Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.419
Filter
1.
Methods Mol Biol ; 2833: 57-64, 2024.
Article in English | MEDLINE | ID: mdl-38949701

ABSTRACT

The use of animal models is still widespread in science but there is a movement away from this manner of experimentation. One option approved by the FDA for human-like studies is the hollow fiber bioreactor (HFS). HFSs are highly controllable, self-contained systems that allow for the modeling of individual tissues and disease phenotypes. Oxygen, drug concentration & half-life, and immune cell invasion are all scalable to human and veterinary conditions using a HFS. There are drawbacks to the systems including cost and contamination so the use of these systems must be carefully managed.With these limitations in mind, the scope of the technology is great. Antimicrobial susceptibility testing (AST) is possible with greater accuracy and clinical validity than classical in vitro techniques making minimal inhibitory concentration (MIC) data generated on the bench more translatable to the clinic.In this chapter, we will outline the background of the HFS and some typical uses.


Subject(s)
Bioreactors , Microbial Sensitivity Tests , Humans , Microbial Sensitivity Tests/methods , Animals , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects
2.
Iran J Med Sci ; 49(6): 369-376, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38952643

ABSTRACT

Background: Spontaneous bacterial peritonitis (SBP) is a fatal complication of ascites fluid infection. The causes of SBP in children differ from those in adults, and these bacteria are frequently resistant to antibiotics. Therefore, this study investigated the clinical findings, bacterial etiology, and antimicrobial resistance in children with SBP. Methods: This study was conducted on all new pediatric ascites patients, who were admitted to the Department of Pediatric Gastroenterology, Namazi Hospital, affiliated with Shiraz University of Medical Sciences (Shiraz, Iran) from 2021 to 2022. Required data such as demographic information, and clinical information such as complete blood count (CBC), erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), Gram staining, blood culture by Automated Blood Culture System (BACTEC), and antibiogram of ascites fluids by disc diffusion method were all collected. Finally, the data were statistically analyzed using SPSS Software (version 26). Besides, the t test, Fisher's exact, Mann-Whitney, and Chi square tests were used for data analysis. In all tests, P≤0.05 was considered statistically significant. Results: The present study examined 62 children with ascites of which 18 (29%) had SBP. The median (IQR) age was 2.5 (8.1) years. Thirty-four (54.8%) of the participants were girls. Abdominal pain was the most common clinical manifestation in patients (54%), and there was a significant association between abdominal pain and SBP (P=0.02). In 12 positive ascites fluid cultures, coagulase-negative staphylococci had the highest frequency (25%), followed by Escherichia coli (16.7%). Third-generation cephalosporins had a 25% sensitivity in the total positive cultures. This sensitivity was 33.3% for Gram-negative cultures and 16.6% for Gram-positive cultures. Conclusion: Although third-generation cephalosporins are recommended as the primary antibiotic for the empirical treatment of SBP, the present study found high bacterial resistance. Finally, empirical therapy should be tailored to each region's bacterial resistance features.


Subject(s)
Anti-Bacterial Agents , Peritonitis , Tertiary Care Centers , Humans , Peritonitis/drug therapy , Peritonitis/microbiology , Child , Female , Male , Iran , Child, Preschool , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Tertiary Care Centers/statistics & numerical data , Tertiary Care Centers/organization & administration , Infant , Adolescent , Drug Resistance, Bacterial/drug effects , Ascites/drug therapy , Bacterial Infections/drug therapy , Microbial Sensitivity Tests/methods , Microbial Sensitivity Tests/statistics & numerical data
3.
Aust J Gen Pract ; 53(7): 499-503, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38959520

ABSTRACT

BACKGROUND AND OBJECTIVES: There were 82.4 million new gonorrhoea cases worldwide in 2020. Dual treatment with ceftriaxone or cefixime and azithromycin or doxycycline is currently recommended for gonorrhoea in Indonesia. However, reduced susceptibility and resistance to cephalosporins and azithromycin are increasing. We evaluated the susceptibility pattern of Neisseria gonorrhoeae to cefixime, ceftriaxone, azithromycin and doxycycline. METHOD: N. gonorrhoeae isolates were obtained from 19 male participants with clinically and laboratory-confirmed gonorrhoea. Antibiotic susceptibility testing was conducted by disc diffusion and interpreted according to Clinical and Laboratory Standards Institute and Centers for Disease Control and Prevention criteria. RESULTS: Reduced susceptibility or resistance was observed against doxycycline in 19 isolates (100%), cefixime in six (31.6%), ceftriaxone in three (15.8%) and azithromycin in zero (0%) isolates. DISCUSSION: A dual treatment regimen with ceftriaxone and azithromycin can still be recommended as first-line therapy for gonorrhoea in Indonesia. Antibiotic susceptibility surveillance of N. gonorrhoeae should be routinely conducted.


Subject(s)
Anti-Bacterial Agents , Azithromycin , Ceftriaxone , Doxycycline , Gonorrhea , Microbial Sensitivity Tests , Neisseria gonorrhoeae , Humans , Indonesia , Neisseria gonorrhoeae/drug effects , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Gonorrhea/drug therapy , Male , Microbial Sensitivity Tests/methods , Azithromycin/therapeutic use , Doxycycline/therapeutic use , Ceftriaxone/therapeutic use , Ceftriaxone/pharmacology , Adult , Cefixime/therapeutic use , Cefixime/pharmacology , Primary Health Care/statistics & numerical data , Drug Resistance, Bacterial/drug effects , Drug Therapy, Combination/methods
4.
Med Sci Monit ; 30: e945583, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38967286

ABSTRACT

Antimicrobial resistance and the associated morbidity and mortality from untreatable common infectious organisms is an increasing threat to global public health. In 2019, the Antimicrobial Resistance Collaborators identified that antimicrobial resistance was directly responsible for up to 1.27 million deaths worldwide and was associated with up to 4.95 million deaths, with low-income and middle-income countries being the most severely affected. In 2019, before the COVID-19 pandemic began, they predicted that antimicrobial resistance could result in 10 million deaths per year by 2050, overtaking cancer as a leading cause of death worldwide. Therefore, there is an urgent need for new approaches to antimicrobial treatment. In June 2024, the findings from researchers at the Ineos Oxford Institute for Antimicrobial Research (IOI) and the Oxford University Department of Pharmacology in the UK reported the use of a small molecule that can work alongside antibiotics to suppress the development of antimicrobial resistance in bacteria. The SOS inhibitor molecule has been called OXF-077. This editorial aims to highlight the global threats from increasing antimicrobial resistance and the urgent need for new molecules that function through novel mechanisms of action, including molecular antimicrobial adjuvants.


Subject(s)
Drug Development , Humans , Drug Development/methods , Anti-Infective Agents/pharmacology , Anti-Infective Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Drug Resistance, Microbial , Drug Resistance, Bacterial/drug effects , COVID-19 , Global Health , SARS-CoV-2/drug effects
5.
Methods Mol Biol ; 2833: 129-143, 2024.
Article in English | MEDLINE | ID: mdl-38949707

ABSTRACT

Antibiotic resistance is a global challenge likely to cost trillions of dollars in excess costs in the health system and more importantly, millions of lives every year. A major driver of resistance is the absence of susceptibility testing at the time a healthcare worker needs to prescribe an antimicrobial. The effect is that many prescriptions are unintentionally wasted and expose mutable organisms to antibiotics increasing the risk of resistance emerging. Often simplistic solutions are applied to this growing issue, such as a naïve drive to increase the speed of drug susceptibility testing. This puts a spotlight on a technological solution and there is a multiplicity of such candidate DST tests in development. Yet, if we do not define the necessary information and the speed at which it needs to be available in the clinical decision-making progress as well as the necessary integration into clinical pathways, then little progress will be made. In this chapter, we place the technological challenge in a clinical and systems context. Further, we will review the landscape of some promising technologies that are emerging and attempt to place them in the clinic where they will have to succeed.


Subject(s)
Anti-Bacterial Agents , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests/methods , Humans , Drug Resistance, Bacterial/drug effects , Bacteria/drug effects
6.
Int J Mol Sci ; 25(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38928191

ABSTRACT

Staphylococcus aureus bacteremia continues to be associated with significant morbidity and mortality, despite improvements in diagnostics and management. Persistent infections pose a major challenge to clinicians and have been consistently shown to increase the risk of mortality and other infectious complications. S. aureus, while typically not considered an intracellular pathogen, has been proven to utilize an intracellular niche, through several phenotypes including small colony variants, as a means for survival that has been linked to chronic, persistent, and recurrent infections. This intracellular persistence allows for protection from the host immune system and leads to reduced antibiotic efficacy through a variety of mechanisms. These include antimicrobial resistance, tolerance, and/or persistence in S. aureus that contribute to persistent bacteremia. This review will discuss the challenges associated with treating these complicated infections and the various methods that S. aureus uses to persist within the intracellular space.


Subject(s)
Anti-Bacterial Agents , Bacteremia , Staphylococcal Infections , Staphylococcus aureus , Bacteremia/drug therapy , Bacteremia/microbiology , Humans , Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Animals , Drug Resistance, Bacterial/drug effects
7.
J Hazard Mater ; 475: 134817, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38878444

ABSTRACT

Microplastics and antibiotics coexist in aquatic environments, especially in freshwater aquaculture areas. However, as the second largest production of polyvinyl chloride (PVC) in the world, the effects of co-exposure to microplastics particles and antibiotics on changes in antibiotic resistance gene (ARG) profiles and the microbial community structure of aquatic organism gut microorganisms are poorly understood. Therefore, in this study, carp (Cyprinus carpio) were exposed to single or combined PVC microplastic contamination and oxytetracycline (OTC) or sulfamethazine (SMZ) for 8 weeks. PVC microplastics can enrich potential pathogenic bacteria, such as Enterobacter and Acinetobacter, among intestinal microorganisms. The presence of PVC microplastics enhanced the selective enrichment and dissemination risk of ARGs. PVC microplastics combined with OTC (OPVC) treatment significantly increased the abundance of tetracycline resistance genes (1.40-fold) compared with that in the OTC exposure treatment, revealing an obvious co-selection effect. However, compared with those in the control group, the total abundance of ARGs and MGEs in the OPVC treatment groups were significantly lower, which was correlated with the reduced abundances of the potential host Enterobacter. Overall, our results emphasized the diffusion and spread of ARGs are more influenced by PVC microplastics than by antibiotics, which may lead to antibiotic resistance in aquaculture.


Subject(s)
Anti-Bacterial Agents , Carps , Microplastics , Oxytetracycline , Polyvinyl Chloride , Water Pollutants, Chemical , Animals , Microplastics/toxicity , Water Pollutants, Chemical/toxicity , Oxytetracycline/toxicity , Carps/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Gastrointestinal Microbiome/drug effects , Bacteria/drug effects , Bacteria/genetics , Sulfamethazine/toxicity , Genes, Bacterial/drug effects , Drug Resistance, Microbial/genetics , Drug Resistance, Bacterial/genetics , Drug Resistance, Bacterial/drug effects
8.
PLoS Biol ; 22(6): e3002694, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38900845

ABSTRACT

Fungi and bacteria coexist in many polymicrobial communities, yet the molecular basis of their interactions remains poorly understood. Here, we show that the fungus Candida albicans sequesters essential magnesium ions from the bacterium Pseudomonas aeruginosa. To counteract fungal Mg2+ sequestration, P. aeruginosa expresses the Mg2+ transporter MgtA when Mg2+ levels are low. Thus, loss of MgtA specifically impairs P. aeruginosa in co-culture with C. albicans, but fitness can be restored by supplementing Mg2+. Using a panel of fungi and bacteria, we show that Mg2+ sequestration is a general mechanism of fungal antagonism against gram-negative bacteria. Mg2+ limitation enhances bacterial resistance to polymyxin antibiotics like colistin, which target gram-negative bacterial membranes. Indeed, experimental evolution reveals that P. aeruginosa evolves C. albicans-dependent colistin resistance via non-canonical means; antifungal treatment renders resistant bacteria colistin-sensitive. Our work suggests that fungal-bacterial competition could profoundly impact polymicrobial infection treatment with antibiotics of last resort.


Subject(s)
Anti-Bacterial Agents , Candida albicans , Colistin , Magnesium , Pseudomonas aeruginosa , Magnesium/pharmacology , Magnesium/metabolism , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/pharmacology , Candida albicans/drug effects , Candida albicans/metabolism , Colistin/pharmacology , Microbial Sensitivity Tests , Polymyxins/pharmacology , Drug Resistance, Bacterial/drug effects , Microbial Interactions/drug effects
9.
Molecules ; 29(11)2024 May 25.
Article in English | MEDLINE | ID: mdl-38893378

ABSTRACT

Metabolic reprogramming mediates antibiotic efficacy. However, metabolic adaptation of microbes evolving from antibiotic sensitivity to resistance remains undefined. Therefore, untargeted metabolomics was conducted to unveil relevant metabolic reprogramming and potential intervention targets involved in gentamicin resistance. In total, 61 metabolites and 52 metabolic pathways were significantly altered in gentamicin-resistant E. coli. Notably, the metabolic reprogramming was characterized by decreases in most metabolites involved in carbohydrate and amino acid metabolism, and accumulation of building blocks for nucleotide synthesis in gentamicin-resistant E. coli. Meanwhile, fatty acid metabolism and glycerolipid metabolism were also significantly altered in gentamicin-resistant E. coli. Additionally, glycerol, glycerol-3-phosphate, palmitoleate, and oleate were separately defined as the potential biomarkers for identifying gentamicin resistance in E. coli. Moreover, palmitoleate and oleate could attenuate or even abolished killing effects of gentamicin on E. coli, and separately increased the minimum inhibitory concentration of gentamicin against E. coli by 2 and 4 times. Furthermore, palmitoleate and oleate separately decreased intracellular gentamicin contents, and abolished gentamicin-induced accumulation of reactive oxygen species, indicating involvement of gentamicin metabolism and redox homeostasis in palmitoleate/oleate-promoted gentamicin resistance in E. coli. This study identifies the metabolic reprogramming, potential biomarkers and intervention targets related to gentamicin resistance in bacteria.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Escherichia coli , Fatty Acids, Monounsaturated , Gentamicins , Oleic Acid , Gentamicins/pharmacology , Gentamicins/metabolism , Escherichia coli/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Oleic Acid/metabolism , Oleic Acid/pharmacology , Drug Resistance, Bacterial/drug effects , Anti-Bacterial Agents/pharmacology , Fatty Acids, Monounsaturated/metabolism , Fatty Acids, Monounsaturated/pharmacology , Microbial Sensitivity Tests , Metabolomics/methods , Metabolic Networks and Pathways/drug effects , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects
10.
PeerJ ; 12: e17463, 2024.
Article in English | MEDLINE | ID: mdl-38827315

ABSTRACT

Background: The use of antimicrobials to treat food animals may result in antimicrobial residues in foodstuffs of animal origin. The European Medicines Association (EMA) and World Health Organization (WHO) define safe antimicrobial concentrations in food based on acceptable daily intakes (ADIs). It is unknown if ADI doses of antimicrobials in food could influence the antimicrobial susceptibility of human-associated bacteria. Objectives: This aim of this study was to evaluate if the consumption of ADI doses of erythromycin could select for erythromycin resistance in a Galleria mellonella model of Streptococcus pneumoniae infection. Methods: A chronic model of S. pneumoniae infection in G. mellonella larvae was used for the experiment. Inoculation of larvae with S. pneumoniae was followed by injections of erythromycin ADI doses (0.0875 and 0.012 µg/ml according to EMA and WHO, respectively). Isolation of S. pneumoniae colonies was then performed on selective agar plates. Minimum inhibitory concentrations (MICs) of resistant colonies were measured, and whole genome sequencing (WGS) was performed followed by variant calling to determine the genetic modifications. Results: Exposure to single doses of both EMA and WHO ADI doses of erythromycin resulted in the emergence of erythromycin resistance in S. pneumoniae. Emergent resistance to erythromycin was associated with a mutation in rplA, which codes for the L1 ribosomal protein and has been linked to macrolide resistance in previous studies. Conclusion: In our in vivo model, even single doses of erythromycin that are classified as acceptable by the WHO and EMA induced significant increases in erythromycin MICs in S. pneumoniae. These results suggest the need to include the induction of antimicrobial resistance (AMR) as a significant criterion for determining ADIs.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Erythromycin , Larva , Microbial Sensitivity Tests , Moths , Streptococcus pneumoniae , Erythromycin/pharmacology , Animals , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/genetics , Anti-Bacterial Agents/pharmacology , Moths/microbiology , Moths/drug effects , Drug Resistance, Bacterial/genetics , Drug Resistance, Bacterial/drug effects , Larva/microbiology , Larva/drug effects , Pneumococcal Infections/drug therapy , Pneumococcal Infections/microbiology , Disease Models, Animal , Humans
11.
Nat Commun ; 15(1): 4901, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851779

ABSTRACT

Antimicrobial resistance remains a significant global threat, driving up mortality rates worldwide. Ribosomally synthesized and post-translationally modified peptides have emerged as a promising source of novel peptide antibiotics due to their diverse chemical structures. Here, we report the discovery of new aminovinyl-(methyl)cysteine (Avi(Me)Cys)-containing peptide antibiotics through a synergistic approach combining biosynthetic rule-based omics mining and heterologous expression. We first bioinformatically identify 1172 RiPP biosynthetic gene clusters (BGCs) responsible for Avi(Me)Cys-containing peptides formation from a vast pool of over 50,000 bacterial genomes. Subsequently, we successfully establish the connection between three identified BGCs and the biosynthesis of five peptide antibiotics via biosynthetic rule-guided metabolic analysis. Notably, we discover a class V lanthipeptide, massatide A, which displays excellent activity against gram-positive pathogens, including drug-resistant clinical isolates like linezolid-resistant S. aureus and methicillin-resistant S. aureus, with a minimum inhibitory concentration of 0.25 µg/mL. The remarkable performance of massatide A in an animal infection model, coupled with a relatively low risk of resistance and favorable safety profile, positions it as a promising candidate for antibiotic development. Our study highlights the potential of Avi(Me)Cys-containing peptides in expanding the arsenal of antibiotics against multi-drug-resistant bacteria, offering promising drug leads in the ongoing battle against infectious diseases.


Subject(s)
Anti-Bacterial Agents , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Peptides, Cyclic/pharmacology , Peptides, Cyclic/chemistry , Humans , Multigene Family , Mice , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/genetics , Antimicrobial Peptides/metabolism , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Drug Resistance, Bacterial/genetics , Drug Resistance, Bacterial/drug effects , Genome, Bacterial/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Computational Biology/methods , Cysteine/metabolism , Cysteine/chemistry
12.
PLoS One ; 19(6): e0304980, 2024.
Article in English | MEDLINE | ID: mdl-38905247

ABSTRACT

BACKGROUND: Antibiotic resistance genes (ARGs) transfer rapidly among bacterial species all over the world contributing to the aggravation of antibiotic resistance crisis. Antibiotics at sub-inhibitory concentration induce horizontal gene transfer (HRT) between bacteria, especially through conjugation. The role of common non-antibiotic pharmaceuticals in the market in disseminating antibiotic resistance is not well studied. OBJECTIVES: In this work, we indicated the effect of some commonly used non-antibiotic pharmaceuticals including antiemetic (metoclopramide HCl) and antispasmodics (hyoscine butyl bromide and tiemonium methyl sulfate) on the plasmid-mediated conjugal transfer of antibiotic resistance genes between pathogenic E. coli in the gastric intestinal tract (GIT). METHODS: Broth microdilution assay was used to test the antibacterial activity of the tested non-antibiotic pharmaceuticals. A conjugation mating system was applied in presence of the studied non-antibiotic pharmaceuticals to test their effect on conjugal transfer frequency. Plasmid extraction and PCR were performed to confirm the conjugation process. Transmission electron microscopy (TEM) was used for imaging the effect of non-antibiotic pharmaceuticals on bacterial cells. RESULTS: No antibacterial activity was reported for the used non-antibiotic pharmaceuticals. Plasmid-mediated conjugal transfer between isolates was induced by metoclopramide HCl but suppressed by hyoscine butyl bromide. Tiemonium methylsulfate slightly promoted conjugal transfer. Aggregation between cells and periplasmic bridges was clear in the case of metoclopramide HCl while in presence of hyoscine butyl bromide little affinity was observed. CONCLUSION: This study indicates the contribution of non-antibiotic pharmaceuticals to the dissemination and evolution of antibiotic resistance at the community level. Metoclopramide HCl showed an important role in the spread of antibiotic resistance.


Subject(s)
Escherichia coli , Gene Transfer, Horizontal , Plasmids , Escherichia coli/drug effects , Escherichia coli/genetics , Plasmids/genetics , Metoclopramide/pharmacology , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Drug Resistance, Bacterial/drug effects , Conjugation, Genetic , Drug Resistance, Microbial/genetics , Drug Resistance, Microbial/drug effects
13.
J Hazard Mater ; 474: 134697, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38823102

ABSTRACT

Airborne pathogens severely threaten public health worldwide. Air disinfection is essential to ensure public health. However, excessive use of disinfectants may endanger environmental and ecological security due to the residual disinfectants and their by-products. This study systematically evaluated disinfection efficiency, induction of multidrug resistance, and the underlying mechanisms of disinfectants (NaClO and H2O2) on airborne bacteria. The results showed that airborne bacteria were effectively inactivated by atomized NaClO (>160 µg/L) and H2O2 (>320 µg/L) after 15 min. However, some bacteria still survived after disinfection by atomized NaClO (0-80 µg/L) and H2O2 (0-160 µg/L), and they exhibited significant increases in antibiotic resistance. The whole-genome sequencing of the resistant bacteria revealed distinct mutations that were responsible for both antibiotic resistance and virulence. This study also provided evidences and insights into possible mechanisms underlying the induction of antibiotic resistance by air disinfection, which involved intracellular reactive oxygen species formation, oxidative stress responses, alterations in bacterial membranes, activation of efflux pumps, and the thickening of biofilms. The present results also shed light on the role of air disinfection in inducing antibiotic resistance, which could be a crucial factor contributing to the global spread of antibiotic resistance through the air.


Subject(s)
Bacteria , Disinfectants , Disinfection , Hydrogen Peroxide , Hydrogen Peroxide/pharmacology , Disinfectants/pharmacology , Disinfection/methods , Bacteria/drug effects , Bacteria/genetics , Air Microbiology , Biofilms/drug effects , Sodium Hypochlorite/pharmacology , Drug Resistance, Bacterial/genetics , Drug Resistance, Bacterial/drug effects , Anti-Bacterial Agents/pharmacology , Chlorine/pharmacology , Reactive Oxygen Species/metabolism
14.
J Colloid Interface Sci ; 671: 751-769, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38824748

ABSTRACT

Chemotherapy and surgery stand as primary cancer treatments, yet the unique traits of the tumor microenvironment hinder their effectiveness. The natural compound epigallocatechin gallate (EGCG) possesses potent anti-tumor and antibacterial traits. However, the tumor's adaptability to chemotherapy due to its acidic pH and elevated glutathione (GSH) levels, coupled with the challenges posed by drug-resistant bacterial infections post-surgery, impede treatment outcomes. To address these challenges, researchers strive to explore innovative treatment strategies, such as multimodal combination therapy. This study successfully synthesized Cu-EGCG, a metal-polyphenol network, and detailly characterized it by using synchrotron radiation and high-resolution mass spectrometry (HRMS). Through chemodynamic therapy (CDT), photothermal therapy (PTT), and photodynamic therapy (PDT), Cu-EGCG showed robust antitumor and antibacterial effects. Cu+ in Cu-EGCG actively participates in a Fenton-like reaction, generating hydroxyl radicals (·OH) upon exposure to hydrogen peroxide (H2O2) and converting to Cu2+. This Cu2+ interacts with GSH, weakening the oxidative stress response of bacteria and tumor cells. Density functional theory (DFT) calculations verified Cu-EGCG's efficient GSH consumption during its reaction with GSH. Additionally, Cu-EGCG exhibited outstanding photothermal conversion when exposed to 808 nm near-infrared (NIR) radiation and produced singlet oxygen (1O2) upon laser irradiation. In both mouse tumor and wound models, Cu-EGCG showcased remarkable antitumor and antibacterial properties.


Subject(s)
Anti-Bacterial Agents , Antineoplastic Agents , Catechin , Copper , Nanocomposites , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Copper/chemistry , Copper/pharmacology , Nanocomposites/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Animals , Mice , Humans , Catechin/chemistry , Catechin/pharmacology , Catechin/analogs & derivatives , Microbial Sensitivity Tests , Drug Resistance, Bacterial/drug effects , Photochemotherapy , Wound Infection/drug therapy , Wound Infection/pathology , Wound Infection/microbiology , Drug Screening Assays, Antitumor , Staphylococcus aureus/drug effects , Photothermal Therapy , Particle Size , Escherichia coli/drug effects , Cell Survival/drug effects , Cell Line, Tumor , Surface Properties , Cell Proliferation/drug effects
15.
Phys Chem Chem Phys ; 26(24): 17011-17027, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38835320

ABSTRACT

Pseudomonas aeruginosa, a formidable pathogen renowned for its antimicrobial resistance, poses a significant threat to immunocompromised individuals. In this regard, the MexAB-OprM efflux pump acts as a pivotal line of defense by extruding antimicrobials from bacterial cells. The inner membrane homotrimeric protein MexB captures antibiotics and translocates them into the outer membrane OprM channel protein connected through the MexA adaptor protein. Despite extensive efforts, competitive inhibitors targeting the tight (T) protomer of the MexB protein have not received FDA approval for medical use. Over the past few years, allosteric inhibitors have become popular as alternatives to the classical competitive inhibitor-based approach because of their higher specificity, lower dosage, and reduced toxicological effects. Hence, in this study, we unveiled the existence of a transmembrane allosteric binding pocket of MexB inspired by the recent discovery of an important allosteric inhibitor, BDM88855, for the homolog AcrB protein. While repurposing BDM88855 proved ineffective in controlling the MexB loose (L) protomer, our investigation identified a promising alternative: a chlorine-containing variant of DB08385 (2-Cl DB08385 or Variant 1). Molecular dynamics simulations, including binding free energy estimation coupled with heterogeneous dielectric implicit membrane model (implicit-membrane MM/PBSA), interaction entropy (IE) analysis and potential of mean force (PMF) calculation, demonstrated Variant 1's superior binding affinity to the transmembrane pocket, displaying the highest energy barrier in the ligand unbinding process. To elucidate the allosteric crosstalk between the transmembrane and porter domain of MexB, we employed the 'eigenvector centrality' measure in the linear mutual information obtained from the protein correlation network. Notably, this study confirmed the presence of an allosteric transmembrane site in the MexB L protomer. In addition to this, Variant 1 emerged as a potent regulator of allosteric crosstalk, inducing an 'O-L intermediate state' in the MexB L protomer. This induced state might hold the potential to diminish substrate intake into the access pocket, leading to the ineffective efflux of antibiotics.


Subject(s)
Anti-Bacterial Agents , Bacterial Outer Membrane Proteins , Molecular Dynamics Simulation , Pseudomonas aeruginosa , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Allosteric Regulation/drug effects , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/chemistry , Drug Resistance, Bacterial/drug effects
16.
J Biomed Mater Res B Appl Biomater ; 112(7): e35442, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38923117

ABSTRACT

The development of drug-resistant microorganisms is taking a heavy toll on the biomedical world. Clinical infections are costly and becoming increasingly dangerous as bacteria that once responded to standard antibiotic treatment are developing resistance mechanisms that require innovative treatment strategies. Nitric oxide (NO) is a gaseous molecule produced endogenously that has shown potent antibacterial capabilities in numerous research studies. Its multimechanistic antibacterial methods prevent the development of resistance and have shown potential as an alternative to antibiotics. However, there has yet to be a direct comparison study evaluating the antibacterial properties of NO against antibiotic susceptible and antibiotic-resistant clinically isolated bacterial strains. Herein, standardized lab and clinically isolated drug-resistant bacterial strains are compared side-by-side for growth and viability following treatment with NO released from S-nitrosoglutathione (GSNO), an NO donor molecule. Evaluation of growth kinetics revealed complete killing of E. coli lab and clinical strains at 17.5 mM GSNO, though 15 mM displayed >50% killing and significantly reduced metabolic activity, with greater dose dependence for membrane permeability. Clinical P. aeruginosa showed greater susceptibility to GSNO during growth curve studies, but metabolic activity and membrane permeability demonstrated similar effects for 12.5 mM GSNO treatment of lab and clinical strains. MRSA lab and clinical strains exhibited total killing at 17.5 mM treatment, though metabolic activity was decreased, and membrane permeation began at 12.5 mM for both strains. Lastly, both S. epidermidis strains were killed by 15 mM GSNO, with sensitivities in metabolic activity and membrane permeability at 12.5 mM GSNO. The mirrored antibacterial effects seen by the lab and clinical strains of two Gram-negative and two Gram-positive bacteria reveal the translational success of NO as an antibacterial therapy and potential alternative to standard antibiotic treatment.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Nitric Oxide , Nitric Oxide/pharmacology , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli/drug effects , Escherichia coli/growth & development , Humans , S-Nitrosoglutathione/pharmacology , S-Nitrosoglutathione/chemistry , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/chemistry , Drug Resistance, Bacterial/drug effects , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development
17.
PLoS One ; 19(6): e0303271, 2024.
Article in English | MEDLINE | ID: mdl-38924011

ABSTRACT

Antibiotic persistence is a phenomenon, where a small fraction of a bacterial population expresses a phenotypic variation that allows them to survive antibiotic treatment, which is lethal to the rest of the population. These cells are called persisters cells, and their occurrence has been associated with recurrent disease. Streptococcus agalactiae is a human pathobiont, able to cause invasive infections, and recurrent infections have been reported to occur in both newborns and adults. In this study, we demonstrated that S. agalactiae NEM316 can form persister cells when exposed to antibiotics from different classes. The frequency of persister cell formation was dependent on bacterial growth phase and the class of antibiotics. The ability to form persister cells in response to penicillin was shown to be a general trait among different clinical S. agalactiae isolates, independent of sero- and sequence-type. Taken together, this study shows the existence of antibiotic tolerant S. agalactiae persister cells, which may explain why this bacterial species frequently persists after treatment of invasive infection and can be associated with recurrent disease.


Subject(s)
Anti-Bacterial Agents , Microbial Sensitivity Tests , Streptococcus agalactiae , Streptococcus agalactiae/drug effects , Anti-Bacterial Agents/pharmacology , Humans , Streptococcal Infections/microbiology , Streptococcal Infections/drug therapy , Drug Resistance, Bacterial/drug effects , Penicillins/pharmacology
18.
Molecules ; 29(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38893366

ABSTRACT

The development of antimicrobial drugs with novel structures and clear mechanisms of action that are active against drug-resistant bacteria has become an urgent need of safeguarding human health due to the rise of bacterial drug resistance. The discovery of AMPs and the development of amphipathic peptidomimetics have lay the foundation for novel antimicrobial agents to combat drug resistance due to their overall strong antimicrobial activities and unique membrane-active mechanisms. To break the limitation of AMPs, researchers have invested in great endeavors through various approaches in the past years. This review summarized the recent advances including the development of antibacterial small molecule peptidomimetics and peptide-mimic cationic oligomers/polymers, as well as mechanism-of-action studies. As this exciting interdisciplinary field is continuously expanding and growing, we hope this review will benefit researchers in the rational design of novel antimicrobial peptidomimetics in the future.


Subject(s)
Peptidomimetics , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Peptidomimetics/chemical synthesis , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Drug Resistance, Bacterial/drug effects , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Microbial Sensitivity Tests , Bacteria/drug effects
19.
J Mater Chem B ; 12(23): 5645-5660, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38747306

ABSTRACT

The increasing frequency of drug-resistant pathogens poses serious health issues to humans around the globe, leading to the development of new antibacterial agents to conquer drug resistance and bacterial infections. In view of this, we have synthesized a series of bis-naphthalimides to respond to awful drug resistance. Bioactivity assay and structure-activity relationship disclosed that compounds 5d and 5o exhibit potent antibacterial activity against E. faecalis, outperforming the marketed antibiotics. These drug candidates not only inhibit the biofilm formation of E. faecalis but also display rapid bactericidal properties, thus delaying the development of drug resistance within 20 passages. To explore the mechanism of antibacterial activity against E. faecalis, biofunctional examination was carried out which unveiled that 5d and 5o effectively disrupt bacterial cell membranes, causing the leakage of cytoplasmic contents and metabolic activity loss. Concurrently, 5d and 5o effectively intercalate with DNA to block DNA replication, causing the build-up of excessive reactive oxygen species and inhibiting the glutathione activity, ultimately leading to oxidative damage of E. faecalis and cell death. In addition, these compounds readily bind with HSA with a high binding constant, indicating that these drug candidates could be easily delivered to the target site. The above finding manifested that these newly synthesized bis-naphthalimides with multitargeting antibacterial properties offer a new prospect to overcome drug resistance.


Subject(s)
Anti-Bacterial Agents , Enterococcus faecalis , Microbial Sensitivity Tests , Naphthalimides , Enterococcus faecalis/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Naphthalimides/chemistry , Naphthalimides/pharmacology , Humans , Structure-Activity Relationship , Biofilms/drug effects , Drug Resistance, Bacterial/drug effects , Molecular Structure , Cell Death/drug effects
20.
J Hazard Mater ; 473: 134698, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38788587

ABSTRACT

Zero-valent iron (ZVI) has been extensively studied for its capacity to remove various contaminants in the environments. However, whether ZVI affects bacterial resistance to antibiotics has not been fully explored. Herein, it was unexpected that, compared with microscale ZVI (mZVI), nanoscale ZVI (nZVI) facilitated the susceptibility of Pseudomonas aeruginosa (P. aeruginosa) to chloramphenicol (CAP), with a decrease in the minimal inhibitory concentration (MIC) of about 60 %, demonstrating a nanosize-specific effect. nZVI enhanced CAP accumulation in P. aeruginosa via inhibitory effect on efflux pumps activated by MexT, thus conferring the susceptibility of P. aeruginosa to CAP. Circular dichroism spectroscopy revealed that the structure of MexT was changed during the evolution. More importantly, molecular dynamic simulations uncovered that, once the structure of MexT changed, it would be more likely to interact with nZVI, resulting in more serious changes in its secondary structure, which was consistent with the increasing susceptibility of P. aeruginosa to CAP. Collectively, this study elucidated the size-specific effect and the underlying mechanism of ZVI on the bacterial evolution of susceptibility toward antibiotics, highlighting the potentials of nZVI-based technologies on the prevention of bacterial resistance to antibiotics, one of the most important issue for globally public health.


Subject(s)
Anti-Bacterial Agents , Chloramphenicol , Drug Resistance, Bacterial , Iron , Microbial Sensitivity Tests , Pseudomonas aeruginosa , Pseudomonas aeruginosa/drug effects , Chloramphenicol/pharmacology , Chloramphenicol/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Iron/chemistry , Drug Resistance, Bacterial/drug effects , Metal Nanoparticles/chemistry , Molecular Dynamics Simulation , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...