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.
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
3.
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
4.
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
5.
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
6.
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
7.
J Infect Dis ; 208(10): 1545-52, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23997234

ABSTRACT

BACKGROUND: Bacterial genotoxins provoke DNA damage and carcinogenesis. The Escherichia coli uropathogenic-specific protein gene, usp, and its linked genes, imu1-3, are associated with strains from pyelonephritis, prostatitis, and bacteremia of urinary tract origin. While the Usp C-terminal domain exhibits similarity with DNase-like colicins and pyocins, its role and mechanisms of action, as well as those of the 3 associated proteins, is unknown. METHODS: We isolated Usp and Imu1-3 and examined their activity on plasmid DNA, human umbilical vein endothelial cells, and human embryonic kidney cells (cell line HEK293). The affect of Usp and Imu1-3 was assessed by MTT and Comet assays, infection assays, caspase 3/7 activity, fluorescently labeled actin staining, and Western blotting. RESULTS: Usp possesses DNase activity and, particularly when coapplied with Imu2, exhibits genotoxic activity in mammalian cells. Infection assays demonstrated that E. coli usp(+) imu1-3(+) affects the viability of mammalian cells, induces increased caspase 3/7 activity, and perturbs cell cytoskeleton structure. CONCLUSIONS: Usp is a novel E. coli genotoxin active against mammalian cells. Optimal in vivo activity of Usp requires Imu2. Infection with E. coli usp(+) imu1-3(+) induces a response characteristic of apoptosis.


Subject(s)
Bacteriocins/pharmacology , Escherichia coli Proteins/pharmacology , Mutagens/pharmacology , Bacteriocins/toxicity , Caspases/metabolism , Cell Line , Cell Survival/drug effects , Cytoskeleton/drug effects , Deoxyribonucleases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/toxicity , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Mutagens/toxicity
8.
J Appl Microbiol ; 115(2): 484-94, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23639072

ABSTRACT

AIM: The current study aimed to visualize the damage caused by enterolysin A to the cells of sensitive strains and to find out cleavage site within the peptidoglycan moiety of bacterial cell walls. METHODS AND RESULTS: Enterolysin A produced by a local isolate, Enterococcus faecalis B9510 was found to rapidly kill cells of the sensitive strain Lactococcus lactis ssp. cremoris 2144 during 120 min of treatment as compared to the untreated control where no such effect was observed. Transmission electron microscopy of the enterolysin A-treated cells revealed leaking of the cytoplasmic contents ultimately resulting in complete lysis of cell walls. To find the cleavage site, purified cell walls of L. lactis ssp. cremoris 2144, Pediococcus pentosaceus 43201 and Lactobacillus delbrueckii ssp. bulgaricus ATCC 11842 were treated with enterolysin A, and liberated amino acids were derivatized for N and C terminals and analysed using thin layer chromatography on silica gel with isopropanol as solvent. The results showed that enterolysin A cleaves the peptide bonds at two locations within peptidoglycan subunits. The first location is between L-alanine and D-glutamic acid of the stem peptide and the other location is between L-lysine of the stem peptide and D-aspartic acid of the interpeptide bridge. CONCLUSIONS: Enterolysin A cleaves the peptide bonds within the stem peptide as well as in the interpeptide bridge of Gram-positive bacterial cell walls. This gives a possible reason for the broad spectrum of enterolysin A activity. SIGNIFICANCE AND IMPACT OF THE STUDY: This is the first report identifying the cleavage site of enterolysin A within the cell walls of sensitive bacteria. This will help in identifying potential applications for enterolysin A.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriocins/pharmacology , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/toxicity , Bacteriocins/chemistry , Bacteriocins/toxicity , Cell Wall/drug effects , Cell Wall/metabolism , Enterococcus faecalis/metabolism , Lactococcus lactis/drug effects , Molecular Sequence Data , Pediococcus/drug effects , Peptidoglycan/chemistry , Peptidoglycan/metabolism , Sequence Homology, Amino Acid
9.
Appl Microbiol Biotechnol ; 97(23): 10081-90, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24085393

ABSTRACT

Bacillus amyloliquefaciens FZB42 has been shown to stimulate plant growth and to suppress the growth of plant pathogenic organisms including nematodes. However, the mechanism underlying its effect against nematodes remains unknown. In this study, we screened a random mutant library of B. amyloliquefaciens FZB42 generated by the mariner transposon TnYLB-1 and identified a mutant strain F5 with attenuated nematicidal activity. Reversible polymerase chain reaction revealed that three candidate genes RAMB_007470, yhdY, and prkA that were disrupted by the transposon in strain F5 potentially contributed to its decreased nematicidal activity. Bioassay of mutants impaired in the three candidate genes demonstrated that directed deletion of gene RBAM_007470 resulted in loss of nematicidal activity comparable with that of the F5 triple mutant. RBAM_007470 has been reported as being involved in biosynthesis of plantazolicin, a thiazole/oxazole-modified microcin with hitherto unknown function. Electrospray ionization time-of-flight mass spectrometry (ESI-TOF-MS) analyses of surface extracts revealed that plantazolicin bearing a molecular weight of 1,354 Da was present in wild-type B. amyloliquefaciens FZB42, but absent in the ΔRABM_007470 mutant. Furthermore, bioassay of the organic extract containing plantazolicin also showed a moderate nematicidal activity. We conclude that a novel gene RBAM_007470 and its related metabolite are involved in the antagonistic effect exerted by B. amyloliquefaciens FZB42 against nematodes.


Subject(s)
Antinematodal Agents/metabolism , Bacillus/metabolism , Bacterial Proteins/metabolism , Oligopeptides/biosynthesis , Animals , Antinematodal Agents/chemistry , Antinematodal Agents/toxicity , Bacillus/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/toxicity , Bacteriocins/biosynthesis , Bacteriocins/chemistry , Bacteriocins/genetics , Bacteriocins/toxicity , Base Sequence , Molecular Sequence Data , Nematoda/drug effects , Oligopeptides/genetics , Oligopeptides/toxicity
10.
Microbiology (Reading) ; 158(Pt 11): 2851-2858, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22956757

ABSTRACT

Bacteriocins are ribosomally synthesized antimicrobial peptides produced by Bacteria and some Archaea. The assessment of the toxic potential of antimicrobial peptides is important in order to apply these peptides on an industrial scale. The aim of the present study was to investigate the in vitro cytotoxic and haemolytic potential of bovicin HC5, as well as to determine whether cholesterol influences bacteriocin activity on model membranes. Nisin, for which the mechanism of action is well described, was used as a reference peptide in our assays. The viability of three distinct eukaryotic cell lines treated with bovicin HC5 or nisin was analysed by using the MTT assay and cellular morphological changes were determined by light microscopy. The haemolytic potential was evaluated by using the haemoglobin liberation assay and the role of cholesterol on bacteriocin activity was examined by using model membranes composed of DOPC (1,2-dioleoyl-sn-glycero-3-phosphocholine) and DPoPC (1,2-dipalmitoyl-sn-glycero-3-phosphocholine). The IC(50) of bovicin HC5 and nisin against Vero cells was 65.42 and 13.48 µM, respectively. When the MTT assay was performed with MCF-7 and HepG2 cells, the IC(50) obtained for bovicin HC5 was 279.39 and 289.30 µM, respectively, while for nisin these values were 105.46 and 112.25 µM. The haemolytic activity of bovicin HC5 against eukaryotic cells was always lower than that determined for nisin. The presence of cholesterol did not influence the activity of either bacteriocin on DOPC model membranes, but nisin showed reduced carboxyfluorescein leakage in DPoPC membranes containing cholesterol. In conclusion, bovicin HC5 only exerted cytotoxic effects at concentrations that were greater than the concentration needed for its biological activity, and the presence of cholesterol did not affect its interaction with model membranes.


Subject(s)
Bacteriocins/toxicity , Cholesterol/metabolism , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , Animals , Bacteriocins/chemistry , Cell Line , Cell Survival/drug effects , Chlorocebus aethiops , Hemolytic Agents/toxicity , Humans , Vero Cells
11.
Appl Environ Microbiol ; 78(18): 6592-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22773653

ABSTRACT

A novel phenotype was recently identified in which specific strains of Escherichia coli inhibit competing E. coli strains via a mechanism that was designated "proximity-dependent inhibition" (PDI). PDI-expressing (PDI(+)) E. coli is known to inhibit susceptible (PDI(-)) E. coli strains, including several enterohemorrhagic (EHEC) and enterotoxigenic (ETEC) E. coli strains. In this study, every strain from a genetically diverse panel of E. coli O157:H7 (n = 25) and additional strains of E. coli serovar O26 were susceptible to the PDI phenotype. LIVE/DEAD staining was consistent with inhibition by killing of susceptible cells. Comparative genome analysis identified the genetic component of PDI, which is composed of a plasmid-borne (Incl1) operon encoding a putative microcin and associated genes for transport, immunity, and microcin activation. Transfer of the plasmid to a PDI(-) strain resulted in transfer of the phenotype, and deletion of the genes within the operon resulted in loss of the inhibition phenotype. Deletion of chromosomally encoded tolC also resulted in loss of the inhibitory phenotype, and this confirmed that the putative microcin is most likely secreted via a type I secretion pathway. Deletion of an unrelated plasmid gene did not affect the PDI phenotype. Quantitative reverse transcription (RT)-PCR demonstrated that microcin expression is correlated with logarithmic-phase growth. The ability to inhibit a diversity of E. coli strains indicates that this microcin may influence gut community composition and could be useful for control of important enteric pathogens.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/toxicity , Antibiosis , Bacteriocins/isolation & purification , Bacteriocins/toxicity , Enterohemorrhagic Escherichia coli/drug effects , Enterohemorrhagic Escherichia coli/physiology , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Escherichia coli Proteins/genetics , Gene Deletion , Gene Expression Profiling , Gene Transfer, Horizontal , Genes, Bacterial , Molecular Sequence Data , Operon , Plasmids , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
12.
Bioorg Med Chem Lett ; 22(22): 6919-22, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23040731

ABSTRACT

Naturally occurring furanosteroids such as viridin and wortmannin have long been known as potent inhibitors of the lipid kinase PI-3K. We have been interested in directly accessing analogs of these complex natural products from abundant steroid feedstock materials. In this communication, we describe the synthesis of viridin/wortmannin hybrid molecules from readily available building blocks that function as PI-3K inhibitors and maintain their electrophilic properties. The compounds also show anti-proliferative effects against a breast cancer line.


Subject(s)
Androstenes/chemistry , Bacteriocins/chemistry , Protein Kinase Inhibitors/chemistry , Steroids/chemistry , Androstadienes/chemistry , Androstenes/chemical synthesis , Androstenes/toxicity , Bacteriocins/chemical synthesis , Bacteriocins/toxicity , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Survival/drug effects , Crystallography, X-Ray , Female , Humans , MCF-7 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/toxicity , Protein Structure, Tertiary , Wortmannin
13.
Environ Monit Assess ; 184(11): 6627-35, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22131013

ABSTRACT

Bacteria play a significant role in water contamination. Chemicals are mostly used for the treatment of bacteriologically contaminated water. The use of bacterial interactions is a new approach to limit the pathogens' growth. Detection of antimicrobial substances produced by lactic acid bacteria against the waterborne pathogens is the objective of this work. Microbiological and biochemical methods were used to identify lactic acid bacteria having an antimicrobial activity. Evaluation of antimicrobial activity with growth kinetic measurements was performed. Four isolates of lactic acid bacteria obtained from whey and curd were identified. The predominant species belonging to the Lactobacillus genera are: Lactobacillus rhamnosus, Lactobacillus sakei, Lactobacillus paracasei, and Lactobacillus paraplantarum. The present study revealed that the Lactobacillus consortium is able to inhibit Staphylococcus aureus's growth along with Escherichia coli and Vibrio species. In mixed culture, after 24 h, the Lactobacillus consortium reduces the growth of S. aureus by 2.03 log; moreover, the growth of the latter bacteria totally ceased after 72 h of incubation. The protein produced by the Lactobacillus consortium was responsible for arresting the growth of S. aureus.


Subject(s)
Bacteria/growth & development , Biological Control Agents , Lactobacillus/physiology , Water Microbiology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/toxicity , Bacteriocins/metabolism , Bacteriocins/toxicity , Escherichia coli/growth & development , Microbial Sensitivity Tests , Staphylococcus aureus/growth & development
14.
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
15.
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
16.
Curr Opin Microbiol ; 11(5): 401-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18848642

ABSTRACT

Type A (I) lantibiotics are cationic antimicrobial peptides that have a potential usefulness in treating infectious diseases. They are known to have a potent and broad spectrum of activity, an insignificant cytotoxicity, and demonstrated efficacy in animal infection models, suggesting therapeutic potential. In this review, topics pertaining to their basic structure, mode of bactericidal activity, pharmacology, and methods of manufacture are described.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriocins/pharmacology , Amino Acid Sequence , Animals , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/toxicity , Bacterial Infections/drug therapy , Bacteriocins/biosynthesis , Bacteriocins/chemistry , Bacteriocins/toxicity , Humans , Molecular Sequence Data
17.
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
18.
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
19.
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
20.
Mikrobiol Z ; 71(6): 21-6, 2009.
Article in Ukrainian | MEDLINE | ID: mdl-20455429

ABSTRACT

A comparative analysis of results of toxicological research of microbiological preparations on the basis of different species of nitrogen-fixing microorganisms of Azotobacter, Agrobacterium, Azospirillum general and pathogenic properties of strains-producers has been carried out. A possibility to improve methodical principles of toxicological estimation and hygienic regulation of associative nitrogen-fixing microorganisms-producers and preparations on their basis in the industrial objects and environment is substantiated. The paper is presented in Ukrainian.


Subject(s)
Azospirillum/pathogenicity , Azotobacter/pathogenicity , Bacteriocins/toxicity , Biological Products/toxicity , Nitrogen/metabolism , Rhizobium/pathogenicity , Animals , Azospirillum/metabolism , Azotobacter/metabolism , Bacteriocins/isolation & purification , Biological Products/isolation & purification , Biopharmaceutics/standards , Lethal Dose 50 , Mice , Rats , Rhizobium/metabolism , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL