Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.023
Filter
1.
World J Microbiol Biotechnol ; 40(9): 287, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39090427

ABSTRACT

Bacteriocins are antimicrobial peptides produced by bacteria to prevent the growth of pathogens. Combining bacteriocins with metal nanoparticles, like silver nanoparticles (AgNPs), has developed into a viable strategy to get over bacteriocin limitations. In this study, bacteriocin BacZY05 was extracted from Bacillus subtilis ZY05 and purified using various techniques. The resulting purified bacteriocin was then combined with silver nanoparticles to form bacteriocin silver nanoconjugates (BacZY05-AgNPs). The physicochemical properties of the BacZY05-AgNPs were characterized using various analytical techniques. The mean diameter of the synthesized AgNPs was approximately 20-60 nm with an oval or spherical shape. The antimicrobial activity of the BacZY05-AgNPs was evaluated against several indicator strains by their zone of inhibition (ZOI), using the agar well diffusion method. Compared to bacteriocin (ZOI- 13 to 20 mm) and AgNPs (ZOI- 10-22 mm) alone, the antibacterial activity data demonstrated a 1.3-1.5-fold increase in the activity of bacteriocin-nanoconjugates (ZOI- 22 to 26 mm). For Staphylococcus aureus MTCC3103 and Klebsiella pneumoniae MTCC109, BacZY05-capped AgNPs exhibited the lowest minimum inhibitory concentration (MIC), measuring 10.93 µg/mL. For Salmonella typhi NCIM2501, the MIC was 28.75 µg/mL. The highest MIC value was 57.5 µg/mL for Escherichia coli DH5α and Vibrio cholerae MTCC3909. With BacZY05-capped AgNPs, the lowest minimum bactericidal concentration (MBC) of 28.75 µg/mL was observed for Staphylococcus aureus MTCC31003. In the cases of Salmonella typhi NCIM2501 and Klebsiella pneumoniae MTCC109 concentration was 57.5 µg/mL. Vibrio cholerae MTCC3909 and Escherichia coli DH5α had the highest MBC values at 115 µg/mL.


Subject(s)
Anti-Bacterial Agents , Bacillus subtilis , Bacteriocins , Klebsiella pneumoniae , Metal Nanoparticles , Microbial Sensitivity Tests , Nanoconjugates , Silver , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Silver/pharmacology , Silver/chemistry , Bacteriocins/pharmacology , Bacteriocins/chemistry , Bacteriocins/biosynthesis , Metal Nanoparticles/chemistry , Staphylococcus aureus/drug effects , Nanoconjugates/chemistry , Bacillus subtilis/drug effects , Klebsiella pneumoniae/drug effects , Escherichia coli/drug effects
2.
Nat Commun ; 15(1): 6332, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068147

ABSTRACT

Bacteriocins are antimicrobial peptides that are naturally produced by many bacteria. They hold great potential in the fight against antibiotic resistant bacteria, including ESKAPE pathogens. Engineered live biotherapeutic products (eLBPs) that secrete bacteriocins can be created to deliver targeted bacteriocin production. Here we develop a modular bacteriocin secretion platform that can be used to express and secrete multiple bacteriocins from non-pathogenic Escherichia coli host strains. As a proof of concept we create Enterocin A (EntA) and Enterocin B (EntB) secreting strains that show strong antimicrobial activity against Enterococcus faecalis and Enterococcus faecium in vitro, and characterise this activity in both solid culture and liquid co-culture. We then develop a Lotka-Volterra model that can be used to capture the interactions of these competitor strains. We show that simultaneous exposure to EntA and EntB can delay Enterococcus growth. Our system has the potential to be used as an eLBP to secrete additional bacteriocins for the targeted killing of pathogenic bacteria.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Enterococcus faecalis , Enterococcus faecium , Escherichia coli , Bacteriocins/pharmacology , Bacteriocins/metabolism , Bacteriocins/biosynthesis , Enterococcus faecalis/metabolism , Enterococcus faecalis/drug effects , Enterococcus faecalis/genetics , Enterococcus faecium/metabolism , Enterococcus faecium/genetics , Enterococcus faecium/drug effects , Escherichia coli/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Coculture Techniques
3.
Int J Biol Macromol ; 277(Pt 1): 133916, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39033897

ABSTRACT

Bacteriocins are a diverse group of ribosomally synthesised antimicrobial peptides/proteins that play an important role in self-defence. They are widely used as bio-preservatives and effective substitutes for disease eradication. They can be used in conjunction with or as an alternative to antibiotics to minimize the risk of resistance development. There are remarkably few reports indicating resistance to bacteriocins. Although there are many research reports that emphasise heterologous expression of bacteriocin, there are no convincing reports on the significant role that intrinsic and extrinsic factors play in overexpression. A coordinated and cooperative expression system works in concert with multiple genetic elements encoding native proteins, immunoproteins, exporters, transporters and enzymes involved in the post-translational modification of bacteriocins. The simplest way could be to utilise the existing E. coli expression system, which is conventional, widely used for heterologous expression and has been further extended for bacteriocin expression. In this article, we will review the intrinsic and extrinsic factors, advantages, disadvantages and major problems associated with bacteriocin overexpression in E. coli. Finally, we recommend the most effective strategies as well as numerous bacteriocin expression systems from E. coli, Lactococcus, Kluveromyces lactis, Saccharomyces cerevisiae and Pichia pastoris for their suitability for successful overexpression.


Subject(s)
Bacteriocins , Escherichia coli , Bacteriocins/genetics , Bacteriocins/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression
4.
Microb Cell Fact ; 23(1): 175, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38872163

ABSTRACT

INTRODUCTION: Bacterial infections and the rising antimicrobial resistance pose a significant threat to public health. Pseudomonas aeruginosa produces bacteriocins like pyocins, especially S-type pyocins, which are promising for biological applications. This research focuses on clinical P. aeruginosa isolates to assess their bacteriocin production, inhibitory spectrum, chemical structure, antibacterial agents, and preservative potential. METHODS: The identification of P. aeruginosa was conducted through both phenotypic and molecular approaches. The inhibitory spectrum and antibacterial potential of the isolates were assessed. The kinetics of antibacterial peptide production were investigated, and the activity of bacteriocin was quantified in arbitrary units (AU ml-1). Physico-chemical characterization of the antibacterial peptides was performed. Molecular weight estimation was carried out using SDS-PAGE. qRT-PCR analysis was employed to validate the expression of the selected candidate gene. RESULT: The antibacterial activity of P. aeruginosa was attributed to the secretion of bacteriocin compounds, which belong to the S-type pyocin family. The use of mitomycin C led to a significant 65.74% increase in pyocin production by these isolates. These S-type pyocins exhibited the ability to inhibit the growth of both Gram-negative (P. mirabilis and P. vulgaris) and Gram-positive (S. aureus, S. epidermidis, E. hirae, S. pyogenes, and S. mutans) bacteria. The molecular weight of S-type pyocin was 66 kDa, and its gene expression was confirmed through qRT-PCR. CONCLUSION: These findings suggest that S-type pyocin hold significant potential as therapeutic agents against pathogenic strains. The Physico-chemical resistance of S-type pyocin underscores its potential for broad applications in the pharmaceutical, hygiene, and food industries.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Microbial Sensitivity Tests , Pseudomonas aeruginosa , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/biosynthesis , Bacteriocins/biosynthesis , Bacteriocins/pharmacology , Bacteriocins/metabolism , Pyocins/metabolism , Pyocins/pharmacology , Pyocins/biosynthesis , Humans , Pseudomonas Infections/microbiology , Pseudomonas Infections/drug therapy
5.
Int J Mol Sci ; 25(11)2024 May 26.
Article in English | MEDLINE | ID: mdl-38891977

ABSTRACT

Class IIa bacteriocins produced in lactic acid bacteria are short cationic peptides with antimicrobial activity. In the search for new biopreservation agents, class IIa bacteriocins are considered to be the best potential candidates, not only due to their large abundance but also because of their high biological activity and excellent thermal stability. However, regulated by the biosynthetic regulatory system, the natural class IIa bacteriocin yield is low, and the extraction process is complicated. The biotechnological production of class IIa bacteriocins in various cell factories has been attempted to improve this situation. In this review, we focus on the application of biotechnological routes for class IIa bacteriocin production. The drawbacks and improvements in the production of class IIa bacteriocins in various cell factories are discussed. Furthermore, we present the main challenge of class IIa bacteriocins, focusing on increasing their production by constructing suitable cell factories. Recombinant bacteriocins have made considerable progress from inclusion body formation, dissolved form and low antibacterial activity to yield recovery. The development of prospective cell factories for the biotechnological production of bacteriocins is still required, which may facilitate the application of bacteriocins in the food industry.


Subject(s)
Bacteriocins , Biotechnology , Bacteriocins/biosynthesis , Biotechnology/methods , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/pharmacology , Recombinant Proteins/metabolism , Recombinant Proteins/biosynthesis , Lactobacillales/metabolism
6.
BMC Microbiol ; 24(1): 226, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937695

ABSTRACT

BACKGROUND: Bacterial antimicrobial resistance poses a severe threat to humanity, necessitating the urgent development of new antibiotics. Recent advances in genome sequencing offer new avenues for antibiotic discovery. Paenibacillus genomes encompass a considerable array of antibiotic biosynthetic gene clusters (BGCs), rendering these species as good candidates for genome-driven novel antibiotic exploration. Nevertheless, BGCs within Paenibacillus genomes have not been extensively studied. RESULTS: We conducted an analysis of 554 Paenibacillus genome sequences, sourced from the National Center for Biotechnology Information database, with a focused investigation involving 89 of these genomes via antiSMASH. Our analysis unearthed a total of 848 BGCs, of which 716 (84.4%) were classified as unknown. From the initial pool of 554 Paenibacillus strains, we selected 26 available in culture collections for an in-depth evaluation. Genomic scrutiny of these selected strains unveiled 255 BGCs, encoding non-ribosomal peptide synthetases, polyketide synthases, and bacteriocins, with 221 (86.7%) classified as unknown. Among these strains, 20 exhibited antimicrobial activity against the gram-positive bacterium Micrococcus luteus, yet only six strains displayed activity against the gram-negative bacterium Escherichia coli. We proceeded to focus on Paenibacillus brasilensis, which featured five new BGCs for further investigation. To facilitate detailed characterization, we constructed a mutant in which a single BGC encoding a novel antibiotic was activated while simultaneously inactivating multiple BGCs using a cytosine base editor (CBE). The novel antibiotic was found to be localized to the cell wall and demonstrated activity against both gram-positive bacteria and fungi. The chemical structure of the new antibiotic was elucidated on the basis of ESIMS, 1D and 2D NMR spectroscopic data. The novel compound, with a molecular weight of 926, was named bracidin. CONCLUSIONS: This study outcome highlights the potential of Paenibacillus species as valuable sources for novel antibiotics. In addition, CBE-mediated dereplication of antibiotics proved to be a rapid and efficient method for characterizing novel antibiotics from Paenibacillus species, suggesting that it will greatly accelerate the genome-based development of new antibiotics.


Subject(s)
Anti-Bacterial Agents , Genome, Bacterial , Multigene Family , Paenibacillus , Paenibacillus/genetics , Paenibacillus/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/biosynthesis , Peptide Synthases/genetics , Polyketide Synthases/genetics , Bacteriocins/genetics , Bacteriocins/pharmacology , Bacteriocins/biosynthesis , Biosynthetic Pathways/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Discovery/methods
7.
Appl Microbiol Biotechnol ; 108(1): 370, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38861018

ABSTRACT

Members of the genus Lysinibacillus attract attention for their mosquitocidal, bioremediation, and plant growth-promoting abilities. Despite this interest, comprehensive studies focusing on genomic traits governing plant growth and stress resilience in this genus using whole-genome sequencing are still scarce. Therefore, we sequenced and compared the genomes of three endophytic Lysinibacillus irui strains isolated from Canary Island date palms with the ex-type strain IRB4-01. Overall, the genomes of these strains consist of a circular chromosome with an average size of 4.6 Mb and a GC content of 37.2%. Comparative analysis identified conserved gene clusters within the core genome involved in iron acquisition, phosphate solubilization, indole-3-acetic acid biosynthesis, and volatile compounds. In addition, genome analysis revealed the presence of genes encoding carbohydrate-active enzymes, and proteins that confer resistance to oxidative, osmotic, and salinity stresses. Furthermore, pathways of putative novel bacteriocins were identified in all genomes. This illustrates possible common plant growth-promoting traits shared among all strains of L. irui. Our findings highlight a rich repertoire of genes associated with plant lifestyles, suggesting significant potential for developing inoculants to enhance plant growth and resilience. This study is the first to provide insights into the overall genomic signatures and mechanisms of plant growth promotion and biocontrol in the genus Lysinibacillus. KEY POINTS: • Pioneer study in elucidating plant growth promoting in L. irui through comparative genomics. • Genome mining identified biosynthetic pathways of putative novel bacteriocins. • Future research directions to develop L. irui-based biofertilizers for sustainable agriculture.


Subject(s)
Bacillaceae , Genome, Bacterial , Genomics , Bacillaceae/genetics , Bacillaceae/metabolism , Base Composition , Multigene Family , Arecaceae/microbiology , Plant Development , Whole Genome Sequencing , Bacteriocins/genetics , Bacteriocins/metabolism , Bacteriocins/biosynthesis , Phylogeny , Plant Growth Regulators/metabolism , Indoleacetic Acids/metabolism , Stress, Physiological
8.
Gut Microbes ; 16(1): 2369338, 2024.
Article in English | MEDLINE | ID: mdl-38899682

ABSTRACT

Gut bacteria are known to produce bacteriocins to inhibit the growth of other bacteria. Consequently, bacteriocins have attracted increased attention as potential microbiome-editing tools. In this study we examine the inhibitory spectrum of 75 class II bacteriocins against 48 representative gut microbiota species. The bacteriocins were heterologously expressed in Escherichia coli and evaluated in vitro, ex vivo and in vivo. In vitro assays revealed 22 bacteriocins to inhibit at least one species and showed selective inhibition patterns against species implicated in certain disorders and diseases. Three bacteriocins were selected for ex vivo assessment on mouse feces. Based on 16S rRNA sequencing of the cultivated feces we showed that the two bacteriocins: Actifencin (#13) and Bacteroidetocin A (#22) selectively inhibited the growth of Lactobacillus and Bacteroides, respectively. The probiotic: E. coli Nissle 1917 was engineered to express these two bacteriocins in mice. However, the selective inhibitory patterns found in the in vitro and ex vivo experiments could not be observed in vivo. Our study describes a methodology for heterologous high throughput bacteriocin expression and screening and elucidates the inhibitory patterns of class II bacteriocins on the gut microbiota.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Escherichia coli , Feces , Gastrointestinal Microbiome , Bacteriocins/genetics , Bacteriocins/pharmacology , Bacteriocins/metabolism , Bacteriocins/biosynthesis , Animals , Mice , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/biosynthesis , RNA, Ribosomal, 16S/genetics , Lactobacillus/genetics , Lactobacillus/metabolism , Lactobacillus/drug effects , Bacteria/genetics , Bacteria/drug effects , Bacteria/metabolism , Bacteria/classification , Gene Expression
9.
Genomics ; 116(3): 110855, 2024 05.
Article in English | MEDLINE | ID: mdl-38703968

ABSTRACT

Clostridium butyricum is a Gram-positive anaerobic bacterium known for its ability to produce butyate. In this study, we conducted whole-genome sequencing and assembly of 14C. butyricum industrial strains collected from various parts of China. We performed a pan-genome comparative analysis of the 14 assembled strains and 139 strains downloaded from NCBI. We found that the genes related to critical industrial production pathways were primarily present in the core and soft-core gene categories. The phylogenetic analysis revealed that strains from the same clade of the phylogenetic tree possessed similar antibiotic resistance and virulence factors, with most of these genes present in the shell and cloud gene categories. Finally, we predicted the genes producing bacteriocins and botulinum toxins as well as CRISPR systems responsible for host defense. In conclusion, our research provides a desirable pan-genome database for the industrial production, food application, and genetic research of C. butyricum.


Subject(s)
Clostridium butyricum , Genome, Bacterial , Phylogeny , Clostridium butyricum/genetics , Clostridium butyricum/metabolism , Whole Genome Sequencing , Bacteriocins/genetics , Bacteriocins/biosynthesis , Industrial Microbiology , Botulinum Toxins/genetics , Virulence Factors/genetics
10.
Microbiol Spectr ; 12(7): e0374823, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38780256

ABSTRACT

The lasso peptide microcin J25 (MccJ25) possesses strong antibacterial properties and is considered a potential effective component of bacterial disease treatment drugs and safe food preservatives. Although MccJ25 can be heterologously expressed in Bacillus subtilis as we have previously reported, its regulation and accumulation are yet to be understood. Here, we investigated the expression level and stability of MccJ25 in B. subtilis strains with disruption in peptidase genes pepA, pepF, and pepT. Oligoendopeptidase F (PepF) was found to be involved in reduction of the production of MccJ25 by degradation of its precursor peptide. In the pepF mutant, the MccJ25 reached a concentration of 1.68 µM after a cultivation time exceeding 60 hours, while the wild-type strain exhibited a concentration of only 0.14 µM. Moreover, the production of MccJ25 in B. subtilis downregulated the genes associated with sporulation, and this may contribute to its accumulation. Finally, this study provides a strategy to improve the stability and production of MccJ25 in B. subtilis. IMPORTANCE: MccJ25 displays significant antibacterial activity, a well-defined mode of action, exceptional safety, and remarkable stability. Hence, it presents itself as a compelling candidate for an optimal antibacterial or anti-endotoxin medication. The successful establishment of exogenous production of MccJ25 in Bacillus subtilis provides a strategy for reducing its production cost and diversifying its utilization. In this study, we have provided evidence indicating that both peptidase PepF and sporulation are significant factors that limit the expression of MccJ25 in B. subtilis. The ΔpepF and ΔsigF mutants of B. subtilis express MccJ25 with higher production yield and enhanced stability. To sum up, this study developed several better engineered strains of B. subtilis, which greatly reduced the consumption of MccJ25 during the nutrient depletion stage of the host strain, improved its production, and elucidated factors that may be involved in reducing MccJ25 accumulation in B. subtilis.


Subject(s)
Anti-Bacterial Agents , Bacillus subtilis , Bacterial Proteins , Bacteriocins , Spores, Bacterial , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacillus subtilis/growth & development , Bacteriocins/metabolism , Bacteriocins/genetics , Bacteriocins/biosynthesis , Spores, Bacterial/genetics , Spores, Bacterial/growth & development , Spores, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/biosynthesis , Gene Expression Regulation, Bacterial , Peptide Hydrolases/metabolism , Peptide Hydrolases/genetics
11.
Probiotics Antimicrob Proteins ; 16(4): 1130-1147, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38743207

ABSTRACT

Enterococcus faecalis CAUM157 (KACC 81148BP), a Gram-positive bacteria isolated from raw cow's milk, was studied for its bacteriocin production. The antimicrobial activity of CAUM157 was attributed to a two-peptide class IIb bacteriocin with potent activity against food-borne pathogen Listeria monocytogenes and periodontal disease-causing pathogens (Prevotella intermedia KCTC 15693 T and Fusobacterium nucleatum KCTC 2488 T). M157 bacteriocins exhibit high temperature and pH stability and resist hydrolytic enzyme degradation and detergent denaturation, potentially due to their structural conformation. Based on amino acid sequence, M157A and M157B were predicted to be 5.176 kDa and 5.182 kDa in size, respectively. However, purified bacteriocins and chemically synthesized N-formylated M157 peptides both showed 5.204 kDa (M157A) and 5.209 kDa (M157B) molecular mass, confirming the formylation of the N-terminal methionine of both peptides produced by strain CAUM157. Furthermore, the strain demonstrated favorable growth and fermentation with minimal bacteriocin production when cultured in whey-based media, whereas a 1.0% tryptone or soytone supplementation resulted in higher bacteriocin production. Although Ent. faecalis CAUM157 innately harbors genes for virulence factors and antimicrobial resistance (e.g., tetracycline and erythromycin), its bacteriocin production is valuable in circumventing the need for live microorganisms, particularly in food applications for pathogen control.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Enterococcus faecalis , Listeria monocytogenes , Bacteriocins/pharmacology , Bacteriocins/chemistry , Bacteriocins/biosynthesis , Enterococcus faecalis/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Listeria monocytogenes/drug effects , Animals , Cattle , Milk/microbiology
12.
Probiotics Antimicrob Proteins ; 16(4): 1411-1426, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38252201

ABSTRACT

Clostridium tyrobutyricum strain NRRL B-67062 was previously isolated from an ethanol production facility and shown to produce high yields of butyric acid. In addition, the cell-free supernatant of the fermentation broth from NRRL B-67062 contained antibacterial activity against certain Gram-positive bacteria. To determine the source of this antibacterial activity, we report the genome and genome mining of this strain. The complete genome of NRRL B-67062 showed one circular chromosome of 3,242,608 nucleotides, 3114 predicted coding sequences, 79 RNA genes, and a G+C content of 31.0%. Analyses of the genome data for genes potentially associated with antimicrobial features were sought after by using BAGEL-4 and anti-SMASH databases. Among the leads, a polypeptide of 66 amino acids (PEG 446) contains the DUF4177 domain, which is an uncharacterized highly conserved domain (pfam13783). The cloning and expression of the peg446 gene in Escherichia coli and Bacillus subtilis confirmed the antibacterial property against Lactococcus lactis LM 0230, Limosilactobacillus fermentum 0315-25, and Listeria innocua NRRL B-33088 by gel overlay and well diffusion assays. Molecular modeling suggested that PEG 446 contains one alpha-helix and three anti-parallel short beta-sheets. These results will aid further functional studies and facilitate simultaneously fermentative production of both butyric acid and a putative bacteriocin from agricultural waste and lignocellulosic biomass materials.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Clostridium tyrobutyricum , Clostridium tyrobutyricum/genetics , Clostridium tyrobutyricum/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/biosynthesis , Bacteriocins/genetics , Bacteriocins/biosynthesis , Bacteriocins/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Limosilactobacillus fermentum/genetics , Limosilactobacillus fermentum/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Genome, Bacterial , Escherichia coli/genetics
13.
J Dairy Sci ; 107(6): 3468-3477, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38246535

ABSTRACT

Ice cream manufacture commonly results in the accumulation of wasted product that contains valuable food-grade quality components, including fat, carbohydrates, and protein. Methods have been developed for recovering the fat from this waste stream, but this results in the generation of a co-product rich in fermentable carbohydrates. This study aimed to investigate the potential for using this co-product as a fermentation substrate for production of antimicrobial peptides, called bacteriocins, by dairy starter cultures. Results showed that Streptococcus thermophilus B59671 and Lactococcus lactis 11454 produced the broad-spectrum bacteriocins thermophilin 110 and nisin, respectively, when the fermentation substrate was melted ice cream, or a co-product generated by a modified butter churning technique. Bacteriocin production varied depending on the brand and variety of vanilla ice cream used in this study. When an alternate enzyme-assisted fat extraction technique was used, S. thermophilus metabolism was impaired within the resulting co-product, and thermophilin 110 production was not observed. Lactococcus lactis was still able to grow in this co-product, but antimicrobial activity was not observed. Results from this study suggest the co-product generated when using the churning technique is a better choice to use as a base medium for future studies to optimize bacteriocin production.


Subject(s)
Bacteriocins , Fermentation , Ice Cream , Lactobacillales , Bacteriocins/metabolism , Bacteriocins/biosynthesis , Lactobacillales/metabolism , Streptococcus thermophilus/metabolism , Lactococcus lactis/metabolism
14.
Arch Microbiol ; 204(6): 344, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35596088

ABSTRACT

Thurincin H is a bacteriocin produced by Bacillus thuringiensis, it is encoded in a group of ten genes, most of which have been characterized experimentally or by homology. However, the activity of the thnI gene encoding a 95 amino acid ORF remains unknown. In this work, the thnI gene was cloned under the regulation of two promoters and transformed into a sensitive strain to determine if it acts as an immunity protein. In addition, a deletion mutant without the thnI gene was used to test whether thnI is required or not for the biosynthesis of thurincin H. It was concluded that thnI does not provide immunity and is not required to produce thurincin H.


Subject(s)
Bacillus thuringiensis , Bacteriocins , Bacillus thuringiensis/chemistry , Bacterial Proteins/metabolism , Bacteriocins/biosynthesis , Promoter Regions, Genetic
15.
Angew Chem Int Ed Engl ; 61(6): e202114414, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34889011

ABSTRACT

Lanthipeptides are a group of ribosomally synthesized and post-translationally modified peptides with diverse structural features and bioactivities. Gut-microbiota-derived lanthipeptides play important roles in gut homeostasis of the host. Herein, we report the discovery and biosynthesis of class III lantibiotics named amylopeptins, which are derived from the gut microbiota of Sprague-Dawley rats and display a narrow antimicrobial spectrum. In contrast to known class III lanthipeptides, the biosynthesis of amylopeptins employs AmyP, which belongs to a subgroup of S8 family serine proteases, to remove the leader of corresponding precursor peptides in a site-specific manner during the last step of their maturation. Overall, this study shows for the first time that S8 family proteases participate in the biosynthesis of class III lanthipeptides.


Subject(s)
Bacillus amyloliquefaciens/genetics , Bacteriocins/biosynthesis , Peptide Hydrolases/metabolism , Animals , Bacillus amyloliquefaciens/metabolism , Bacteriocins/chemistry , Bacteriocins/genetics , Gastrointestinal Microbiome , Peptide Hydrolases/genetics , Rats , Rats, Sprague-Dawley
16.
PLoS Genet ; 17(11): e1009919, 2021 11.
Article in English | MEDLINE | ID: mdl-34847155

ABSTRACT

Antimicrobial resistance is one of the major threats to Public Health worldwide. Understanding the transfer and maintenance of antimicrobial resistance genes mediated by mobile genetic elements is thus urgent. In this work, we focus on the ColE1-like plasmid family, whose distinctive replication and multicopy nature has given rise to key discoveries and tools in molecular biology. Despite being massively used, the hosts, functions, and evolutionary history of these plasmids remain poorly known. Here, we built specific Hidden Markov Model (HMM) profiles to search ColE1 replicons within genomes. We identified 1,035 ColE1 plasmids in five Orders of γ-Proteobacteria, several of which are described here for the first time. The phylogenetic analysis of these replicons and their characteristic MOBP5/HEN relaxases suggest that ColE1 plasmids have diverged apart, with little transfer across orders, but frequent transfer across families. Additionally, ColE1 plasmids show a functional shift over the last decades, losing their characteristic bacteriocin production while gaining several antimicrobial resistance genes, mainly enzymatic determinants and including several extended-spectrum betalactamases and carbapenemases. Furthermore, ColE1 plasmids facilitate the intragenomic mobilization of these determinants, as various replicons were identified co-integrated with large non-ColE1 plasmids, mostly via transposases. These results illustrate how families of plasmids evolve and adapt their gene repertoires to bacterial adaptive requirements.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacteriocins/biosynthesis , Drug Resistance, Bacterial/genetics , Evolution, Molecular , Gammaproteobacteria/genetics , Genes, Bacterial , Plasmids , Gammaproteobacteria/drug effects , Markov Chains , Phylogeny
17.
Probiotics Antimicrob Proteins ; 13(6): 1820-1832, 2021 12.
Article in English | MEDLINE | ID: mdl-34423377

ABSTRACT

Bacteriocins produced by lactic acid bacteria have potential use as natural food preservatives, which may alleviate current problems associated with the overuse of antibiotics and emerging multi-drug-resistant microbes. In this work, Lactiplantibacillus plantarum RUB1 was found to produce a class IIb bacteriocin with strong antibacterial activity. Except for plnXY encoding putative proteins, L. plantarum RUB1 contains most genes in five operons (plnABCD, plnGHSTUVW, plnMNOP, plnIEF, and plnRLJK) related to bacteriocin synthesis. Adding low (100 and 500 ng/mL) and medium (1 µg/mL) concentrations of PlnA to broth promoted bacteriocin production and upregulated bacteriocin gene plnA, while high concentrations (50 and 200 µg/mL) inhibited expression of these genes. Co-culturing L. plantarum RUB1 with Enterococcus hirae 1003, Enterococcus hirae LWS, Limosilactobacillus fermentum RC4, L. plantarum B6, and even Listeria monocytogenes ATCC 19111 and Staphylococcus aureus ATCC 6538 enhanced bacteriocin activity and expression of bacteriocin-related genes. This study verifies that PlnA can indeed upregulate the expression of bacteriocin genes, and also bacteriocin production can be induced by co-culture with some specific bacteria or their cell-free supernatants. Bacteriocin production by L. plantarum RUB1 is mediated by a quorum sensing mechanism, directly influenced by autoinducing peptide or specific strains. The findings provide new methods and insight into bacteriocin production mechanisms.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Lactobacillus plantarum , Anti-Bacterial Agents/pharmacology , Bacteriocins/biosynthesis , Bacteriocins/pharmacology , Enterococcus hirae/drug effects , Quorum Sensing , Staphylococcus aureus/drug effects
18.
Int J Mol Sci ; 22(16)2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34445321

ABSTRACT

Listeria monocytogenes is an important food-borne pathogen and a serious concern to food industries. Bacteriocins are antimicrobial peptides produced naturally by a wide range of bacteria mostly belonging to the group of lactic acid bacteria (LAB), which also comprises many strains used as starter cultures or probiotic supplements. Consequently, multifunctional strains that produce bacteriocins are an attractive approach to combine a green-label approach for food preservation with an important probiotic trait. Here, a collection of bacterial isolates from raw cow's milk was typed by 16S rRNA gene sequencing and MALDI-Biotyping and supernatants were screened for the production of antimicrobial compounds. Screening was performed with live Listeria monocytogenes biosensors using a growth-dependent assay and pHluorin, a pH-dependent protein reporting membrane damage. Purification by cation exchange chromatography and further investigation of the active compounds in supernatants of two isolates belonging to the species Pediococcus acidilactici and Lactococcus garvieae suggest that their antimicrobial activity is related to heat-stable proteins/peptides that presumably belong to the class IIa bacteriocins. In conclusion, we present a pipeline of methods for high-throughput screening of strain libraries for potential starter cultures and probiotics producing antimicrobial compounds and their identification and analysis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriocins/pharmacology , Drug Discovery/methods , Listeria monocytogenes/drug effects , Probiotics , Animals , Anti-Bacterial Agents/biosynthesis , Bacteriocins/biosynthesis , Lactococcus/isolation & purification , Lactococcus/metabolism , Microbiota , Milk/microbiology , Pediococcus acidilactici/isolation & purification , Pediococcus acidilactici/metabolism
19.
mBio ; 12(4): e0121921, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34281399

ABSTRACT

A membrane-associated lanthipeptide synthetase complex, consisting of the dehydratase NisB, the cyclase NisC, and the ABC transporter NisT, has been described for nisin biosynthesis in the coccoid bacterium Lactococcus lactis. Here, we used advanced fluorescence microscopy to visualize the functional nisin biosynthesis machinery in rod-shaped cells and analyzed its spatial distribution and dynamics employing a platform we developed for heterologous production of nisin in Bacillus subtilis. We observed that NisT, as well as NisB and NisC, were all distributed in a punctate pattern along the cell periphery, opposed to the situation in coccoid cells. NisBTC proteins were found to be highly colocalized, being visualized at the same spots by dual fluorescence microscopy. In conjunction with the successful isolation of the biosynthetic complex NisBTC from the cell membrane, this corroborated that the visual bright foci were the sites for nisin maturation and transportation. A strategy of differential timing of expression was employed to demonstrate the in vivo dynamic assembly of NisBTC, revealing the recruitment by NisT of NisBC to the membrane. Additionally, by use of mutated proteins, the nucleotide binding domain (NBD) of NisT was found to function as a membrane anchor for NisB and/or NisC. We also show that the nisin biosynthesis sites are static and likely associated with proteins residing in lipid rafts. Based on these data, we propose a model for a three-phase production of modified precursor nisin in rod-shaped bacteria, presenting the assembly dynamics of NisBTC and emphasizing the crucial role of NisBC, next to NisT, in the process of precursor nisin translocation. IMPORTANCE Nisin is a model antimicrobial peptide for LanBC-modified lantibiotics that are modified and transported by a membrane synthetase complex. Although the subcellular localization and the assembly process of such a complex in L. lactis have been described in our recent work (J. Chen, A. J. van Heel, and O. P. Kuipers, mBio 11:e02825-20, 2020, https://doi.org/10.1128/mBio.02825-20), it proved difficult to gain a more detailed insight into the exact LanBTC assembly in the L. lactis system. Rod-shaped cells, especially B. subtilis, are better suited to study the assembly dynamics of these protein complexes. In this work, we present evidence for the existence of the lanthipeptide biosynthetic complex by visualizing and isolating the machinery in vivo. The dynamic behavior of the modification machinery and the transporter within the cells was characterized in depth, revealing the dependence of first LanB and LanC on each other and subsequent recruitment of them by LanT during the machinery assembly. Importantly, the elucidation of the dynamic assembly of the complex will facilitate future studies of lanthipeptide transport mechanisms and the structural characterization of the complete complex.


Subject(s)
Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Biosynthetic Pathways , Nisin/biosynthesis , Nisin/genetics , Antimicrobial Peptides/biosynthesis , Antimicrobial Peptides/genetics , Antimicrobial Peptides/metabolism , Bacterial Proteins/genetics , Bacteriocins/biosynthesis , Bacteriocins/genetics , Bacteriocins/metabolism , Biosynthetic Pathways/genetics , Biosynthetic Pathways/physiology , Microscopy, Fluorescence/methods , Nisin/analysis
20.
J Microbiol ; 59(6): 573-583, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33990913

ABSTRACT

Pediococcus acidilactici is a reliable bacteriocin producer and a promising probiotic species with wide application in the food and health industry. However, the underlying genetic features of this species have not been analyzed. In this study, we performed a comprehensive comparative genomic analysis of 41 P. acidilactici strains from various ecological niches. The bacteriocin production of 41 strains were predicted and three kinds of bacteriocin encoding genes were identified in 11 P. acidilactici strains, namely pediocin PA-1, enterolysin A, and colicin-B. Moreover, whole-genome analysis showed a high genetic diversity within the population, mainly related to a large proportion of variable genomes, mobile elements, and hypothetical genes obtained through horizontal gene transfer. In addition, comparative genomics also facilitated the genetic explanation of the adaptation for host environment, which specify the protection mechanism against the invasion of foreign DNA (i.e. CRISPR/Cas locus), as well as carbohydrate fermentation. The 41 strains of P. acidilactici can metabolize a variety of carbon sources, which enhances the adaptability of this species and survival in different environments. This study evaluated the antibacterial ability, genome evolution, and ecological flexibility of P. acidilactici from the perspective of genetics and provides strong supporting evidence for its industrial development and application.


Subject(s)
Genome, Bacterial , Pediococcus acidilactici/genetics , Anti-Bacterial Agents/biosynthesis , Bacteriocins/biosynthesis , Genomics , Pediococcus acidilactici/classification , Pediococcus acidilactici/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL