years proving this is a robust and highly effective strategy to fight resistant pathogens. This minireview summarizes experimental research addressing the potential of the NTP-ATBs combination, particularly for inhibiting planktonic and biofilm growth and treating infections in mouse models caused by methicillin-resistant Staphylococcus aureus or Pseudomonas aeruginosa. The published studies highlight this combination as a promising solution to emerging ATBR, and further research is therefore highly desirable.


Anti-Bacterial Agents , Biofilms , Plasma Gases , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Plasma Gases/pharmacology , Animals , Humans , Biofilms/drug effects , Pseudomonas aeruginosa/drug effects , Mice , Methicillin-Resistant Staphylococcus aureus/drug effects , Drug Resistance, Bacterial , Drug Resistance, Microbial , Pseudomonas Infections/microbiology , Pseudomonas Infections/drug therapy , Disease Models, Animal , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy
7.
Nat Commun ; 15(1): 4036, 2024 May 13.
Article En | MEDLINE | ID: mdl-38740750

Microbial Ni2+ homeostasis underpins the virulence of several clinical pathogens. Ni2+ is an essential cofactor in urease and [NiFe]-hydrogenases involved in colonization and persistence. Many microbes produce metallophores to sequester metals necessary for their metabolism and starve competing neighboring organisms. The fungal metallophore aspergillomarasmine A (AMA) shows narrow specificity for Zn2+, Ni2+, and Co2+. Here, we show that this specificity allows AMA to block the uptake of Ni2+ and attenuate bacterial Ni-dependent enzymes, offering a potential strategy for reducing virulence. Bacterial exposure to AMA perturbs H2 metabolism, ureolysis, struvite crystallization, and biofilm formation and shows efficacy in a Galleria mellonella animal infection model. The inhibition of Ni-dependent enzymes was aided by Zn2+, which complexes with AMA and competes with the native nickelophore for the uptake of Ni2+. Biochemical analyses demonstrated high-affinity binding of AMA-metal complexes to NikA, the periplasmic substrate-binding protein of the Ni2+ uptake system. Structural examination of NikA in complex with Ni-AMA revealed that the coordination geometry of Ni-AMA mimics the native ligand, Ni-(L-His)2, providing a structural basis for binding AMA-metal complexes. Structure-activity relationship studies of AMA identified regions of the molecule that improve NikA affinity and offer potential routes for further developing this compound as an anti-virulence agent.


Bacterial Proteins , Nickel , Nickel/metabolism , Nickel/chemistry , Animals , Virulence/drug effects , Bacterial Proteins/metabolism , Biofilms/drug effects , Zinc/metabolism , Zinc/chemistry , Moths/microbiology , Urease/metabolism , Urease/antagonists & inhibitors , Biological Transport
8.
Sci Rep ; 14(1): 10882, 2024 05 13.
Article En | MEDLINE | ID: mdl-38740792

The aim of this study was to evaluate the antimicrobial efficacy of an air gas soft jet CAP for its potential use in removing oral biofilms, given that plasma-based technologies have emerged as promising methods in periodontology. Two types of biofilms were developed, one by Streptococcus mutans UA 159 bacterial strain and the other by a complex mixture of saliva microorganisms isolated from a patient with periodontitis. This latter biofilm was characterized via Next Generation Sequencing to determine the main bacterial phyla. The CAP source was applied at a distance of 6 mm for different time points. A statistically significant reduction of both CFU count and XTT was already detected after 60 s of CAP treatment. CLSM analysis supported CAP effectiveness in killing the microorganisms inside the biofilm and in reducing the thickness of the biofilm matrix. Cytotoxicity tests demonstrated the possible use of CAP without important side effects towards human gingival fibroblasts cell line. The current study showed that CAP treatment was able to significantly reduce preformed biofilms developed by both S. mutans and microorganisms isolated by a saliva sample. Further studies should be conducted on biofilms developed by additional saliva donors to support the potential of this innovative strategy to counteract oral pathogens responsible for periodontal diseases.


Biofilms , Plasma Gases , Saliva , Streptococcus mutans , Biofilms/drug effects , Biofilms/growth & development , Humans , Plasma Gases/pharmacology , Streptococcus mutans/drug effects , Streptococcus mutans/physiology , Saliva/microbiology , Fibroblasts/microbiology , Fibroblasts/drug effects , Periodontitis/microbiology , Periodontitis/therapy , Cell Line , Mouth/microbiology
9.
J Nanobiotechnology ; 22(1): 232, 2024 May 08.
Article En | MEDLINE | ID: mdl-38720301

Diabetic wounds pose a challenge to healing due to increased bacterial susceptibility and poor vascularization. Effective healing requires simultaneous bacterial and biofilm elimination and angiogenesis stimulation. In this study, we incorporated polyaniline (PANI) and S-Nitrosoglutathione (GSNO) into a polyvinyl alcohol, chitosan, and hydroxypropyltrimethyl ammonium chloride chitosan (PVA/CS/HTCC) matrix, creating a versatile wound dressing membrane through electrospinning. The dressing combines the advantages of photothermal antibacterial therapy and nitric oxide gas therapy, exhibiting enduring and effective bactericidal activity and biofilm disruption against methicillin-sensitive Staphylococcus aureus, methicillin-resistant Staphylococcus aureus, and Escherichia coli. Furthermore, the membrane's PTT effect and NO release exhibit significant synergistic activation, enabling a nanodetonator-like burst release of NO through NIR irradiation to disintegrate biofilms. Importantly, the nanofiber sustained a uniform release of nitric oxide, thereby catalyzing angiogenesis and advancing cellular migration. Ultimately, the employment of this membrane dressing culminated in the efficacious amelioration of diabetic-infected wounds in Sprague-Dawley rats, achieving wound closure within a concise duration of 14 days. Upon applying NIR irradiation to the PVA-CS-HTCC-PANI-GSNO nanofiber membrane, it swiftly eradicates bacteria and biofilm within 5 min, enhancing its inherent antibacterial and anti-biofilm properties through the powerful synergistic action of PTT and NO therapy. It also promotes angiogenesis, exhibits excellent biocompatibility, and is easy to use, highlighting its potential in treating diabetic wounds.


Anti-Bacterial Agents , Bandages , Biofilms , Nitric Oxide , Photothermal Therapy , Rats, Sprague-Dawley , Wound Healing , Animals , Wound Healing/drug effects , Nitric Oxide/pharmacology , Nitric Oxide/metabolism , Rats , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Photothermal Therapy/methods , Male , Chitosan/chemistry , Chitosan/pharmacology , Nanofibers/chemistry , Escherichia coli/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Diabetes Mellitus, Experimental/complications , Staphylococcus aureus/drug effects , Polyvinyl Alcohol/chemistry , Polyvinyl Alcohol/pharmacology , S-Nitrosoglutathione/pharmacology , S-Nitrosoglutathione/chemistry
10.
Int J Nanomedicine ; 19: 3861-3890, 2024.
Article En | MEDLINE | ID: mdl-38708178

Introduction: Cystic fibrosis (CF) is associated with pulmonary Pseudomonas aeruginosa infections persistent to antibiotics. Methods: To eradicate pseudomonal biofilms, solid lipid nanoparticles (SLNs) loaded with quorum-sensing-inhibitor (QSI, disrupting bacterial crosstalk), coated with chitosan (CS, improving internalization) and immobilized with alginate lyase (AL, destroying alginate biofilms) were developed. Results: SLNs (140-205 nm) showed prolonged release of QSI with no sign of acute toxicity to A549 and Calu-3 cells. The CS coating improved uptake, whereas immobilized-AL ensured >1.5-fold higher uptake and doubled SLN diffusion across the artificial biofilm sputum model. Respirable microparticles comprising SLNs in carbohydrate matrix elicited aerodynamic diameters MMAD (3.54, 2.48 µm) and fine-particle-fraction FPF (65, 48%) for anionic and cationic SLNs, respectively. The antimicrobial and/or antibiofilm activity of SLNs was explored in Pseudomonas aeruginosa reference mucoid/nonmucoid strains as well as clinical isolates. The full growth inhibition of planktonic bacteria was dependent on SLN type, concentration, growth medium, and strain. OD measurements and live/dead staining proved that anionic SLNs efficiently ceased biofilm formation and eradicated established biofilms, whereas cationic SLNs unexpectedly promoted biofilm progression. AL immobilization increased biofilm vulnerability; instead, CS coating increased biofilm formation confirmed by 3D-time lapse confocal imaging. Incubation of SLNs with mature biofilms of P. aeruginosa isolates increased biofilm density by an average of 1.5-fold. CLSM further confirmed the binding and uptake of the labeled SLNs in P. aeruginosa biofilms. Considerable uptake of CS-coated SLNs in non-mucoid strains could be observed presumably due to interaction of chitosan with LPS glycolipids in the outer cell membrane of P. aeruginosa. Conclusion: The biofilm-destructive potential of QSI/SLNs/AL inhalation is promising for site-specific biofilm-targeted interventional CF therapy. Nevertheless, the intrinsic/extrinsic fundamentals of nanocarrier-biofilm interactions require further investigation.


Anti-Bacterial Agents , Biofilms , Chitosan , Liposomes , Nanoparticles , Pseudomonas Infections , Pseudomonas aeruginosa , Biofilms/drug effects , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Humans , Pseudomonas Infections/drug therapy , Nanoparticles/chemistry , Chitosan/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Drug Carriers/chemistry , Cystic Fibrosis/drug therapy , Cystic Fibrosis/microbiology , Lipids/chemistry , Lipids/pharmacology , Quorum Sensing/drug effects , A549 Cells , Alginates/chemistry
11.
Carbohydr Polym ; 337: 122160, 2024 Aug 01.
Article En | MEDLINE | ID: mdl-38710575

Sterilisation technologies are essential to eliminate foodborne pathogens from food contact surfaces. However, most of the current sterilisation methods involve high energy and chemical consumption. In this study, a photodynamic inactivation coating featuring excellent antibacterial activity was prepared by dispersing curcumin as a plant-based photosensitiser in a chitosan solution. The coating generated abundant reactive oxygen species (ROS) after light irradiation at 420 nm, which eradicated ≥99.999 % of Escherichia coli O157:H7. It was also found that ROS damaged the cell membrane, leading to the leakage of cell contents and cell shrinkage on the basis of chitosan. In addition, the production of ROS first excited the bacterial antioxidant defence system resulting in the increase of peroxidase (POD) and superoxide dismutase (SOD). ROS levels exceed its capacity, causing damage to the defence system and further oxidative decomposition of large molecules, such as DNA and proteins, eventually leading to the death of E. coli O157:H7. We also found the curcumin/chitosan coating could effectively remove E. coli O157:H7 biofilms by oxidative of extracellular polysaccharides and proteins. All the contributors made the chitosan/curcumin coating an efficient detergent comparable with HClO.


Anti-Bacterial Agents , Biofilms , Chitosan , Curcumin , Escherichia coli O157 , Photosensitizing Agents , Reactive Oxygen Species , Chitosan/chemistry , Chitosan/pharmacology , Curcumin/pharmacology , Curcumin/chemistry , Escherichia coli O157/drug effects , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Reactive Oxygen Species/metabolism , Biofilms/drug effects , Food Microbiology , Light
12.
PLoS One ; 19(5): e0302717, 2024.
Article En | MEDLINE | ID: mdl-38718045

Bacterial pathogens have remained a major public health concern for several decades. This study investigated the antibacterial activities of Miang extracts (at non-neutral and neutral pH) against Bacillus cereus TISTR 747, Escherichia coli ATCC 22595, Salmonella enterica serovar Typhimurium TISTR 292 and Streptococcus mutans DMST 18777. The potential of Polyvinylpolypyrrolidone (PVPP)-precipitated tannin-free Miang extracts in growth-inhibition of the cariogenic Streptococcus mutans DMST 18777 and its biofilms was also evaluated. The tannin-rich fermented extracts had the best bacterial growth inhibition against S. mutans DMST 18777 with an MIC of 0.29 and 0.72 mg/mL for nonfilamentous fungi (NFP) Miang and filamentous-fungi-processed (FFP) Miang respectively. This observed anti-streptococcal activity still remained after PVPP-mediated precipitation of bioactive tannins especially, in NFP and FFP Miang. Characterization of the PVPP-treated extracts using High performance liquid chromatography quadrupole-time of flight-mass spectrometry (HPLC-QToF-MS) analysis, also offered an insight into probable compound classes responsible for the activities. In addition, Crystal violet-staining also showed better IC50 values for NFP Miang (4.30 ± 0.66 mg/mL) and FFP Miang (12.73 ± 0.11 mg/mL) against S. mutans DMST 18777 biofilms in vitro. Homology modeling and molecular docking analysis using HPLC-MS identified ligands in tannin-free Miang supernatants, was performed against modelled S. mutans DMST 18777 sortase A enzyme. The in silico analysis suggested that the inhibition by NFP and FFP Miang might be attributed to the presence of ellagic acid, flavonoid aglycones, and glycosides. Thus, these Miang extracts could be optimized and explored as natural active pharmaceutical ingredients (NAPIs) for applications in oral hygienic products.


Anti-Bacterial Agents , Biofilms , Microbial Sensitivity Tests , Molecular Docking Simulation , Plant Extracts , Streptococcus mutans , Tannins , Streptococcus mutans/drug effects , Streptococcus mutans/growth & development , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Tannins/pharmacology , Tannins/chemistry , Biofilms/drug effects , Biofilms/growth & development , Plant Extracts/pharmacology , Plant Extracts/chemistry , Bacterial Proteins/metabolism
13.
Int J Nanomedicine ; 19: 3991-4005, 2024.
Article En | MEDLINE | ID: mdl-38720939

Purpose: Surgical site infections pose a significant challenge for medical services. Systemic antibiotics may be insufficient in preventing bacterial biofilm development. With the local administration of antibiotics, it is easier to minimize possible complications, achieve drugs' higher concentration at the injured site, as well as provide their more sustained release. Therefore, the main objective of the proposed herein studies was the fabrication and characterization of innovative hydrogel-based composites for local vancomycin (VAN) therapy. Methods: Presented systems are composed of ionically gelled chitosan particles loaded with vancomycin, embedded into biomimetic collagen/chitosan/hyaluronic acid-based hydrogels crosslinked with genipin and freeze-dried to serve in a flake/disc-like form. VAN-loaded carriers were characterized for their size, stability, and encapsulation efficiency (EE) using dynamic light scattering technique, zeta potential measurements, and UV-Vis spectroscopy, respectively. The synthesized composites were tested in terms of their physicochemical and biological features. Results: Spherical structures with sizes of about 200 nm and encapsulation efficiencies reaching values of approximately 60% were obtained. It was found that the resulting particles exhibit stability over time. The antibacterial activity of the developed materials against Staphylococcus aureus was established. Moreover, in vitro cell culture study revealed that the surfaces of all prepared systems are biocompatible as they supported the proliferation and adhesion of the model MG-63 cells. In addition, we have demonstrated significantly prolonged VAN release while minimizing the initial burst effect for the composites compared to bare nanoparticles and verified their desired physicochemical features during swellability, and degradation experiments. Conclusion: It is expected that the developed herein system will enable direct delivery of the antibiotic at an exposed to infections surgical site, providing drugs sustained release and thus will reduce the risk of systemic toxicity. This strategy would both inhibit biofilm formation and accelerate the healing process.


Anti-Bacterial Agents , Chitosan , Hydrogels , Staphylococcus aureus , Vancomycin , Vancomycin/chemistry , Vancomycin/pharmacology , Vancomycin/administration & dosage , Vancomycin/pharmacokinetics , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Hydrogels/chemistry , Hydrogels/pharmacology , Staphylococcus aureus/drug effects , Humans , Chitosan/chemistry , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Drug Carriers/chemistry , Collagen/chemistry , Collagen/pharmacology , Particle Size , Drug Liberation , Surgical Wound Infection/prevention & control , Surgical Wound Infection/drug therapy , Microbial Sensitivity Tests , Biofilms/drug effects
14.
Arch Microbiol ; 206(6): 251, 2024 May 10.
Article En | MEDLINE | ID: mdl-38727840

The prevalence of Candida albicans infection has increased during the past few years, which contributes to the need for new, effective treatments due to the increasing concerns regarding antifungal drug toxicity and multidrug resistance. Butyl isothiocyanate (butylITC) is a glucosinolate derivative, and has shown a significant antifungal effect contrary to Candida albicans. Additionally, how butylITC affects the virulence traits of C. albicans and molecular mode of actions are not well known. Present study shows that at 17.36 mM concentration butylITC inhibit planktonic growth. butylITC initially slowed the hyphal transition at 0.542 mM concentration. butylITC hampered biofilm development, and inhibits biofilm formation at 17.36 mM concentration which was analysed using metabolic assay (XTT assay) and Scanning Electron Microscopy (SEM). In addition, it was noted that butylITC inhibits ergosterol biosynthesis. The permeability of cell membranes was enhanced by butylITC treatment. Moreover, butylITC arrests cells at S-phase and induces intracellular Reactive Oxygen Species (ROS) accumulation in C. albicans. The results suggest that butylITC may have a dual mode of action, inhibit virulence factors and modulate cellular processes like inhibit ergosterol biosynthesis, cell cycle arrest, induces ROS production which leads to cell death in C. albicans.


Antifungal Agents , Biofilms , Candida albicans , Cell Membrane , Isothiocyanates , Oxidative Stress , Reactive Oxygen Species , Candida albicans/drug effects , Candida albicans/physiology , Biofilms/drug effects , Antifungal Agents/pharmacology , Isothiocyanates/pharmacology , Oxidative Stress/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Reactive Oxygen Species/metabolism , Microbial Sensitivity Tests , Cell Cycle/drug effects , Hyphae/drug effects , Hyphae/growth & development , Ergosterol/metabolism
15.
Sci Rep ; 14(1): 10200, 2024 05 03.
Article En | MEDLINE | ID: mdl-38702397

Today, antibiotic therapies that previously worked well against certain bacteria due to their natural sensitivity, are becoming less effective. Honey has been proven to inhibit the biofilm formation of some respiratory bacteria, however few data are available on how the storage time affects the antibacterial effect. The activity of black locust, goldenrod, linden and sunflower honeys from three consecutive years (2020, 2021, 2022) was analyzed in 2022 against Gram-negative (Haemophilus influenzae, H. parainfluenzae, Pseudomonas aeruginosa) and Gram-positive (Streptococcus pneumoniae) bacteria using in vitro microbiological methods. After determining the physicochemical parameters of honey, broth microdilution was applied to determine the minimum inhibitory concentration of each honey type against each bacterium, and crystal violet assay was used to test their antibiofilm effect. The possible mechanism of action was explored with membrane degradation test, while structural changes were illustrated with scanning electron microscopy. Honeys stored for one or two years were darker than fresh honeys, while older honeys had significantly lower antibacterial activity. The most remarkable inhibitory effect was exerted by linden and sunflower honeys, and P. aeruginosa proved to be the most resistant bacterium. Based on our results, honey intended for medicinal purposes should be used as fresh as possible during a treatment.


Anti-Bacterial Agents , Honey , Microbial Sensitivity Tests , Honey/analysis , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Time Factors , Pseudomonas aeruginosa/drug effects , Food Storage/methods , Humans
16.
Appl Microbiol Biotechnol ; 108(1): 316, 2024 May 03.
Article En | MEDLINE | ID: mdl-38700735

Nowadays, it is very important to produce new-generation drugs with antimicrobial properties that will target biofilm-induced infections. The first target for combating these microorganisms, which are the source itself. Antimicrobial peptides, which are more effective than antibiotics due to their ability to kill microorganisms and use a different metabolic pathway, are among the new options today. The aim of this study is to develop new-generation antibiotics that inhibit both biofilm-producing bacteria and the biofilm itself. For this purpose, we designed four different peptides by combining two amino acid forms (D- and L-) with the same sequence having alpha helix structures. It was found that the combined use of these two forms can increase antimicrobial efficacy more than 30-fold. These results are supported by molecular modeling and scanning electron microscopy (SEM), at the same time cytotoxicity (IC50) and hemotoxicity (HC50) values remained within the safe range. Furthermore, antibiofilm activities of these peptides were investigated. Since the existing biofilm inhibition methods in the literature do not technically simulate the exact situation, in this study, we have developed a real-time observable biofilm model and a new detection method based on it, which we call the CoMIC method. Findings have shown that the NET1 peptide with D-leucine amino acid in its structure and the NET3 peptide with D-arginine amino acid in its structure are effective in inhibiting biofilm. As a conclusion, our peptides can be considered as potential next-generation broad-spectrum antibiotic molecule/drug candidates that might be used in biofilm and clinical important bacteria. KEY POINTS: • Antimicrobial peptides were developed to inhibit both biofilms producing bacteria and the biofilm itself. • CoMIC will fill a very crucial gap in understanding biofilms and conducting the necessary quantitative studies. • Molecular modelling studies, NET1 peptide molecules tends to move towards and adhere to the membrane within nanoseconds.


Anti-Bacterial Agents , Antimicrobial Peptides , Biofilms , Microbial Sensitivity Tests , Biofilms/drug effects , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Models, Molecular , Microscopy, Electron, Scanning , Bacteria/drug effects
17.
Technol Cancer Res Treat ; 23: 15330338241250244, 2024.
Article En | MEDLINE | ID: mdl-38693842

Single biofilm biomimetic nanodrug delivery systems based on single cell membranes, such as erythrocytes and cancer cells, have immune evasion ability, good biocompatibility, prolonged blood circulation, and high tumor targeting. Because of the different characteristics and functions of each single cell membrane, more researchers are using various hybrid cell membranes according to their specific needs. This review focuses on several different types of biomimetic nanodrug-delivery systems based on composite biofilms and looks forward to the challenges and possible development directions of biomimetic nanodrug-delivery systems based on composite biofilms to provide reference and ideas for future research.


Antineoplastic Agents , Biofilms , Biomimetics , Drug Delivery Systems , Neoplasms , Humans , Neoplasms/drug therapy , Biofilms/drug effects , Biomimetics/methods , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Biomimetic Materials/chemistry , Animals , Drug Carriers/chemistry
18.
BMC Biotechnol ; 24(1): 28, 2024 May 04.
Article En | MEDLINE | ID: mdl-38702622

Scientists know very little about the mechanisms underlying fish skin mucus, despite the fact that it is a component of the immune system. Fish skin mucus is an important component of defence against invasive infections. Recently, Fish skin and its mucus are gaining interest among immunologists. Characterization was done on the obtained silver nanoparticles Ag combined with Clarias gariepinus catfish epidermal mucus proteins (EMP-Ag-NPs) through UV-vis, FTIR, XRD, TEM, and SEM. Ag-NPs ranged in size from 4 to 20 nm, spherical in form and the angles were 38.10°, 44.20°, 64.40°, and 77.20°, Where wavelength change after formation of EMP-Ag-NPs as indicate of dark brown, the broad band recorded at wavelength at 391 nm. Additionally, the antimicrobial, antibiofilm and anticancer activities of EMP-Ag-NPs was assessed. The present results demonstrate high activity against unicellular fungi C. albicans, followed by E. faecalis. Antibiofilm results showed strong activity against both S. aureus and P. aeruginosa pathogens in a dose-dependent manner, without affecting planktonic cell growth. Also, cytotoxicity effect was investigated against normal cells (Vero), breast cancer cells (Mcf7) and hepatic carcinoma (HepG2) cell lines at concentrations (200-6.25 µg/mL) and current results showed highly anticancer effect of Ag-NPs at concentrations 100, 5 and 25 µg/mL exhibited rounding, shrinkage, deformation and granulation of Mcf7 and HepG2 with IC50 19.34 and 31.16 µg/mL respectively while Vero cells appeared rounded at concentration 50 µg/mL and normal shape at concentration 25, 12.5 and 6.25 µg/ml with IC50 35.85 µg/mL. This study evidence the potential efficacy of biologically generated Ag-NPs as a substitute medicinal agent against harmful microorganisms. Furthermore, it highlights their inhibitory effect on cancer cell lines.


Biofilms , Catfishes , Metal Nanoparticles , Silver , Metal Nanoparticles/chemistry , Biofilms/drug effects , Biofilms/growth & development , Silver/chemistry , Silver/pharmacology , Animals , Humans , Mucus/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Vero Cells , Fish Proteins/pharmacology , Fish Proteins/chemistry , Fish Proteins/metabolism , Chlorocebus aethiops , Cell Line, Tumor , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Candida albicans/drug effects , Epidermis/metabolism
19.
BMC Microbiol ; 24(1): 152, 2024 May 03.
Article En | MEDLINE | ID: mdl-38702660

BACKGROUND: Pseudomonas aeruginosa is a common cause of nosocomial infections. However, the emergence of multidrug-resistant strains has complicated the treatment of P. aeruginosa infections. While polymyxins have been the mainstay for treatment, there is a global increase in resistance to these antibiotics. Therefore, our study aimed to determine the prevalence and molecular details of colistin resistance in P. aeruginosa clinical isolates collected between June 2019 and May 2023, as well as the genetic linkage of colistin-resistant P. aeruginosa isolates. RESULTS: The resistance rate to colistin was 9% (n = 18) among P. aeruginosa isolates. All 18 colistin-resistant isolates were biofilm producers and carried genes associated with biofilm formation. Furthermore, the presence of genes encoding efflux pumps, TCSs, and outer membrane porin was observed in all colistin-resistant P. aeruginosa strains, while the mcr-1 gene was not detected. Amino acid substitutions were identified only in the PmrB protein of multidrug- and colistin-resistant strains. The expression levels of mexA, mexC, mexE, mexY, phoP, and pmrA genes in the 18 colistin-resistant P. aeruginosa strains were as follows: 88.8%, 94.4%, 11.1%, 83.3%, 83.3%, and 38.8%, respectively. Additionally, down-regulation of the oprD gene was observed in 44.4% of colistin-resistant P. aeruginosa strains. CONCLUSION: This study reports the emergence of colistin resistance with various mechanisms among P. aeruginosa strains in Ardabil hospitals. We recommend avoiding unnecessary use of colistin to prevent potential future increases in colistin resistance.


Anti-Bacterial Agents , Bacterial Proteins , Colistin , Microbial Sensitivity Tests , Pseudomonas Infections , Pseudomonas aeruginosa , Transcription Factors , Colistin/pharmacology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/isolation & purification , Anti-Bacterial Agents/pharmacology , Humans , Bacterial Proteins/genetics , Pseudomonas Infections/microbiology , Pseudomonas Infections/epidemiology , Prevalence , Drug Resistance, Multiple, Bacterial/genetics , Biofilms/drug effects , Biofilms/growth & development , Hospitals , Drug Resistance, Bacterial/genetics , Cross Infection/microbiology , Cross Infection/epidemiology , Membrane Transport Proteins/genetics , Porins/genetics
20.
J Appl Microbiol ; 135(5)2024 May 01.
Article En | MEDLINE | ID: mdl-38702839

AIMS: Macroalgae harbor a rich epiphytic microbiota that plays a crucial role in algal morphogenesis and defense mechanisms. This study aims to isolate epiphytic cultivable microbiota from Ulva sp. surfaces. Various culture media were employed to evaluate a wide range of cultivable microbiota. Our objective was to assess the antibacterial and biofilm-modulating activities of supernatants from isolated bacteria. METHODS AND RESULTS: Sixty-nine bacterial isolates from Ulva sp. were identified based on 16S rRNA gene sequencing. Their antibacterial activity and biofilm modulation potential were screened against three target marine bacteria: 45%, mostly affiliated with Gammaproteobacteria and mainly grown on diluted R2A medium (R2Ad), showed strong antibacterial activity, while 18% had a significant impact on biofilm modulation. Molecular network analysis was carried out on four bioactive bacterial supernatants, revealing new molecules potentially responsible for their activities. CONCLUSION: R2Ad offered the greatest diversity and proportion of active isolates. The molecular network approach holds promise for both identifying bacterial isolates based on their molecular production and characterizing antibacterial and biofilm-modulating activities.


Anti-Bacterial Agents , Bacteria , Biofilms , RNA, Ribosomal, 16S , Ulva , Biofilms/drug effects , Biofilms/growth & development , Ulva/microbiology , Anti-Bacterial Agents/pharmacology , RNA, Ribosomal, 16S/genetics , Bacteria/genetics , Bacteria/classification , Bacteria/isolation & purification , Bacteria/drug effects , Microbiota , Phylogeny , Biodiversity , Seaweed/microbiology
...