Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
J Biol Chem ; 300(3): 105694, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38301890

ABSTRACT

Bacteriocins, which have narrow-spectrum activity and limited adverse effects, are promising alternatives to antibiotics. In this study, we identified klebicin E (KlebE), a small bacteriocin derived from Klebsiella pneumoniae. KlebE exhibited strong efficacy against multidrug-resistant K. pneumoniae isolates and conferred a significant growth advantage to the producing strain during intraspecies competition. A giant unilamellar vesicle leakage assay demonstrated the unique membrane permeabilization effect of KlebE, suggesting that it is a pore-forming toxin. In addition to a C-terminal toxic domain, KlebE also has a disordered N-terminal domain and a globular central domain. Pulldown assays and soft agar overlay experiments revealed the essential role of the outer membrane porin OmpC and the Ton system in KlebE recognition and cytotoxicity. Strong binding between KlebE and both OmpC and TonB was observed. The TonB-box, a crucial component of the toxin-TonB interaction, was identified as the 7-amino acid sequence (E3ETLTVV9) located in the N-terminal region. Further studies showed that a region near the bottom of the central domain of KlebE plays a primary role in recognizing OmpC, with eight residues surrounding this region identified as essential for KlebE toxicity. Finally, based on the discrepancies in OmpC sequences between the KlebE-resistant and sensitive strains, it was found that the 91st residue of OmpC, an aspartic acid residue, is a key determinant of KlebE toxicity. The identification and characterization of this toxin will facilitate the development of bacteriocin-based therapies targeting multidrug-resistant K. pneumoniae infections.


Subject(s)
Bacteriocins , Klebsiella pneumoniae , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacteriocins/genetics , Bacteriocins/metabolism , Bacteriocins/pharmacology , Bacteriocins/toxicity , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/metabolism , Porins/genetics , Porins/metabolism , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Protein Domains , Drug Resistance, Multiple, Bacterial/drug effects
2.
Sci Rep ; 12(1): 4570, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35301365

ABSTRACT

Bacteriocins and reuterin are promising antimicrobials for application in food, veterinary, and medical sectors. In the light of their high potential for application in hand sanitizer, we investigated the skin toxicity of reuterin, microcin J25, pediocin PA-1, bactofencin A, and nisin Z in vitro using neutral red and LDH release assays on NHEK cells. We determined their skin sensitization potential using the human cell line activation test (h-CLAT). Their skin irritation potential was measured on human epidermal model EpiDerm™. We showed that the viability and membrane integrity of NHEK cells remained unaltered after exposure to bacteriocins and reuterin at concentrations up to 400 µg/mL and 80 mg/mL, respectively. Furthermore, microcin J25 and reuterin showed no skin sensitization at concentrations up to 100 µg/mL and 40 mg/mL, respectively, while pediocin PA-1, bactofencin A, and nisin Z caused sensitization at concentrations higher than 100 µg/mL. Tissue viability was unaffected in presence of bacteriocins and reuterin at concentrations up to 200 µg/mL and 40 mg/mL, respectively, which was confirmed by measuring cytokine IL-1α and IL-8 levels and by histological analysis. In conclusion, the current study provides scientific evidence that some bacteriocins and reuterin, could be safely applied topically as sanitizers at recommended concentrations.


Subject(s)
Bacteriocins , Bacteriocins/metabolism , Bacteriocins/toxicity , Glyceraldehyde/analogs & derivatives , Humans , Propane
3.
Nucleic Acids Res ; 49(3): 1581-1596, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33434265

ABSTRACT

DNA gyrase, a type II topoisomerase found predominantly in bacteria, is the target for a variety of 'poisons', namely natural product toxins (e.g. albicidin, microcin B17) and clinically important synthetic molecules (e.g. fluoroquinolones). Resistance to both groups can be mediated by pentapeptide repeat proteins (PRPs). Despite long-term studies, the mechanism of action of these protective PRPs is not known. We show that a PRP, QnrB1 provides specific protection against fluoroquinolones, which strictly requires ATP hydrolysis by gyrase. QnrB1 binds to the GyrB protein and stimulates ATPase activity of the isolated N-terminal ATPase domain of GyrB (GyrB43). We probed the QnrB1 binding site using site-specific incorporation of a photoreactive amino acid and mapped the crosslinks to the GyrB43 protein. We propose a model in which QnrB1 binding allosterically promotes dissociation of the fluoroquinolone molecule from the cleavage complex.


Subject(s)
Bacterial Proteins/metabolism , DNA Gyrase/metabolism , Topoisomerase II Inhibitors/toxicity , Adenosine Triphosphate/metabolism , Bacteriocins/toxicity , Ciprofloxacin/toxicity , DNA/metabolism , Escherichia coli/enzymology , Hydrolysis , Organic Chemicals/toxicity , Xanthomonas
4.
FEMS Microbiol Rev ; 45(1)2021 01 08.
Article in English | MEDLINE | ID: mdl-32876664

ABSTRACT

In recent decades, bacteriocins have received substantial attention as antimicrobial compounds. Although bacteriocins have been predominantly exploited as food preservatives, they are now receiving increased attention as potential clinical antimicrobials and as possible immune-modulating agents. Infections caused by antibiotic-resistant bacteria have been declared as a global threat to public health. Bacteriocins represent a potential solution to this worldwide threat due to their broad- or narrow-spectrum activity against antibiotic-resistant bacteria. Notably, despite their role in food safety as natural alternatives to chemical preservatives, nisin remains the only bacteriocin legally approved by regulatory agencies as a food preservative. Moreover, insufficient data on the safety and toxicity of bacteriocins represent a barrier against the more widespread use of bacteriocins by the food and medical industry. Here, we focus on the most recent trends relating to the application of bacteriocins, their toxicity and impacts.


Subject(s)
Bacteriocins/toxicity , Anti-Infective Agents/toxicity , Bacteriocins/standards , Drug Development/trends , Drug and Narcotic Control
5.
PLoS One ; 15(10): e0240545, 2020.
Article in English | MEDLINE | ID: mdl-33057351

ABSTRACT

The bacterial strain, EMM-1, was isolated from the rhizosphere of red maize ("Rojo Criollo") and identified as Pseudomonas protegens EMM-1 based on phylogenetic analysis of 16S rDNA, rpoB, rpoD, and gyrB gene sequences. We uncovered genes involved in the production of antimicrobial compounds like 2,4-diacetylphloroglucinol (2,4-DAPG), pyoluteorin, and lectin-like bacteriocins. These antimicrobial compounds are also produced by other fluorescent pseudomonads alike P. protegens. Double-layer agar assay showed that P. protegens EMM-1 inhibited the growth of several multidrug-resistant (MDR) bacteria, especially clinical isolates of the genera Klebsiella and ß-hemolytic Streptococcus. This strain also displayed inhibitory effects against diverse fungi, such as Aspergillus, Botrytis, and Fusarium. Besides, a crude extract of inhibitory substances secreted into agar was obtained after the cold-leaching process, and physicochemical characterization was performed. The partially purified inhibitory substances produced by P. protegens EMM-1 inhibited the growth of Streptococcus sp. and Microbacterium sp., but no inhibitory effect was noted for other bacterial or fungal strains. The molecular weight determined after ultrafiltration was between 3 and 10 kDa. The inhibitory activity was thermally stable up to 60°C (but completely lost at 100°C), and the inhibitory activity remained active in a wide pH range (from 3 to 9). After treatment with a protease from Bacillus licheniformis, the inhibitory activity was decreased by 90%, suggesting the presence of proteic natural compounds. All these findings suggested that P. protegens EMM-1 is a potential source of antimicrobials to be used against pathogens for humans and plants.


Subject(s)
Anti-Infective Agents/toxicity , Bacteriocins/toxicity , Pseudomonas/metabolism , Anti-Infective Agents/isolation & purification , Anti-Infective Agents/metabolism , Anti-Infective Agents/therapeutic use , Antibiosis , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Bacteriocins/isolation & purification , Bacteriocins/metabolism , Bacteriocins/therapeutic use , Drug Resistance, Multiple, Bacterial/drug effects , Microbial Sensitivity Tests , Mycoses/drug therapy , Mycoses/microbiology , Plant Diseases/prevention & control , Rhizosphere , Zea mays/microbiology
6.
Structure ; 28(5): 528-539.e9, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32220302

ABSTRACT

Phenomycin is a bacterial mini-protein of 89 amino acids discovered more than 50 years ago with toxicity in the nanomolar regime toward mammalian cells. The protein inhibits the function of the eukaryotic ribosome in cell-free systems and appears to target translation initiation. Several fundamental questions concerning the cellular activity of phenomycin, however, have remained unanswered. In this paper, we have used morphological profiling to show that direct inhibition of translation underlies the toxicity of phenomycin in cells. We have performed studies of the cellular uptake mechanism of phenomycin, showing that endosomal escape is the toxicity-limiting step, and we have solved a solution phase high-resolution structure of the protein using NMR spectroscopy. Through bioinformatic as well as functional comparisons between phenomycin and two homologs, we have identified a peptide segment, which constitutes one of two loops in the structure that is critical for the toxicity of phenomycin.


Subject(s)
Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/toxicity , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Bacteriocins/pharmacokinetics , Bacteriocins/toxicity , Cell Line , Endosomes/drug effects , Endosomes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , MCF-7 Cells , Mice , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/toxicity , Structure-Activity Relationship
7.
Toxins (Basel) ; 12(2)2020 02 05.
Article in English | MEDLINE | ID: mdl-32033406

ABSTRACT

Intestinal microbiota exerts protective effects against the infection of various bacterial pathogens, including Listeria monocytogenes, a major foodborne pathogen whose infection can lead to a disease (listeriosis) with a high fatality rate. As a strategy to mitigate the action of the intestinal microbiota, pathogens often produce antimicrobial proteinaceous compounds such as bacteriocins. In this review, we summarize the information currently available for the well-characterized L. monocytogenes bacteriocin listeriolysin S, with the emphasis on its intriguing mode of action as a virulence factor, which promotes the infection of L. monocytogenes by changing the composition of the intestinal microbiota. We then discuss another intriguing L. monocytogenes bacteriocin Lmo2776 that specifically inhibits the inflammogenic species, Prevotella copri, in the intestinal microbiota, reducing superfluous inflammation while weakening virulence. In addition, we describe relatively less studied phage tail-like Listeria bacteriocins (monocins) and elaborate on the possibility that these monocins could be involved in enhancing pathogenicity. In spite of the burgeoning interest in the roles played by the intestinal microbiota against the L. monocytogenes infection, our understanding on the virulence factors affecting the intestinal microbiota is still lacking, calling for further studies on bacteriocins that could function as novel virulence factors.


Subject(s)
Bacteriocins , Gastrointestinal Microbiome/drug effects , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Virulence Factors , Animals , Bacteriocins/genetics , Bacteriocins/toxicity , Host-Pathogen Interactions , Humans , Virulence , Virulence Factors/genetics , Virulence Factors/toxicity
8.
Biocontrol Sci ; 24(4): 185-192, 2019.
Article in English | MEDLINE | ID: mdl-31875610

ABSTRACT

Lactic Acid Bacteria (LAB) are generally recognized as safe. It has been used to increase the shelf-life of fermented products, and its antimicrobial action is based on the metabolites secretions, such as lactic acid, hydrogen peroxide, reuterin, bacteriocins and the like-bacteriocins substances. It has been proven that LAB are able to inhibit deteriorating bacteria of raw meat, but improper handling of live cultures could lead to spoilage. So, the use of their bacteriocins, small antimicrobial peptides, could be an alternative. Besides reducing the number of spoilage bacteria, it seeks to inhibit pathogenic bacteria such as Salmonella, enterohemorrhagic Escherichia coli and Listeria. The food industry uses few bacteriocins and now bacterial resistance has been reported. For that reason, the search of novel bacteriocins produced by LAB is a priority. Moreover, the natural microbiota of meat could be a reservoir of LAB.


Subject(s)
Food Microbiology , Food Storage/methods , Lactobacillales/physiology , Meat/microbiology , Animals , Bacteriocins/metabolism , Bacteriocins/pharmacology , Bacteriocins/toxicity , Drug Resistance, Microbial , Food Contamination/prevention & control , Food Preservation/methods , Food Preservatives/metabolism , Food Preservatives/pharmacology , Food Preservatives/toxicity , Food Safety/methods , Humans , Meat Products/microbiology , Microbiota/physiology , Red Meat/microbiology
9.
PLoS Pathog ; 15(9): e1008029, 2019 09.
Article in English | MEDLINE | ID: mdl-31545853

ABSTRACT

Although Escherichia coli Nissle 1917 (EcN) has been used therapeutically for over a century, the determinants of its probiotic properties remain elusive. EcN produces two siderophore-microcins (Mcc) responsible for an antagonistic activity against other Enterobacteriaceae. EcN also synthesizes the genotoxin colibactin encoded by the pks island. Colibactin is a virulence factor and a putative pro-carcinogenic compound. Therefore, we aimed to decouple the antagonistic activity of EcN from its genotoxic activity. We demonstrated that the pks-encoded ClbP, the peptidase that activates colibactin, is required for the antagonistic activity of EcN. The analysis of a series of ClbP mutants revealed that this activity is linked to the transmembrane helices of ClbP and not the periplasmic peptidase domain, indicating the transmembrane domain is involved in some aspect of Mcc biosynthesis or secretion. A single amino acid substitution in ClbP inactivates the genotoxic activity but maintains the antagonistic activity. In an in vivo salmonellosis model, this point mutant reduced the clinical signs and the fecal shedding of Salmonella similarly to the wild type strain, whereas the clbP deletion mutant could neither protect nor outcompete the pathogen. The ClbP-dependent antibacterial effect was also observed in vitro with other E. coli strains that carry both a truncated form of the Mcc gene cluster and the pks island. In such strains, siderophore-Mcc synthesis also required the glucosyltransferase IroB involved in salmochelin production. This interplay between colibactin, salmochelin, and siderophore-Mcc biosynthetic pathways suggests that these genomic islands were co-selected and played a role in the evolution of E. coli from phylogroup B2. This co-evolution observed in EcN illustrates the fine margin between pathogenicity and probiotic activity, and the need to address both the effectiveness and safety of probiotics. Decoupling the antagonistic from the genotoxic activity by specifically inactivating ClbP peptidase domain opens the way to the safe use of EcN.


Subject(s)
Escherichia coli/physiology , Mutagens/toxicity , Probiotics/therapeutic use , Animals , Antibiosis/genetics , Antibiosis/physiology , Bacteriocins/genetics , Bacteriocins/metabolism , Bacteriocins/toxicity , Biosynthetic Pathways/genetics , Enterobactin/analogs & derivatives , Enterobactin/genetics , Enterobactin/physiology , Enterobactin/toxicity , Escherichia coli/genetics , Escherichia coli/pathogenicity , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/physiology , Female , Genes, Bacterial , Genomic Islands , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Multigene Family , Mutation , Peptide Hydrolases/chemistry , Peptide Hydrolases/genetics , Peptide Hydrolases/physiology , Peptides/genetics , Peptides/physiology , Peptides/toxicity , Polyketides/toxicity , Probiotics/toxicity , Protein Domains , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/therapy , Salmonella typhimurium , Siderophores/genetics , Siderophores/physiology , Siderophores/toxicity , Virulence Factors/genetics , Virulence Factors/physiology , Virulence Factors/toxicity
10.
Int J Antimicrob Agents ; 53(6): 838-843, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30928682

ABSTRACT

The alarming burden of antibiotic resistance in nosocomial pathogens warrants the discovery and development of new and effective antimicrobial compounds. Small cationic antimicrobial peptides seem to be a promising therapeutic alternative to fight multi-drug resistance. This study investigated the in-vitro potential of a previously reported lantibiotic, paenibacillin, from the clinical perspective. An antimicrobial peptide, M152-P4, was isolated, purified and characterized from a mud isolate, and its susceptibility was determined in clinical isolates of Staphylococcus aureus and Enterococcus spp. Time-kill kinetics, resistance, probable mode of action, haemolytic activity and mammalian cytotoxicity were investigated. M152-P4 was identified as paenibacillin based on mass spectroscopy data, amino acid analysis and biosynthetic gene cluster analysis. It had potent antibacterial activity against the Gram-positive pathogens tested, with minimum inhibitory concentrations from 0.1 to 1.56 µM. It appeared very challenging for S. aureus to develop resistance to this compound. Also, paenibacillin penetrated the outer layer of bacteria, and depolarized the membrane completely by creating pores in the plasma membrane with better potential than nisin. Paenibacillin showed no haemolysis up to 60 µM, and the half maximal inhibitory concentration on mammalian cell lines was >100 µM. These results highlight the excellent antibacterial properties of paenibacillin in clinically relevant pathogens. It is stable in the presence of serum, and non-haemolytic and non-cytotoxic even above the therapeutic concentration. Further research efforts regarding toxicity and in-vivo efficacy are necessary to develop paenibacillin as a next-generation therapeutic drug to overcome multi-drug resistance in Gram-positive pathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Bacteriocins/pharmacology , Enterococcus/drug effects , Paenibacillus/metabolism , Staphylococcus aureus/drug effects , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/toxicity , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/isolation & purification , Antimicrobial Cationic Peptides/toxicity , Bacteriocins/chemistry , Bacteriocins/isolation & purification , Bacteriocins/toxicity , Biosynthetic Pathways/genetics , Cell Line , Cell Survival/drug effects , Drug Resistance, Bacterial , Humans , Mass Spectrometry , Microbial Sensitivity Tests , Microbial Viability/drug effects , Multigene Family , Paenibacillus/classification , Paenibacillus/isolation & purification , Sequence Analysis, Protein , Sewage/microbiology
11.
Toxicol Appl Pharmacol ; 374: 32-40, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31034929

ABSTRACT

Clostridium difficile associated disease (CDAD) is the leading infectious cause of antibiotic-associated diarrhea and colitis in the United States. Both the incidence and severity of CDAD have been increased over the past two decades. We evaluated the maximum tolerated dose (MTD) and toxicokinetics of OG253, a novel lantibiotic in development for the treatment of CDAD. OG253 was orally administered to Wistar Han rats as enteric-coated capsules in a one-day dose escalation study, followed by a seven-day repeated dose toxicokinetics study. All three doses of OG253 (6.75, 27 and 108 mg/day) were generally well-tolerated with no treatment-related clinical signs, alterations in body weight or food consumption in both one-day acute tolerability and seven-days repeated dose tolerability and toxicokinetics study. OG253 capsule administration neither significantly alter the weight of organs nor affect the hematology, coagulation, clinical biochemistry parameters and urine pH compared to placebo capsule administered rats. LC-MS/MS analysis did not detect OG253 in the plasma, indicating that OG253 is not absorbed into the blood from the rat gastrointestinal tract. Glandular atrophy of the rectal mucosa was noticed in two out of six rats administered with a high dose of OG253. Surprisingly, we found that OG253 treatment significantly lowered both serum cholesterol and triglyceride levels in both sexes of rats. Overall, there was a 29.8 and 61.38% decrease in the serum cholesterol and triglyceride levels, respectively as compared to placebo-treated rats. The well-tolerated high dose of OG253 (425.7 mg/kg/day) is recommended as the MTD for safety and efficacy studies. Further preclinical study is needed to evaluate the safety profile of OG253 under longer exposure.


Subject(s)
Bacteriocins/administration & dosage , Bacteriocins/toxicity , Animals , Bacteriocins/chemistry , Bacteriocins/pharmacokinetics , Capsules , Dose-Response Relationship, Drug , Female , Male , Molecular Structure , Random Allocation , Rats , Rats, Wistar , Toxicokinetics
12.
Braz J Microbiol ; 50(2): 369-377, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30852798

ABSTRACT

The bacteriocinogenic Enterococcus hirae ST57ACC recently isolated from a Brazilian artisanal cheese was subjected here to additional analyses in order to evaluate its bacteriocin production and the potential influence of ABC transporter system in its expression. Besides these physiological and molecular aspects, the bacteriocin was evaluated for its cytotoxicity against HT-29. Differences in the inoculum size had no impact on the growth of E. hirae ST57ACC; however, the bacteriocin was only produced after 9 h of growth when the strain was inoculated at 5% or 10% (v/v), with similar levels of bacteriocin production obtained by both conventional growth and batch fermentation. Furthermore, potential expression of ABC transporters corresponding to the bacteriocin transport and sugar metabolism was identified. In terms of adverse effects, when a semi-purified fraction of the bacteriocin and the cell-free supernatant were tested against HT-29, total cell viability was similar to observed on untreated cells, indicating the absence of cytotoxic effect. Based on the obtained results, E. hirae ST57ACC can produce its bacteriocin at industrial level by using bioreactors, its bacteriocin expression is potentially influenced by the ABC transporter system, and no cytotoxic effects were observed on HT-29 cells, indicating its potential use as a bio-preservative.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacteriocins/biosynthesis , Bacteriocins/toxicity , Cheese/microbiology , Enterococcus hirae/metabolism , Bacteriocins/genetics , Brazil , Carbohydrate Metabolism , Cell Line , Enterococcus hirae/genetics , Enterococcus hirae/isolation & purification , Food Preservatives , HT29 Cells , Humans
13.
ISME J ; 13(2): 237-249, 2019 02.
Article in English | MEDLINE | ID: mdl-30171255

ABSTRACT

To better understand the potential for antagonistic interactions between members of the same bacterial species, we have surveyed bacteriocin killing activity across a diverse suite of strains of the phytopathogen Pseudomonas syringae. Our data demonstrate that killing activity from phage-derived bacteriocins of P. syringae (R-type syringacins) is widespread. Despite a high overall diversity of bacteriocin activity, strains can broadly be classified into five main killing types and two main sensitivity types. Furthermore, we show that killing activity switches frequently between strains and that switches correlate with localized recombination of two genes that together encode the proteins that specify bacteriocin targeting. Lastly, we demonstrate that phage-derived bacteriocin killing activity can be swapped between strains simply through expression of these two genes in trans. Overall, our study characterizes extensive diversity of killing activity for phage-derived bacteriocins of P. syringae across strains and highlights the power of localized recombination to alter phenotypes that mediate strain interactions during evolution of natural populations and communities.


Subject(s)
Bacteriocins/toxicity , Bacteriophages/metabolism , Pseudomonas syringae/genetics , Recombination, Genetic , Antibiosis , Bacteriocins/biosynthesis , Pseudomonas syringae/classification , Pseudomonas syringae/metabolism , Pseudomonas syringae/virology
14.
Toxicon ; 149: 20-25, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29715468

ABSTRACT

Cyanobacteria produce many biologically active metabolites synthesized via nonribosomal synthetic pathways such as cyclic microcystins (MCs) and linear aeruginosins (Aers). The present study aimed to investigate the effects of different MC variants and the newly isolated aerugenosin Aer-865 on macrophages, which represent one of the key effector cells within the innate immune responses. Specifically, our study included RAW 264.7 macrophage activation associated with production of cytotoxic and cytostatic nitric oxide (NO) as well as pro-inflammatory mediators like tumor necrosis factor α (TNFα) and interleukin 6 (IL-6). From the compounds investigated, commonly occurring MC variants (-RR, -YR) and Aer-865 had no significant effects within the non-cytotoxic concentrations tested, i.e. 0.001-1 µM for MCs and 0.1-50 µM for Aer-865. In contrast to known immunoactive MC-LR, the negligible immunomodulatory potential of tested MC congeners could be related to their differences in structure. The knowledge of MC structure-specific activities contributes to the understanding of complex toxicity of different MC variants and most importantly their mixtures. This study is one of the first study that evaluate the effect of larger set of cyanobacterial peptides on macrophages and compare their immunomodulatory potential.


Subject(s)
Bacterial Toxins/toxicity , Bacteriocins/toxicity , Immunomodulation/drug effects , Macrophages/drug effects , Marine Toxins/toxicity , Microcystins/toxicity , Animals , Bacterial Toxins/chemistry , Bacteriocins/chemistry , Cyanobacteria Toxins , Macrophages/immunology , Marine Toxins/chemistry , Mice , Microcystins/chemistry , RAW 264.7 Cells
15.
JACC Cardiovasc Imaging ; 11(12): 1823-1833, 2018 12.
Article in English | MEDLINE | ID: mdl-29454770

ABSTRACT

OBJECTIVES: The purpose of this study was to evaluate the feasibility of imaging apoptosis in experimental ischemia-reperfusion model by technetium-99m (99mTc)-labeled Duramycin, and compare it to an established tracer, 99mTc-labeled Annexin-V, which has a relative disadvantage of high radiation burden to nontarget organs. BACKGROUND: During apoptosis, the cell membrane phospholipids-phosphatidylserine (PS) and phosphatidylethanolamine (PE) are exposed and can be targeted by Annexin-V and Duramycin, respectively, for in vivo imaging. Identification of a reversible cell death process should permit therapeutic intervention to help reduce myocyte loss and left ventricle dysfunction. METHODS: In a 40-min left coronary artery ischemia-reperfusion model in 17 rabbits, 7 mCi of 99mTc-labeled Duramycin (n = 10), 99mTc-linear Duramycin (a negative tracer control; n = 3), or 99mTc-Annexin-V (a positive tracer-control; n = 4) were intravenously administered 30 min after reperfusion. Of the 10 Duramycin group animals, 4 animals were treated with an antiapoptotic agent, minocycline at the time of reperfusion. In vivo and ex vivo micro-single-photon emission computed tomography (µSPECT) and micro-computed tomography (µCT) imaging was performed 3 h after reperfusion, followed by quantitative assessment of tracer uptake and pathological characterization. Fluorescent Duramycin and Annexin-V were injected in 4 rats to visualize colocalization in infarct areas in a 40-min left coronary artery occlusion and 30-min reperfusion model. RESULTS: Intense uptake of Duramycin and Annexin-V was observed in the apical (infarcted) areas. The percent injected dose per gram uptake of Duramycin in apical region (0.751 ± 0.262%) was significantly higher than remote area in same animals (0.045 ± 0.029%; p < 0.01). Duramycin uptake was insignificantly lower than Annexin-V uptake (1.23 ± 0.304%; p > 0.01) but demonstrated substantially lower radiation burden to kidneys (0.358 ± 0.210% vs. 1.58 ± 0.316%, respectively; p < 0.001). Fluorescence studies with Duramycin and Annexin V showed colocalization in infarct areas. Minocycline treatment substantially resolved Duramycin uptake (0.354% ± 0.0624%; p < 0.01). CONCLUSIONS: Duramycin is similarly effective in imaging apoptotic cell death as Annexin-V with lower nontarget organ radiation. Clinical feasibility of apoptosis imaging with a PE-seeking tracer should be tested.


Subject(s)
Annexin A5/administration & dosage , Apoptosis , Bacteriocins/administration & dosage , Molecular Imaging/methods , Myocardial Infarction/diagnostic imaging , Myocardial Reperfusion Injury/diagnostic imaging , Myocardium/pathology , Organotechnetium Compounds/administration & dosage , Phosphatidylethanolamines/metabolism , Radiopharmaceuticals/administration & dosage , Tomography, Emission-Computed, Single-Photon , Animals , Annexin A5/toxicity , Bacteriocins/toxicity , Disease Models, Animal , Feasibility Studies , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Organotechnetium Compounds/toxicity , Organs at Risk , Predictive Value of Tests , Rabbits , Radiopharmaceuticals/toxicity , Risk Assessment , Time Factors , X-Ray Microtomography
16.
J Clin Invest ; 126(6): 2321-33, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27159394

ABSTRACT

Cell ablation is a powerful tool for studying cell lineage and/or function; however, current cell-ablation models have limitations. Intermedilysin (ILY), a cytolytic pore-forming toxin that is secreted by Streptococcus intermedius, lyses human cells exclusively by binding to the human complement regulator CD59 (hCD59), but does not react with CD59 from nonprimates. Here, we took advantage of this feature of ILY and developed a model of conditional and targeted cell ablation by generating floxed STOP-CD59 knockin mice (ihCD59), in which expression of human CD59 only occurs after Cre-mediated recombination. The administration of ILY to ihCD59+ mice crossed with various Cre-driver lines resulted in the rapid and specific ablation of immune, epithelial, or neural cells without off-target effects. ILY had a large pharmacological window, which allowed us to perform dose-dependent studies. Finally, the ILY/ihCD59-mediated cell-ablation method was tested in several disease models to study immune cell functionalities, hepatocyte and/or biliary epithelial damage and regeneration, and neural cell damage. Together, the results of this study demonstrate the utility of the ihCD59 mouse model for studying the effects of cell ablation in specific organ systems in a variety of developmental and disease states.


Subject(s)
Bacteriocins/administration & dosage , CD59 Antigens/physiology , Animals , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/pathology , Bacteriocins/toxicity , CD59 Antigens/genetics , Gene Knock-In Techniques , Hemolysis/genetics , Hemolysis/physiology , Hepatitis, Animal/etiology , Hepatitis, Animal/genetics , Hepatitis, Animal/immunology , Hepatocytes/drug effects , Hepatocytes/immunology , Hepatocytes/pathology , Humans , Integrases , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
17.
Biomolecules ; 5(2): 1178-94, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26111195

ABSTRACT

Lactic acid bacteria (LAB) have historically been used in food fermentations to preserve foods and are generally-recognized-as-safe (GRAS) by the FDA for use as food ingredients. In addition to lactic acid; some strains also produce bacteriocins that have been proposed for use as food preservatives. In this study we examined the inhibition of Listeria monocytogenes 39-2 by neutralized and non-neutralized bacteriocin preparations (Bac+ preps) produced by Lactobacillus curvatus FS47; Lb. curvatus Beef3; Pediococcus acidilactici Bac3; Lactococcus lactis FLS1; Enterococcus faecium FS56-1; and Enterococcus thailandicus FS92. Activity differences between non-neutralized and neutralized Bac+ preps in agar spot assays could not readily be attributed to acid because a bacteriocin-negative control strain was not inhibitory to Listeria in these assays. When neutralized and non-neutralized Bac+ preps were used in microplate growth inhibition assays against L. monocytogenes 39-2 we observed some differences attributed to acid inhibition. A microplate growth inhibition assay was used to compare inhibitory reactions of wild-type and bacteriocin-resistant variants of L. monocytogenes to differentiate bacteriocins with different modes-of-action (MOA) whereby curvaticins FS47 and Beef3, and pediocin Bac3 were categorized to be in MOA1; enterocins FS92 and FS56-1 in MOA2; and lacticin FLS1 in MOA3. The microplate bacteriocin MOA assay establishes a platform to evaluate the best combination of bacteriocin preparations for use in food applications as biopreservatives against L. monocytogenes.


Subject(s)
Anti-Bacterial Agents/toxicity , Bacteriocins/toxicity , High-Throughput Screening Assays/methods , Listeria monocytogenes/drug effects , Anti-Bacterial Agents/chemistry , Bacteriocins/chemistry , Enterococcus/chemistry , Enterococcus/metabolism , Lactobacillus/chemistry , Lactobacillus/metabolism , Microbial Sensitivity Tests/methods , Pediococcus/chemistry , Pediococcus/metabolism
18.
Toxins (Basel) ; 7(1): 110-28, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25590277

ABSTRACT

Gardnerella vaginalis produces cytolysin vaginolysin (VLY), which has been suggested to be a contributor to bacterial vaginosis pathogenesis. VLY along with intermedilysin (ILY) from Streptococcus intermedius have been attributed to a group of cholesterol-dependent cytolysins (CDCs) whose pore-forming activity depends on human CD59 (hCD59). Here, we show that different types of cells lacking hCD59 are susceptible to VLY-mediated lysis, albeit to different extents. We analyze the effects of both hCD59 and cholesterol on VLY cytolytic activity. We show that VLY binds to cholesterol-rich membranes of non-human cells, while VLY with an impaired cholesterol recognition site retains binding to the hCD59-containing cells. We further demonstrate that cholesterol binding by VLY is sufficient to trigger the formation of oligomeric complexes on cholesterol rich-liposomes lacking hCD59. Thus, VLY may induce cell lysis following two alternative pathways. One requires only cholesterol and does not depend on hCD59. The second pathway involves hCD59 contribution similarly to ILY. Apparently, under physiological conditions VLY acts in the most effective way by accepting the assistance of hCD59.


Subject(s)
Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , CD59 Antigens/metabolism , Cholesterol/metabolism , Cytotoxins/toxicity , Adult , Animals , Bacteriocins/toxicity , CD59 Antigens/genetics , CHO Cells , Cells, Cultured , Cricetulus , Erythrocytes/drug effects , Erythrocytes/metabolism , HeLa Cells , Humans , Mice , Streptolysins/toxicity
19.
Exp Parasitol ; 144: 14-21, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24929146

ABSTRACT

Through increased awareness and improved diagnostics, microsporidiosis has now been identified in a broader range of human populations; however current therapies are inconsistently effective. Recently, probiotics were determined as means for the control of intestinal parasitic infections through their secretory products; bacteriocins. This is the first study on the effect of bacteriocin produced by Lactobacillus acidophilus CH1 bacteriocin, with or without gold nanoparticles (Au-NPs), against intestinal microsporidiosis in immunosuppressed mice. Fecal and intestinal spore loads, besides viability, extrusion and infectivity of spores from treated animals were assessed. Results showed that the anti-microsporidial effects of bacteriocin were significantly potent. This efficiency was further potentiated upon conjugating bacteriocins with Au-NPs, as it induced a strikingly sustained reduction in fecal spore shedding after cessation of therapy by 1 week (94.26%). Furthermore, reduction in intestinal spore load was highest in bacteriocin/Au-NPs-inoculated mice (89.7%) followed by bacteriocin-inoculated group (73.5%). Spores encountered from stool of bacteriocin/Au-NPs group showed 92.4% viability, versus 93.7% in bacteriocin group. Spore extrusion and infectivity were most inhibited by exposure to bacteriocin/Au-NPs. Safety of bacteriocin/Au-NPs was also verified. Thus, considering the results of the present work, L. acidophilus CH1-derived bacteriocin can present a powerful safe therapy against intestinal microsporidiosis.


Subject(s)
Bacteriocins/pharmacology , Enterocytozoon/drug effects , Lactobacillus acidophilus/metabolism , Metal Nanoparticles , Microsporidiosis/drug therapy , Analysis of Variance , Animals , Bacteriocins/administration & dosage , Bacteriocins/therapeutic use , Bacteriocins/toxicity , Drug Synergism , Feces/parasitology , Gold , Humans , Intestine, Small/parasitology , Kidney/drug effects , Liver/drug effects , Male , Metal Nanoparticles/therapeutic use , Metal Nanoparticles/toxicity , Mice , Spores, Fungal/isolation & purification
20.
Appl Environ Microbiol ; 80(13): 3835-41, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24747893

ABSTRACT

Carnocyclin A (CCLA) is an antimicrobial peptide produced by Carnobacterium maltaromaticum ATCC PTA-5313, which can be used to control the growth of Listeria monocytogenes in ready-to-eat meat products. The aim of this research was to elucidate the cellular responses of L. monocytogenes 08-5923 exposed to a sublethal dose of CCLA. Microarray, quantitative reverse transcription-PCR, tandem mass spectrometry, and electron microscopy were used to investigate the alteration in gene expression, protein production, and morphological changes in cells of Listeria following treatment with CCLA. The genes involved in metabolism (baiE, trn, and pykA), cell wall synthesis (murZ and dacB2), and cell division (clpE and divIVA) were upregulated following a 15-min exposure to CCLA as a result of stress responses. Genes involved in cell division, cell wall synthesis, flagellar synthesis, and metabolism were downregulated after 4 h as a result of adaptation. Analysis of total soluble proteins confirmed the downregulation of pykA and gnd after 4 h of exposure to CCLA. The absence of flagella was observed in L. monocytogenes following 30 h of exposure to CCLA. A sublethal dose of CCLA induced adaptation in L. monocytogenes 08-5923 by inhibition of expression of genes and proteins critical for synthesis of cell wall structures and maintaining metabolic functions. Both the mannose- and cellobiose-specific phosphotransferase systems could be targets for CCLA.


Subject(s)
Bacteriocins/toxicity , Listeria monocytogenes/drug effects , Peptides, Cyclic/toxicity , Stress, Physiological , Gene Expression Profiling , Gene Expression Regulation, Bacterial/drug effects , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Listeria monocytogenes/ultrastructure , Microarray Analysis , Microscopy, Electron , Proteome/analysis , Real-Time Polymerase Chain Reaction , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...