Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Rev. argent. microbiol ; 53(4): 31-40, Dec. 2021. graf
Article in English | LILACS | ID: biblio-1376419

ABSTRACT

ABSTRACT Increasing antimicrobial resistance amongStaphylococcus aureusnecessitates a new antimicrobial with a different site of action. We have isolated a novel cyclic peptide-1 (ASP-1) fromBacillussubtiliswith potent activity against methicillin-resistantS. aureus(MRSA) at a minimum inhibitory concentration (MIC) of 8-64μg/ml. Scanning electron micrographs demonstrated drastic changes in the cellular architecture of ASP-1 treated cells ofS. aureusATCC 29213 and an MRSA clinical isolate at MICs, with damages to the cell wall, membrane lysis and probable leakage of cytoplasmic contents at minimum bactericidal concentrations. The ultrastructure alterations induced by ASP-1 have also been compared with those of oxacillin-treated MRSA cells at its MIC using scanning electron microscopy.


RESUMEN El incremento de la resistencia antimicrobiana entre los tipos deS. aureusexige un nuevo agente antimicrobiano con un sitio de acción diferente. Aislamos un nuevo péptido cíclico (ASP-1) deBacillussubtiliscon potente actividad frente aS. aureusresistente a meticilina (SARM) en una concentración inhibitoria mínima (CIM) de 8-64μg/ml. Las micrografías obtenidas con microscopio electrónico de barrido mostraron cambios drásticos en la arquitectura celular de las células deS. aureusATCC 29213 tratadas con ASP-1, y un aislamiento clínico de SARM a la CIM, con daños a la pared celular, lisis de la membrana y probable fuga de contenido citoplasmático a concentraciones bactericidas mínimas. Comparamos también, las alteraciones de la ultraestructura inducidas por ASP-1 con las de células de SARM tratadas con oxacilina a su CIM, utilizando microscopio electrónico de barrido.


Subject(s)
Peptides, Cyclic/pharmacology , Methicillin-Resistant Staphylococcus aureus , Anti-Bacterial Agents , Bacillus subtilis/chemistry , Microscopy, Electron, Scanning , Microbial Sensitivity Tests , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Anti-Bacterial Agents/pharmacology
2.
PLoS One ; 16(10): e0258607, 2021.
Article in English | MEDLINE | ID: mdl-34648570

ABSTRACT

Staphylococcus aureus and Methicillin-resistant S. aureus (MRSA) remains one of the major concerns of healthcare associated and community-onset infections worldwide. The number of cases of treatment failure for infections associated with resistant bacteria is on the rise, due to the decreasing efficacy of current antibiotics. Notably, Acrophialophora levis, a thermophilous fungus species, showed antibacterial activity, namely against S. aureus and clinical MRSA strains. The ethyl acetate extract of culture filtrate was found to display significant activity against S. aureus and MRSA with a minimum inhibitory concentration (MIC) of 1 µg/mL and 4 µg/mL, respectively. Scanning electron micrographs demonstrated drastic changes in the cellular architecture of metabolite treated cells of S. aureus and an MRSA clinical isolate. Cell wall disruption, membrane lysis and probable leakage of cytoplasmic are hallmarks of the antibacterial effect of fungal metabolites against MRSA. The ethyl acetate extract also showed strong antioxidant activity using two different complementary free radicals scavenging methods, DPPH and ABTS with efficiency of 55% and 47% at 1 mg/mL, respectively. The total phenolic and flavonoid content was found to be 50 mg/GAE and 20 mg/CAE, respectively. More than ten metabolites from different classes were identified: phenolic acids, phenylpropanoids, sesquiterpenes, tannins, lignans and flavonoids. In conclusion, the significant antibacterial activity renders this fungal strain as a bioresource for natural compounds an interesting alternative against resistant bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Biological Factors/pharmacology , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Sordariales/chemistry , Acetates/chemistry , Anti-Bacterial Agents/chemistry , Antioxidants/chemistry , Biological Factors/chemistry , Flavonoids/isolation & purification , Hydroxybenzoates/isolation & purification , India , Lignans/isolation & purification , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Sesquiterpenes/isolation & purification , Tannins/isolation & purification
3.
Int J Biol Macromol ; 181: 540-551, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-33766592

ABSTRACT

Biomaterial research has improved the delivery and efficacy of drugs over a wide range of pharmaceutical applications. The objective of this study was to synthesize benzodioxane coupled piperazine decorated chitosan silver nanoparticle (Bcp*C@AgNPs) against methicillin-resistant Staphylococcus aureus (MRSA) and to assess the nanoparticle as an effective candidate for antibacterial and anti-biofilm care. Antibacterial activity of the compound was examined and minimum inhibitory concentration (MIC) was observed at (10.21 ± 0.03 ZOI) a concentration of 200 µg/mL. The Bcp*C@AgNPs interferes with surface adherence of MRSA, suggesting an anti-biofilm distinctive property that is verified for the first time by confocal laser microscopic studies. By ADMET studies the absorption, distribution, metabolism, excretion and toxicity of the compound was examined. The interaction solidity and the stability of the compound when surrounded by water molecules were analyzed by docking and dynamic simulation analysis. The myoblast cell line (L6) was considered for toxicity study and was observed that the compound exhibited less toxic effect. This current research highlights the biocidal efficiency of Bcp*C@AgNPs with their bactericidal and anti-biofilm properties over potential interesting clinical trial targets in future.


Subject(s)
Biofilms/drug effects , Chitosan/chemical synthesis , Dioxanes/pharmacology , Metal Nanoparticles/chemistry , Methicillin-Resistant Staphylococcus aureus/physiology , Molecular Docking Simulation , Molecular Dynamics Simulation , Piperazine/pharmacology , Silver/pharmacology , Animals , Anti-Infective Agents/pharmacology , Cell Line , Chitosan/chemistry , Fluorescence , Ligands , Metal Nanoparticles/ultrastructure , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Microbial Sensitivity Tests , Piperazine/chemistry , Plankton/drug effects , Rats , Toxicity Tests
4.
Rev Argent Microbiol ; 53(4): 281-286, 2021.
Article in English | MEDLINE | ID: mdl-33608109

ABSTRACT

Increasing antimicrobial resistance among Staphylococcus aureus necessitates a new antimicrobial with a different site of action. We have isolated a novel cyclic peptide-1 (ASP-1) from Bacillussubtilis with potent activity against methicillin-resistant S. aureus (MRSA) at a minimum inhibitory concentration (MIC) of 8-64µg/ml. Scanning electron micrographs demonstrated drastic changes in the cellular architecture of ASP-1 treated cells of S. aureus ATCC 29213 and an MRSA clinical isolate at MICs, with damages to the cell wall, membrane lysis and probable leakage of cytoplasmic contents at minimum bactericidal concentrations. The ultrastructure alterations induced by ASP-1 have also been compared with those of oxacillin-treated MRSA cells at its MIC using scanning electron microscopy.


Subject(s)
Anti-Bacterial Agents , Methicillin-Resistant Staphylococcus aureus , Peptides, Cyclic/pharmacology , Anti-Bacterial Agents/pharmacology , Bacillus subtilis/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Microbial Sensitivity Tests , Microscopy, Electron, Scanning
5.
Sci Rep ; 11(1): 4240, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33608582

ABSTRACT

The herbal products proved to be more promising antimicrobials even though their antimicrobial activity is milder than commercially available antibiotics. Moreover, herbal drugs may act synergistically with antibiotics to kill microbes. In this study, we aimed to enhance the activity of penicillin against MRSA through combination with the active saponin fraction isolated from the Zygophyllum album plant. Three different types of metabolites (saponins, sterols, and phenolics) have been extracted from Zygophyllum album with ethanol and purified using different chromatographic techniques. The antibacterial activity of crude extract and the separated metabolites were checked against MRSA isolates, Saponin fraction (ZA-S) was only the active one followed by the crude extract. Therefore, the compounds in this fraction were identified using ultra-high-performance liquid chromatography connected to quadrupole time-of-flight mass spectrometry (UHPLC/QTOF-MS) operated in positive and negative ionization modes. UHPLC/QTOF-MS revealed the presence of major six ursane-type tritepenoidal saponins (Quinovic acid, Quinovic acid 3ß-O-ß-D-quinovopyranoside, Zygophylloside C, Zygophylloside G, Zygophylloside K and Ursolic acid), in addition to Oleanolic acid. Interaction studies between saponin fraction and penicillin against MRSA were performed through the checkerboard method and time-kill assay. According to checkerboard results, only three combinations showed a fractional inhibitory concentration index less than 0.5 at concentrations of (62.5 + 312.5, 62.5 + 156.25, and 62.5 + 78.125 of penicillin and ZA-S, respectively). Time kill assay results showed that the highest reduction in log10 colony-forming unit (CFU)/ml of initial inoculum of MRSA after 24 h occurred by 3.7 at concentrations of 62.5 + 312.5 (µg/µg)/ml of penicillin and ZA-S, respectively. Thus, the combination between saponin fraction of Zygophyllum album and penicillin with these concentrations could be a potential agent against MRSA that can serve as possible model for new antibacterial drug.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Chromatography, High Pressure Liquid , Combined Modality Therapy , Disease Management , Drug Discovery , Drug Synergism , Drug Therapy, Combination , Humans , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Microbial Sensitivity Tests , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Treatment Outcome
6.
J Microbiol Immunol Infect ; 54(3): 527-530, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33060039

ABSTRACT

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) threatens human health. A local CA-MRSA with ST8/SCCmecIVl (CA-MRSA/J) has emerged in Japan, being associated with progression from bullous impetigo to potentially fatal invasive infection. We found that CA-MRSA/J has unique bacterial surface structures, spikes, spikes with a cap, and long spikes, reflecting clinical origins.


Subject(s)
Community-Acquired Infections/microbiology , Methicillin-Resistant Staphylococcus aureus/chemistry , Humans , Japan , Leukocidins/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Microscopy, Electron , Staphylococcal Infections/microbiology
7.
Int J Nanomedicine ; 15: 4779-4791, 2020.
Article in English | MEDLINE | ID: mdl-32753866

ABSTRACT

BACKGROUND: Considering the timeline required for the development of novel antimicrobial drugs, increased attention should be given to repurposing old drugs and improving antimicrobial efficacy, particularly for chronic infections associated with biofilms. Methicillin-susceptible Staphylococcus aureus (MSSA) and methicillin-resistant S. aureus (MRSA) are common causes of biofilm-associated infections but produce different biofilm matrices. MSSA biofilm cells are typically embedded in an extracellular polysaccharide matrix, whereas MRSA biofilms comprise predominantly of surface proteins and extracellular DNA (eDNA). Nanoparticles (NPs) have the potential to enhance the delivery of antimicrobial agents into biofilms. However, the mechanisms which influence the interactions between NPs and the biofilm matrix are not yet fully understood. METHODS: To investigate the influence of NPs surface chemistry on vancomycin (VAN) encapsulation and NP entrapment in MRSA and MSSA biofilms, mesoporous silica nanoparticles (MSNs) with different surface functionalization (bare-B, amine-D, carboxyl-C, aromatic-A) were synthesised using an adapted Stöber method. The antibacterial efficacy of VAN-loaded MSNs was assessed against MRSA and MSSA biofilms. RESULTS: The two negatively charged MSNs (MSN-B and MSN-C) showed a higher VAN loading in comparison to the positively charged MSNs (MSN-D and MSN-A). Cellular binding with MSN suspensions (0.25 mg mL-1) correlated with the reduced viability of both MSSA and MRSA biofilm cells. This allowed the administration of low MSNs concentrations while maintaining a high local concentration of the antibiotic surrounding the bacterial cells. CONCLUSION: Our data suggest that by tailoring the surface functionalization of MSNs, enhanced bacterial cell targeting can be achieved, leading to a novel treatment strategy for biofilm infections.


Subject(s)
Anti-Infective Agents/pharmacology , Biofilms , Methicillin-Resistant Staphylococcus aureus/physiology , Nanoparticles/chemistry , Biofilms/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Microbial Sensitivity Tests , Nanoparticles/ultrastructure , Proton Magnetic Resonance Spectroscopy , Silicon Dioxide/chemistry , Vancomycin/pharmacology
8.
Pharmacol Res ; 160: 105067, 2020 10.
Article in English | MEDLINE | ID: mdl-32650057

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) infections are one of the most serious surgery complications, and their prevention is of utmost importance. Flufenamic acid is a non-steroid anti-inflammatory drug approved for clinical use to relieve inflammation and pain in rheumatoid arthritis patients. In this study, we explored the antibacterial efficacy of flufenamic acid and the mechanisms underlying this effect. By using minimal inhibitory concentration (MIC), time-kill, resistance induction assays, and the antibiotic synergy test, we demonstrated that flufenamic acid inhibited the growth of methicillin-resistant staphylococci and did not induce resistance when it was used at the MIC. Furthermore, flufenamic acid acted synergistically with the beta-lactam antibiotic oxacillin and did not show significant toxicity toward mammalian cells. The biofilm inhibition assay revealed that flufenamic acid could prevent biofilm formation on medical implants and destroy the ultrastructure of the bacterial cell wall. RNA sequencing and quantitative RT-PCR indicated that flufenamic acid inhibited the expression of genes associated with peptidoglycan biosynthesis, beta-lactam resistance, quorum sensing, and biofilm formation. Furthermore, flufenamic acid efficiently ameliorated a local infection caused by MRSA in mice. In conclusion, flufenamic acid may be a potent therapeutic compound against MRSA infections and a promising candidate for antimicrobial coating of implants and surgical devices.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Flufenamic Acid/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Ampicillin Resistance/genetics , Animals , Drug Synergism , Gene Expression Regulation, Bacterial/drug effects , Humans , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Mice , Microbial Sensitivity Tests , Oxacillin/pharmacology , Quorum Sensing/drug effects , Thoracic Wall/drug effects , Thoracic Wall/ultrastructure
9.
Int J Med Microbiol ; 310(5): 151435, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32654773

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is one of the important causes of food poisoning and infectious diseases worldwide, it can produce a large number of virulence factors, enhance the colonization ability of the host so that it can quickly colonize and spread on the surface of the objects. Essential oil (EO) is one of the natural products with antimicrobial properties, can be used as an important source of antibacterial agent discovery, and has a broad development prospect. However, the unclear mechanisms of antibacterial action have become an obstacle to its further development and use. Hence, the objective of the present study was to reveal the antibacterial mechanism of EO from Amomum villosum Lour (A villosum Lour) against MRSA using label-free quantitative proteomics, investigate the effect of EO on the bacterial proteome, enzymatic activities and leakage of bacterial intracellular biomacromolecule. Proteomic analysis of MRSA in the presence of EO found that a total of 144 differential expressed proteins (DEPs) between the control and treatment group, in which 42 proteins were distinctly up-regulated and 102 proteins were down-regulated. Besides, sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis, determination of cell membrane permeability and apoptosis, scanning electron microscopy (SEM) observations, bacterial surface hydrophobicity, and biofilm formation measurement were performed. Collectively, the above results indicated that the cell membrane damage by EO leads to the loss of membrane integrity and causes leakage of intracellular macromolecular substances, inhibition of protein, and biofilm synthesis. These findings manifested that EO exerts antibacterial effect by multiple avenues and expands our understanding of the antibacterial mechanism, it has potential application value in food preservative and pharmaceutical industries.


Subject(s)
Apoptosis/drug effects , Bacterial Proteins/metabolism , Biofilms/drug effects , Cell Membrane Permeability/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/metabolism , Oils, Volatile/pharmacology , Amomum/chemistry , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Biosynthetic Pathways , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Microscopy, Electron, Scanning , Oils, Volatile/chemistry , Proteome
10.
J Ethnopharmacol ; 261: 113167, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-32730885

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Cochlospermum regium, known as "algodãozinho", is an important plant belonging to Brazilian biodiversity used in traditional medicine to treat infections, wounds and skin conditions. AIM OF THE STUDY: To assess the effects of aqueous and ethanolic extracts from C. regium leaves on methicillin-resistant Staphylococcus aureus planktonic cells and biofilm formation. MATERIAL AND METHODS: The phytochemical characterization of the extracts was carried out by quantification of flavonoids, phenols and tannins and HPLC-DAD. Minimum inhibitory concentrations, cell viability, and enzyme activity inhibition were determined in planktonic cells exposed to C. regium extracts. The effect of the extracts on biofilms was assessed by quantifying colony-forming units (CFUs) and the extracellular matrix, and by visualizing the biofilm structure using scanning electron microscopy. RESULTS: Leaf extract contents showed high concentration of phenols and the gallic and ellagic acids were identified. The extracts showed potent antimicrobial activities at concentrations ranging from 62.5-250 µg/mL, and decreased coagulase activity. In addition, the extracts prevented biofilm formation, and the aqueous extract completely inhibited its formation. CONCLUSIONS: C. regium extracts stand out as promising alternative treatments for the prevention and treatment of methicillin-resistant Staphylococcus aureus infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Bixaceae , Methicillin-Resistant Staphylococcus aureus/drug effects , Plant Extracts/pharmacology , Anti-Bacterial Agents/isolation & purification , Biofilms/growth & development , Bixaceae/chemistry , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Microbial Sensitivity Tests , Microbial Viability/drug effects , Plant Extracts/isolation & purification , Plant Leaves/chemistry
11.
J Ethnopharmacol ; 254: 112669, 2020 May 23.
Article in English | MEDLINE | ID: mdl-32087316

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The development of new inhibitors of bacterial virulence factors from natural origin has recently received significant attention. Callistemon citrinus Skeels is an important plant of great medicinal value. Its antimicrobial activity is well documented. Although several compounds were isolated from this plant, the actual bioactive compounds responsible for its antimicrobial activity are still unrevealed. AIM OF THE STUDY: To evaluate the effect of C. citrinus crude extract and isolated compounds on methicillin-resistant and sensitive Staphylococcus aureus. MATERIALS AND METHODS: The methylene chloride-methanol extract (MME) of C. citrinus leaves was prepared by Soxhlet apparatus. Biologically guided fractionation of MME was accomplished using several normal and reversed phase silica gel columns. The potency of MME and its isolated compounds against methicillin-resistant Staphylococcus aureus (MRSA) and methicillin-sensitive S. aureus (MSSA) was evaluated. In addition, the mechanism of resistance was studied using three virulence factors; antibiofilm activity, inhibition of staphyloxanthin biosynthesis and effect on acid tolerance. Ultrastructural changes in MRSA and MSSA were observed by TEM to understand mode of action of these compounds. RESULTS: Pulverulentone A (C1), 8- desmethyl eucalyptin (C2) and eucalyptin (C3) were isolated from the most bioactive fraction of MME. Confocal scanning laser microscopy images revealed that C. citrinus isolated compounds destroyed the intact architecture of biofilm, thickness and reduced its biomass. Pulverulentone A (C1) showed the most potent anti-biofilm activity up to 71% and 62.3% against MRSA and MSSA, respectively. It also exhibited the highest inhibition of staphyloxanthin biosynthesis of MRSA and MSSA by 55.6% and 54.5%, respectively. The bacterial cell membrane was compromised, losing its integrity and releasing important cellular constituents when exposed to C1-C3 CONCLUSIONS: C. citrinus phenolics and acylphloroglucinols may serve as potential source of plant-based antibacterials and thus could be implicated to control MRSA biofilm formation.


Subject(s)
Biofilms/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Myrtaceae/chemistry , Plant Extracts/pharmacology , Staphylococcus aureus/drug effects , Xanthophylls/antagonists & inhibitors , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Microbial Sensitivity Tests , Microscopy, Electron, Scanning Transmission , Phytochemicals/isolation & purification , Phytochemicals/pharmacology , Plant Extracts/chemistry , Plant Leaves/chemistry , Staphylococcus aureus/ultrastructure
12.
Int J Biol Macromol ; 144: 9-18, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31821826

ABSTRACT

Kappa-Carrageenan wrapped ZnO nanoparticles (KC-ZnO NPs) was synthesized, physico-chemically characterized and evaluated their biocompatibility and antimicrobial therapy against MRSA. XRD showed the highly crystalline and hexagonal phase structure of ZnO NPs. FETEM confirmed the spherical and hexagonal shaped particle with the mean size of 97.03 ± 9.05 nm. The synthesized KC-ZnO NPs exhibited significant antibacterial activity against MRSA. The biofilm growth of MRSA was greatly inhibited at 100 µg/ml as observed through live and dead cell assay. KC-ZnO NPs have shown invitro anti-inflammatory activity (82%) at 500 µg/ml. KC-ZnO NPs was non-toxic to NIH3T3 mouse embryonic fibroblasts cell lines. Further, no apoptotic and necrotic mediated death in NIH3T3 mouse embryonic fibroblasts cells were noticed by flow cytometric analysis. KC-ZnO NPs have good biocompatibility as recorded by the least hemolysis percentage (<3%) up to 100 µg/ml, which is much lesser than the acceptable limit. In addition, ecosafety analysis has shown that KC-ZnO NPs and kappa karrageenan (0-500 µg/ml) caused no mortality of A. salina after 48 h. However, bare zinc acetate has shown 35% mortality of A. salina after 48 h. The results conclude that KC-ZnO NPs could be a novel antibacterial therapy for the treatment of MRSA associated infectious.


Subject(s)
Carrageenan/pharmacology , Drug Delivery Systems , Metal Nanoparticles/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Zinc Oxide/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Artemia/drug effects , Biocompatible Materials/pharmacology , Biofilms/drug effects , Carrageenan/chemical synthesis , Carrageenan/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Embryo, Mammalian/cytology , Erythrocytes/drug effects , Erythrocytes/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Metal Nanoparticles/ultrastructure , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Mice , Microbial Sensitivity Tests , NIH 3T3 Cells , Reactive Oxygen Species/metabolism
13.
ACS Nano ; 13(12): 13965-13984, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31730327

ABSTRACT

Multidrug-resistant (MDR) bacterial infections are a severe threat to public health owing to their high risk of fatality. Noticeably, the premature degradation and undeveloped imaging ability of antibiotics still remain challenging. Herein, a selenium nanosystem in response to a bacteria-infected microenvironment is proposed as an antibiotic substitute to detect and inhibit methicillin-resistant Staphylococcus aureus (MRSA) with a combined strategy. Using natural red blood cell membrane (RBCM) and bacteria-responsive gelatin nanoparticles (GNPs), the Ru-Se@GNP-RBCM nanosystem was constructed for effective delivery of Ru-complex-modified selenium nanoparticles (Ru-Se NPs). Taking advantage of natural RBCM, the immune system clearance was reduced and exotoxins were neutralized efficiently. GNPs could be degraded by gelatinase in pathogen-infected areas in situ; therefore, Ru-Se NPs were released to destroy the bacteria cells. Ru-Se NPs with intense fluorescence imaging capability could accurately monitor the infection treatment process. Moreover, excellent in vivo bacteria elimination and a facilitated wound healing process were confirmed by two kinds of MRSA-infected mice models. Overall, the above advantages proved that the prepared nanosystem is a promising antibiotic alternative to combat the ever-threatening multidrug-resistant bacteria.


Subject(s)
Biomimetics , Drug Resistance, Multiple, Bacterial/drug effects , Nanoparticles/chemistry , Selenium/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Infections , Biocompatible Materials/pharmacology , Cattle , Cell Membrane Permeability/drug effects , Disease Models, Animal , Drug Liberation , Endocytosis/drug effects , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Escherichia coli/drug effects , Escherichia coli/ultrastructure , Fluorescence , Gelatin/chemistry , Immune Evasion/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Mice, Inbred BALB C , Microbial Sensitivity Tests , Nanoparticles/ultrastructure , Reactive Oxygen Species/metabolism
14.
PLoS One ; 14(11): e0224816, 2019.
Article in English | MEDLINE | ID: mdl-31730663

ABSTRACT

Two membrane-intercalating conjugated oligoelectrolytes (COEs), namely COE-D8 and COE-S6, were combined to achieve enhanced antimicrobial efficacy. COE-D8 has a shorter molecular length than COE-S6 and is typical of effective antimicrobial COE molecules, presumably due to its prominent membrane disrupting function. In contrast, COE-D6 exhibits lower efficacy against bacteria and lower toxicity toward mammalian cells. Surprisingly, after supplementing 8 µM COE-S6, the minimum inhibitory concentration (MIC) of COE-D8 against methicillin-resistant Staphylococcus aureus (MRSA) was improved 8-fold, from 0.5 µM to 0.063 µM (0.050 µg mL-1). No increased toxicity toward mammalian cells was observed by the combination of COEs, as indicated by cytotoxicity measurements using the 3T3 cell line. Indeed, there is an extended ratio between the half maximal inhibitory concentration based on 3T3 cells to MIC against MRSA from 12 to greater than 256. Biophysical experiments using liposome models suggest that COE-S6 promotes the interactions between COE-D8 and lipid bilayers, which is in agreement with damages of cellular permeability and morphology, as observed by confocal microscopy and scanning electron microscopy. The application of a combined mixture of COEs further demonstrates their promising potential as a new class of antimicrobial agents with high efficacy and selectivity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Electrolytes/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , 3T3 Cells , Adult , Animals , Anti-Bacterial Agents/chemistry , Biophysical Phenomena , Cell Death/drug effects , Drug Synergism , Electrolytes/chemistry , Hep G2 Cells , Humans , Male , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Mice , Microbial Sensitivity Tests
15.
Small ; 15(46): e1902647, 2019 11.
Article in English | MEDLINE | ID: mdl-31614073

ABSTRACT

Integrating multiple strategies of antibacterial mechanisms into one has been proven to have tremendous promise for improving antimicrobial efficiency. Hence, dual-valent platinum nanoparticles (dvPtNPs) with a zero-valent platinum core (Pt0 ) and bi-valent platinum shell (Pt2+ ions), combining photothermal and photodynamic therapy, together with "chemotherapy," emerge as spatiotemporally light-activatable platinum nano-antibiotics. Under near-infrared (NIR) exposure, the multiple antibacterial modes of dvPtNPs are triggered. The Pt0 core reveals significant hyperthermia via effective photothermal conversion while an immediate release of chemotherapeutic Pt2+ ions occurs through hyperthermia-initiated destabilization of metallic interactions, together with reactive oxygen species (ROS) level increase, thereby resulting in synergistic antibacterial effects. The precise cooperative effects between photothermal, photodynamic, and Pt2+ antibacterial effects are achieved on both Gram-negative Escherichia coli and Gram-positive methicillin-resistant Staphylococcus aureus, where bacterial viability and colony-forming units are significantly reduced. Moreover, similar results are observed in mice subcutaneous abscess models. Significantly, after NIR treatment, dvPtNP exhibits a more robust bacteria-killing efficiency than other PtNP groups, owing to its integration of dramatic damage to the bacterial membrane and DNA, and alteration to ATP and ROS metabolism. This study broadens the avenues for designing and synthesizing antibacterial materials with higher efficiency.


Subject(s)
Anti-Bacterial Agents/pharmacology , Light , Nanoparticles/chemistry , Platinum/pharmacology , Animals , Escherichia coli/drug effects , Escherichia coli/ultrastructure , Female , Metal Nanoparticles/ultrastructure , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , NIH 3T3 Cells
16.
Int J Nanomedicine ; 14: 7975-7985, 2019.
Article in English | MEDLINE | ID: mdl-31632012

ABSTRACT

BACKGROUND: Multidrug-resistant bacteria such as extended-spectrum beta-lactamase (ESBL), Enterobacteriaceae, and methicillin-resistant Staphylococcus aureus (MRSA) pose a challenge to the human health care system. MRSA is among the major causes of hospital-acquired and community infections. METHODS: Therefore, in the present study, we evaluated the antibacterial activity of silver nanoparticles synthesized by Fusarium oxysporum (AgNPbio) in combination with simvastatin against reference and multidrug-resistant bacterial strains. RESULTS: Simvastatin showed a minimal inhibitory concentration (MIC) ranging from 0.062 to 0.25 mg mL-1 against MRSA. AgNPbio with a size of 77.68± 33.95 nm and zeta potential -34.6 ± 12.7 mV showed an MIC of 0.212 mg mL-1 against S. aureus including MRSA strains. The checkerboard assay and time-kill curves exhibited a synergistic effect of the simvastatin-AgNPbio combination on antibacterial activity against MRSA strains. The combination of simvastatin and AgNPbio demonstrated antibacterial activity against Escherichia coli producing ESBL. Scanning electron microscopy showed the formation of cell surface protrusions after treatment with AgNPbio and the formation of a large amorphous mass after treatment with simvastatin, both in MRSA. CONCLUSION: Our results indicate that the combination of AgNPbio and simvastatin could be a great future alternative in the control of bacterial infections, where, when combined with simvastatin, smaller doses of AgNPbio are required, with the same antibacterial activity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli/drug effects , Fusarium/metabolism , Metal Nanoparticles/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Silver/pharmacology , Simvastatin/pharmacology , Cell Death/drug effects , Drug Synergism , Erythrocytes/drug effects , Fusarium/drug effects , Fusarium/ultrastructure , Humans , Metal Nanoparticles/ultrastructure , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Microbial Sensitivity Tests
17.
Acta Biomater ; 96: 537-546, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31302297

ABSTRACT

This work describes a novel strategy to combat methicillin-resistant Staphylococcus aureus (MRSA) via the reactivation of inert antibiotics. This strategy exploits a multifunctional system consisting of bioactive glass-ceramic microparticles with antibacterial properties combined with various antibiotics to kill MRSA. Specifically, sol-gel derived silver-doped bioactive glass-ceramic microparticles (Ag-BG) combined with antibiotics that MRSA resists such as oxacillin or fosfomycin, significantly decreased the viability of MRSA. Ag-BG also potentiated the activity of vancomycin on static bacteria, which are typically resistant to this antibiotic. Notably, the synergistic activity is restricted to cell-envelope acting antibiotics as Ag-BG supplementation did not increase the efficacy of gentamicin. Bacteria viability assays and electron microscopy images demonstrate that Ag-BG synergizes to restore antibacterial activity to antibiotics that MRSA resists. The low cytotoxicity previously studied against oral bacteria, together with the known regenerative properties presented in previous studies, and the unique antibacterial properties observed in this work when they are combined with antibiotics, make this multifunctional system a promising approach for healing infected tissue. STATEMENT OF SIGNIFICANCE: This study addresses a very significant issue in the field of antibiotic resistance presenting an innovative way to clear MRSA, by utilizing bioactive glass-ceramic microparticles in combination with antibiotics. Multifunctional glass-ceramic microparticles doped with silver ions (Ag-BG) have been previously observed to exhibit bioactive and antibacterial properties. In this study Ag-BG microparticles were observed to synergize with antibiotics restoring their sensitivity against MRSA. This research work presents a novel approach to resurrect ineffective antibiotics and render them effective against MRSA. Cytotoxicity to eukaryotic cells is not anticipated, as it has been previously observed that these microparticles can trigger hard and soft dental tissue regeneration, when they are utilized in certain concentrations. This study opens a new avenue in the treatment of multidrug resistance bacteria.


Subject(s)
Anti-Bacterial Agents , Ceramics , Glass/chemistry , Methicillin-Resistant Staphylococcus aureus/growth & development , Silver , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Ceramics/chemistry , Ceramics/pharmacology , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Silver/chemistry , Silver/pharmacology
18.
Proc Natl Acad Sci U S A ; 116(33): 16529-16534, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31358625

ABSTRACT

Treatment of Staphylococcus aureus infections is complicated by the development of antibiotic tolerance, a consequence of the ability of S. aureus to enter into a nongrowing, dormant state in which the organisms are referred to as persisters. We report that the clinically approved anthelmintic agent bithionol kills methicillin-resistant S. aureus (MRSA) persister cells, which correlates with its ability to disrupt the integrity of Gram-positive bacterial membranes. Critically, bithionol exhibits significant selectivity for bacterial compared with mammalian cell membranes. All-atom molecular dynamics (MD) simulations demonstrate that the selectivity of bithionol for bacterial membranes correlates with its ability to penetrate and embed in bacterial-mimic lipid bilayers, but not in cholesterol-rich mammalian-mimic lipid bilayers. In addition to causing rapid membrane permeabilization, the insertion of bithionol increases membrane fluidity. By using bithionol and nTZDpa (another membrane-active antimicrobial agent), as well as analogs of these compounds, we show that the activity of membrane-active compounds against MRSA persisters positively correlates with their ability to increase membrane fluidity, thereby establishing an accurate biophysical indicator for estimating antipersister potency. Finally, we demonstrate that, in combination with gentamicin, bithionol effectively reduces bacterial burdens in a mouse model of chronic deep-seated MRSA infection. This work highlights the potential repurposing of bithionol as an antipersister therapeutic agent.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Membrane/drug effects , Drug Repositioning , Methicillin-Resistant Staphylococcus aureus/drug effects , Animals , Bithionol/pharmacology , Cell Membrane Permeability/drug effects , Cholesterol/chemistry , Disease Models, Animal , Drug Synergism , Gentamicins/pharmacology , Lipid Bilayers/chemistry , Membrane Fluidity/drug effects , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Molecular Dynamics Simulation , Phosphatidylcholines/chemistry , Structure-Activity Relationship , Unilamellar Liposomes
19.
J Med Microbiol ; 68(8): 1129-1136, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31241446

ABSTRACT

PURPOSE: Staphylococcus aureus causes a wide range of infections, such as endocarditis, pneumonia, osteomyelitis, skin and soft tissue infections, and implant/in-dwelling device-related infections. S. aureus poses a significant challenge to clinicians because of its ability to rapidly acquire multi-drug resistance and quickly progress into a recurrent, chronic infection by biofilm formation. Levonadifloxacin (WCK 771) is a novel broad-spectrum antibacterial agent (it recently completed a phase 3 trial in India) with a differentiated mechanism of action involving high affinity to staphylococcal DNA gyrase, and is active against multi-drug-resistant (MDR) S. aureus, including those that are resistant to quinolones. The present study investigated the bactericidal activity of levonadifloxacin against biofilm-embedded S. aureus clinical isolates in comparison with other anti-S. aureus drugs. METHODOLOGY: The bactericidal activity of levonadifloxacin and comparator drugs such as vancomycin, linezolid and daptomycin was evaluated against planktonic and biofilm-encapsulated recent methicillin- and quinolone-resistant S. aureus clinical isolates using time-kill, biofilm eradication and scanning electron microscopy analysis. RESULTS: Levonadifloxacin displayed a consistent ≥90 % bacterial kill rate against biofilm-embedded organisms, while vancomycin and linezolid displayed variable activity and daptomycin did not show any activity. Scanning electron microscopy images further confirmed the efficacy of levonadifloxacin against biofilm, showing the disruption of biofilm structure and a corresponding reduction in the viable bacterial count. CONCLUSION: These results show that levonadifloxacin has an improved bactericidal effect on biofilm-embedded quinolone-resistant S. aureus and meticillin-resistant S. aureus, and that it can be a promising treatment option for such infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , Fluoroquinolones/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Biofilms/growth & development , Humans , Methicillin/pharmacology , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/physiology , Methicillin-Resistant Staphylococcus aureus/ultrastructure , Microbial Sensitivity Tests , Microbial Viability/drug effects , Quinolones/pharmacology , Staphylococcal Infections/microbiology , Time Factors
20.
Appl Biochem Biotechnol ; 189(4): 1291-1303, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31254228

ABSTRACT

With growing concern about the possible risks and side effects of antibiotic drugs, more and more natural products with antibacterial activity are studied as the substitutes. In this paper, the antibacterial activity of hydroquinone and arbutin in Ainsliaea bonatii was investigated, which both displayed relatively strong antibacterial activity against Staphylococcus aureus (SA), methicillin-resistant S. aureus (MRSA), and extended spectrum ß-lactamase S. aureus (ESBL-SA). The antibacterial mechanism of hydroquinone had been explored by scanning electron microscopy (SEM), alkaline phosphatase (AKP), and bacterial extracellular protein leakage. Results showed that hydroquinone could destroy the bacterial cell wall and membrane, increase permeability, lead leakage of intracellular substance affect synthesis of protein, and influence expression of genes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Arbutin/pharmacology , Hydroquinones/pharmacology , Lamiaceae/chemistry , Methicillin-Resistant Staphylococcus aureus/metabolism , Anti-Bacterial Agents/chemistry , Arbutin/chemistry , Hydroquinones/chemistry , Methicillin-Resistant Staphylococcus aureus/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...