Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.751
Filter
1.
Nat Commun ; 15(1): 7057, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152109

ABSTRACT

Due to envelope differences between Gram-positive and Gram-negative bacteria, engineering precision bactericidal contractile nanomachines requires atomic-level understanding of their structures; however, only those killing Gram-negative bacteria are currently known. Here, we report the atomic structures of an engineered diffocin, a contractile syringe-like molecular machine that kills the Gram-positive bacterium Clostridioides difficile. Captured in one pre-contraction and two post-contraction states, each structure fashions six proteins in the bacteria-targeting baseplate, two proteins in the energy-storing trunk, and a collar linking the sheath with the membrane-penetrating tube. Compared to contractile machines targeting Gram-negative bacteria, major differences reside in the baseplate and contraction magnitude, consistent with target envelope differences. The multifunctional hub-hydrolase protein connects the tube and baseplate and is positioned to degrade peptidoglycan during penetration. The full-length tape measure protein forms a coiled-coil helix bundle homotrimer spanning the entire diffocin. Our study offers mechanical insights and principles for designing potent protein-based precision antibiotics.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Clostridioides difficile , Bacteriocins/chemistry , Bacteriocins/metabolism , Bacteriocins/pharmacology , Clostridioides difficile/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Models, Molecular , Gram-Positive Bacteria/drug effects , Peptidoglycan/metabolism , Peptidoglycan/chemistry , Crystallography, X-Ray
2.
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
3.
World J Microbiol Biotechnol ; 40(10): 290, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39102120

ABSTRACT

Strain Lactiplantibacillus plantarum D1 with bacteriocin producing ability was found in the intestine of Gambusia affinis. The bacteriocin was found to have high inhibitory activity against multiple Streptococcus species and several other Gram-positive and Gram-negative bacteria. Bacteriocin was purified from culture supernatant by ion-exchange chromatography, Sep-Pak C18 cartridge, and reverse-phase high-performance liquid chromatography (RP-HPLC). Matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectral analysis determined that purified bacteriocin has a molecular mass of 2,731 Da. A partial N-terminal sequence KRKKHKXQIYNNGM was obtained from the Edman analysis. The N-terminal sequence was employed to search against a translation of the draft genome of strain D1. The translated full amino acid sequence of the mature peptide is as follows: NH2- KRKKHKCQIYNNGMPTGQYRWC, which has a molecular weight of 2738 Da. A BLAST search revealed that this bacteriocin was most similar to bactofencin A but differed from it with three amino acid residues. No identical peptide or protein has been previously reported, and this peptide, termed bactofencin YH, was therefore considered to be a new bacteriocin produced by Lactiplantibacillus plantarum D1.


Subject(s)
Amino Acid Sequence , Anti-Bacterial Agents , Bacteriocins , Molecular Weight , Streptococcus , Bacteriocins/pharmacology , Bacteriocins/chemistry , Bacteriocins/isolation & purification , Bacteriocins/metabolism , Streptococcus/drug effects , Streptococcus/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Microbial Sensitivity Tests , Animals , Chromatography, High Pressure Liquid , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects
4.
World J Microbiol Biotechnol ; 40(10): 307, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39162916

ABSTRACT

Antimicrobial resistance poses a significant threat to humanity, and the development of new antibiotics is urgently needed. Our research has focused on thiopeptide antibiotics such as micrococcin P2 (MP2) and derivatives thereof as new anti-infective agents. Thiopeptides are sulfur-rich, structurally complex substances that exhibit potent activity against Gram-positive pathogens and Mycobacteria species, including clinically resistant strains. The clinical development of thiopeptides has been hampered by the lack of efficient synthetic platforms to conduct detailed structure-activity relationship studies of these natural products. The present contribution touches upon efficient synthetic routes to MP2 that laid the groundwork for clinical translation. The medicinal chemistry campaign on MP2 has been guided by computational molecular dynamic simulations and parallel investigations to improve drug-like properties, such as enhancing the aqueous solubility and optimizing antibacterial activity. Such endeavors have enabled identification of promising lead compounds, AJ-037 and AJ-206, against Mycobacterium avium complex (MAC). Extensive in vitro studies revealed that these compounds exert potent activity against MAC species, a subspecies of non-tuberculous mycobacteria (NTM) that proliferate inside macrophages. Two additional pre-clinical candidates have been identified: AJ-024, for the treatment of Clostridioides difficile infections, and AJ-147, for methicillin-resistant Staphylococcus aureus impetigo. Both compounds compare quite favorably with current first-line treatments. In particular, the ability of AJ-147 to downregulate pro-inflammatory cytokines adds a valuable dimension to its clinical use. In light of above, these new thiopeptide derivatives are well-poised for further clinical development.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bacteriocins/pharmacology , Bacteriocins/chemistry , Humans , Structure-Activity Relationship , Molecular Dynamics Simulation , Peptides/pharmacology , Peptides/chemistry , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Clostridioides difficile/drug effects
5.
Gut Microbes ; 16(1): 2387139, 2024.
Article in English | MEDLINE | ID: mdl-39106231

ABSTRACT

Bacteriocins are broad or narrow-spectrum antimicrobial compounds that have received significant scientific attention due to their potential to treat infections caused by antibiotic-resistant pathogenic bacteria. The genome of Bifidobacterium pseudocatenulatum MM0196, an antimicrobial-producing, fecal isolate from a healthy pregnant woman, was shown to contain a gene cluster predicted to encode Pseudocin 196, a novel lantibiotic, in addition to proteins involved in its processing, transport and immunity. Following antimicrobial assessment against various indicator strains, protease-sensitive Pseudocin 196 was purified to homogeneity from cell-free supernatant. MALDI TOF mass spectrometry confirmed that the purified antimicrobial compound corresponds to a molecular mass of 2679 Da, which is consistent with that deduced from its genetic origin. Pseudocin 196 is classified as a lantibiotic based on its similarity to lacticin 481, a lanthionine ring-containing lantibiotic produced by Lactococcus lactis. Pseudocin 196, the first reported bacteriocin produced by a B. pseudocatenulatum species of human origin, was shown to inhibit clinically relevant pathogens, such as Clostridium spp. and Streptococcus spp. thereby highlighting the potential application of this strain as a probiotic to treat and prevent bacterial infections.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Bifidobacterium , Bacteriocins/pharmacology , Bacteriocins/genetics , Bacteriocins/metabolism , Bacteriocins/chemistry , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bifidobacterium/genetics , Bifidobacterium/drug effects , Bifidobacterium/metabolism , Female , Clostridium/genetics , Clostridium/drug effects , Clostridium/metabolism , Feces/microbiology , Streptococcus/drug effects , Streptococcus/genetics , Streptococcus/metabolism , Pregnancy , Multigene Family , Microbial Sensitivity Tests , Genome, Bacterial , Probiotics/pharmacology
6.
Appl Environ Microbiol ; 90(8): e0084524, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39078127

ABSTRACT

Campylobacter jejuni (C. jejuni) is one of the most common causes of foodborne infections worldwide and a major contributor to diarrheal diseases. This study aimed to explore the ability of commensal gut bacteria to control C. jejuni infection. Bacterial strains from the intestinal mucosa of broilers were screened in vitro against C. jejuni ATCC BAA1153. The cell-free supernatant (CFS) of Ligilactobacillus salivarius UO.C249 showed potent dose-dependent antimicrobial activity against the pathogen, likely due to the presence of bacteriocin-like moieties, as confirmed by protease treatment. Genome and exoproteome analyses revealed the presence of known bacteriocins, including Abp118. The genome of Lg. salivarius UO.C249 harbors a 1.8-Mb chromosome and a 203-kb megaplasmid. The strain was susceptible to several antibiotics and had a high survival rate in the simulated chicken gastrointestinal tract (GIT). Post-protease treatment revealed residual inhibitory activity, suggesting alternative antimicrobial mechanisms. Short-chain fatty acid (SCFA) quantification confirmed non-inhibitory levels of acetic (24.4 ± 1.2 mM), isovaleric (34 ± 1.0 µM), and butyric (32 ± 2.5 µM) acids. Interestingly, extracellular vesicles (EVs) isolated from the CFS of Lg. salivarius UO.C249 were found to inhibit C. jejuni ATCC BAA-1153. Proteome profiling of these EVs revealed the presence of unique proteins distinct from bacteriocins identified in CFS. The majority of the identified proteins in EVs are located in the membrane and play roles in transmembrane transport and peptidoglycan degradation, peptidase, proteolysis, and hydrolysis. These findings suggest that although bacteriocins are a primary antimicrobial mechanism, EV production also contributes to the inhibitory activity of Lg. salivarius UO.C249 against C. jejuni. IMPORTANCE: Campylobacter jejuni (C. jejuni) is a major cause of gastroenteritis and a global public health concern. The increasing antibiotic resistance and lack of effective alternatives in livestock production pose serious challenges for controlling C. jejuni infections. Therefore, alternative strategies are needed to control this pathogen, especially in the poultry industry where it is prevalent and can be transmitted to humans through contaminated food products. In this study, Ligilactobacillus salivarius UO.C249 isolated from broiler intestinal mucosa inhibited C. jejuni and exhibited important probiotic features. Beyond bacteriocins, Lg. salivarius UO.C249 secretes antimicrobial extracellular vesicles (EVs) with a unique protein set distinct from bacteriocins that are involved in transmembrane transport and peptidoglycan degradation. Our findings suggest that beyond bacteriocins, EV production is also a distinct inhibitory signaling mechanism used by Lg. salivarius UO.C249 to control C. jejuni. These findings hold promise for the application of probiotic EVs for pathogen control.


Subject(s)
Bacteriocins , Campylobacter jejuni , Chickens , Extracellular Vesicles , Ligilactobacillus salivarius , Probiotics , Bacteriocins/pharmacology , Bacteriocins/metabolism , Bacteriocins/genetics , Probiotics/pharmacology , Campylobacter jejuni/drug effects , Campylobacter jejuni/genetics , Campylobacter jejuni/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/chemistry , Animals , Chickens/microbiology , Ligilactobacillus salivarius/physiology , Anti-Bacterial Agents/pharmacology , Campylobacter Infections/microbiology , Campylobacter Infections/veterinary , Campylobacter Infections/prevention & control
7.
Biotechnol Adv ; 75: 108415, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39033836

ABSTRACT

Glycosylated bacteriocins, known as glycocins, were first discovered in 2011. These bioactive peptides are produced by bacteria to gain survival advantages. They exhibit diverse types of glycans and demonstrate varied antimicrobial activity. Currently, there are 13 experimentally known glycocins, with over 250 identified in silico across different bacterial phyla. Notably, glycocins are recognized for their glycan-mediated antimicrobial activity, proving effective against drug-resistant and foodborne pathogens. Many glycocins contain rare S-linked glycans. Glycosyltransferases (GTs), responsible for transferring sugar to glycocins and involved in glycocin biosynthesis, often cluster together in the producer's genome. This clustering makes them valuable for custom glycoengineering with diverse substrate specificities. Heterologous expression of glycocins has paved the way for the establishment of microbial factories for glycopeptide and glycoconjugate production across various industries. In this review, we emphasize the primary roles of fully and partially characterized glycocins and their glycosylating enzymes. Additionally, we explore how specific glycan structures facilitate these functions in antibacterial activities. Furthermore, we discuss newer approaches and increasing efforts aimed at exploiting bacterial glycobiology for the development of food preservatives and as replacements or complements to traditional antibiotics, particularly in the face of antibiotic-resistant pathogenic bacteria.


Subject(s)
Glycosyltransferases , Glycosyltransferases/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/chemistry , Polysaccharides/chemistry , Polysaccharides/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bacteria/drug effects , Bacteria/metabolism , Glycosylation , Bacteriocins/chemistry , Bacteriocins/metabolism , Bacteriocins/pharmacology , Bacteriocins/genetics , Anti-Infective Agents/pharmacology , Anti-Infective Agents/metabolism , Anti-Infective Agents/chemistry
8.
Postepy Biochem ; 70(2): 266-278, 2024 07 01.
Article in Polish | MEDLINE | ID: mdl-39083475

ABSTRACT

Bacteriocins are peptides or proteins produced by bacteria to kill or inhibit the growth of other bacteria inhabiting the same ecological niche. The growing interest in bacteriocins reflects their potential use in food preservation and treatment of infections caused by antibiotic-resistant pathogenic bacteria, among other applications. The number of published studies on the identification of new bacteriocin-producing strains is constantly increasing. At the same time, there is a noticeable lack of research describing the mechanisms of action of most newly identified bacteriocins, as well as the mechanisms leading to the development of resistance to these bacteriocins and cross-resistance to antibiotics. Detailed understanding of these issues will allow to develop guidelines ensuring the most effective, safe and long-term use of bacteriocins without the risk of resistance development. This work describes the main assumptions of the doctoral dissertation of Aleksandra Tymoszewska, which objectives were to characterize the mechanisms of action and of resistance to class II bacteriocins of Gram-positive bacteria. Using the model bacterium Lactococcus lactis, two groups of bacteriocins were examined: (i) garvicins Q, A, B and C, and BacSJ; and (ii) aureocin A53 (AurA53)- and enterocin L50 (EntL50)-like bacteriocins. Bacteriocins of group (i) have been shown to recognize susceptible cells and form pores in the cell membrane using a specific receptor, the mannose-specific phosphotransferase system (Man-PTS), and sensitive bacteria have been shown to acquire resistance to the these bacteriocins by modifying the structure of Man-PTS. On the other hand, the acquisition of resistance to group (ii) membrane-targeting and receptor-independent bacteriocins occurs through changes in the structure of the bacterial cell wall and membrane, which are induced by changes in the expression of proteins involved in lipid metabolism or components of the YsaCB-KinG-LlrG regulatory system. The results shed new light on previous views on the mechanisms of action of bacteriocins and open up opportunities for their further study.


Subject(s)
Bacteriocins , Bacteriocins/pharmacology , Bacteriocins/metabolism , Gram-Positive Bacteria/drug effects , Drug Resistance, Bacterial , Anti-Bacterial Agents/pharmacology , Lactococcus lactis/metabolism
9.
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
10.
Biomolecules ; 14(7)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39062544

ABSTRACT

Cancer poses a severe threat to human health. Although conventional chemotherapy remains a cornerstone of cancer treatment, its significant side effects and the growing issue of drug resistance necessitate the urgent search for more efficient and less toxic anticancer drugs. In recent years, bacteriocins, antimicrobial peptides of microbial origin, have garnered significant attention due to their targeted antitumor activity. This unique activity is mainly attributed to their cationic and amphiphilic nature, which enables bacteriocins to specifically kill tumor cells without harming normal cells. When involving non-membrane-disrupting mechanisms, such as apoptosis induction, cell cycle blockade, and metastasis inhibition, the core mechanism of action is achieved by disrupting cell membranes, which endows bacteriocins with low drug resistance and high selectivity. However, the susceptibility of bacteriocins to hydrolysis and hemolysis in vivo limits their clinical application. To overcome these challenges, structural optimization of bacteriocins or their combination with nanotechnology is proposed for future development. This review aims to study the mechanism of action and current research status of bacteriocins as anticancer treatments, thus providing new insights for their clinical development and application.


Subject(s)
Antineoplastic Agents , Bacteriocins , Neoplasms , Bacteriocins/therapeutic use , Bacteriocins/pharmacology , Bacteriocins/chemistry , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Animals , Apoptosis/drug effects
11.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39000321

ABSTRACT

In the postantibiotic era, the pathogenicity and resistance of pathogens have increased, leading to an increase in intestinal inflammatory disease. Bacterial infections remain the leading cause of animal mortality. With increasing resistance to antibiotics, there has been a significant decrease in resistance to both inflammation and disease in animals, thus decreasing production efficiency and increasing production costs. These side effects have serious consequences and have detracted from the development of China's pig industry. Microcin C7 (McC7) demonstrates potent antibacterial activity against a broad spectrum of pathogens, stable physicochemical properties, and low toxicity, reducing the likelihood of resistance development. Thus, McC7 has received increasing attention as a potential clinical antibacterial and immunomodulatory agent. McC7 has the potential to serve as a new generation of antibiotic substitutes; however, its commercial applications in the livestock and poultry industry have been limited. In this review, we summarize and discuss the biosynthesis, biochemical properties, structural characteristics, mechanism of action, and immune strategies of McC7. We also describe the ability of McC7 to improve intestinal health. Our aim in this study was to provide a theoretical basis for the application of McC7 as a new feed additive or new veterinary drug in the livestock and poultry breeding industry, thus providing a new strategy for alleviating resistance through feed and mitigating drug resistance. Furthermore, this review provides insight into the new functions and anti-infection mechanisms of bacteriocin peptides and proposes crucial ideas for the research, product development, and application of bacteriocin peptides in different fields, such as the food and medical industries.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Bacteriocins/pharmacology , Bacteriocins/chemistry , Bacteriocins/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Animals , Immunomodulating Agents/pharmacology , Immunomodulating Agents/chemistry , Immunomodulating Agents/therapeutic use , Swine , Humans
12.
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
13.
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
14.
J Appl Microbiol ; 135(7)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38925659

ABSTRACT

AIMS: This study aimed to prospect and isolate lactic acid bacteria (LAB) from an artisanal cheese production environment, to assess their safety, and to explore their bacteriocinogenic potential against Listeria monocytogenes. METHODS AND RESULTS: Samples were collected from surfaces of an artisanal-cheese production facility and after rep-PCR and 16S rRNA sequencing analysis, selected strains were identified as to be belonging to Lactococcus garvieae (1 strain) and Enterococcus faecium (14 isolates, grouped into three clusters) associated with different environments (worktables, cheese mold, ripening wooden shelves). All of them presented bacteriocinogenic potential against L. monocytogenes ATCC 7644 and were confirmed as safe (γ-hemolytic, not presenting antibiotic resistance, no mucus degradation properties, and no proteolytic or gelatinase enzyme activity). Additionally, cell growth, acidification and bacteriocins production kinetics, bacteriocin stability in relation to different temperatures, pH, and chemicals were evaluated. According to performed PCR analysis all studied strains generated positive evidence for the presence of entA and entP genes (for production of enterocins A and enterocins P, respectively). However, pediocin PA-1 associated gene was recorded only in DNA obtained from E. faecium ST02JL and Lc. garvieae ST04JL. CONCLUSIONS: It is worth considering the application of these safe LAB or their bacteriocins in situ as an alternative means of controlling L. monocytogenes in cheese production environments, either alone or in combination with other antimicrobials.


Subject(s)
Bacteriocins , Cheese , Enterococcus faecium , Food Microbiology , Lactococcus , Listeria monocytogenes , Cheese/microbiology , Enterococcus faecium/genetics , Enterococcus faecium/isolation & purification , Enterococcus faecium/metabolism , Lactococcus/genetics , Lactococcus/isolation & purification , Bacteriocins/pharmacology , Brazil , Listeria monocytogenes/genetics , Listeria monocytogenes/drug effects , RNA, Ribosomal, 16S/genetics , Anti-Bacterial Agents/pharmacology
15.
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
16.
Appl Environ Microbiol ; 90(7): e0030024, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38832774

ABSTRACT

Staphylococcus aureus is a common bacterium on the skin and in the nose that sometimes causes severe illness. Bacteriocins, antimicrobial peptides, or proteins produced by bacteria are candidates for the treatment of S. aureus infection. In this study, we found that a clinical Staphylococcus epidermidis strain, KSE112, produced the lantibiotic Pep5, which showed anti-S. aureus activity. The complete nucleotide sequence of the Pep5-encoding plasmid was determined. Several S. aureus two-component regulatory systems (TCSs) are known to be involved in bacteriocin susceptibility. Therefore, susceptibility tests were performed using TCS-inactivated S. aureus mutants to determine which TCS is responsible for Pep5 susceptibility; the ΔgraRS mutant exhibited increased susceptibility to Pep5, while the ΔsrrAB mutant exhibited decreased susceptibility. GraRS is known to regulate dltABCD and mprF in concert with vraFG, and Pep5 susceptibility was significantly increased in the ΔdltABCD, ΔmprF, and ΔvraFG mutants. Regarding the ΔsrrAB mutant, cross-resistance to aminoglycosides was observed. As aminoglycoside activity is known to be affected by aerobic respiration, we focused on qoxABCD and cydAB, which are quinol oxidase genes that are necessary for aerobic respiration and have downregulated the expression in the ΔsrrAB mutant. We constructed ΔqoxABCD and ΔcydAB mutants and found that qoxABCD inactivation decreased susceptibility to Pep5 and aminoglycosides. These results indicate that reduced aerobic respiration due to the reduced qoxABCD expression in the ΔsrrAB mutant decreased Pep5 activity.IMPORTANCEThe emergence of drug-resistant bacteria, including MRSA, is a severe health problem worldwide. Thus, the development of novel antimicrobial agents, including bacteriocins, is needed. In this report, we found a Pep5-producing strain with anti-S. aureus activity. We determined the complete sequence of the Pep5-encoding plasmid for the first time. However, in S. aureus, GraRS and its effectors conferred decreased susceptibility to Pep5. We also revealed that another TCS, SrrAB, affects susceptibility Pep5 and other lantibiotics by controlling aerobic respiration. In our study, we investigated the efficacy of Pep5 against S. aureus and other Gram-positive bacteria and revealed that respiratory constancy regulated by TCS is required for the antimicrobial activity of nisin, nukacin, and Pep5. These findings provide important information for the clinical application of bacteriocins and suggest that they have different properties among similar pore-forming lantibiotics.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Bacteriocins , Staphylococcus aureus , Staphylococcus epidermidis , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/drug effects , Bacteriocins/pharmacology , Bacteriocins/genetics , Bacteriocins/metabolism , Anti-Bacterial Agents/pharmacology , Staphylococcal Infections/microbiology , Microbial Sensitivity Tests , Humans , Gene Expression Regulation, Bacterial , Repressor Proteins
17.
Int J Biol Macromol ; 274(Pt 1): 133047, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38857722

ABSTRACT

Bacterial cellulose (BC) has been found extensive applications in diverse domains for its exceptional attributes. However, the lack of antibacterial properties hampers its utilization in food and biomedical sectors. Leucocin, a bacteriocin belonging to class IIa, is synthesized by Leuconostoc that demonstrates potent efficacy against the foodborne pathogen, Listeria monocytogenes. In the current study, co-culturing strategy involving Kosakonia oryzendophytica FY-07 and Leuconostoc carnosum 4010 was used to confer anti-listerial activity to BC, which resulted in the generation of leucocin-containing BC (BC-L). The physical characteristics of BC-L, as determined by X-ray diffraction (XRD), Fourier-transform infrared spectroscopy (FTIR), and thermogravimetric analysis (TGA), were similar to the physical characteristics of BC. Notably, the experimental results of disc diffusion and growth curve indicated that the BC-L film exhibited a potent inhibitory effect against L. monocytogenes. Scanning electron microscopy (SEM) showed that BC-L exerts its bactericidal activity by forming pores on the bacterial cell wall. Despite the BC-L antibacterial mechanism, which involves pore formation, the mammalian cell viability remained unaffected by the BC-L film. The measurement results of zeta potential indicated that the properties of BC changed after being loaded with leucocin. Based on these findings, the anti-listerial BC-L generated through this co-culture system holds promise as a novel effective antimicrobial agent for applications in meat product preservation and packaging.


Subject(s)
Anti-Bacterial Agents , Cellulose , Listeria monocytogenes , Listeria monocytogenes/drug effects , Listeria monocytogenes/growth & development , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Cellulose/chemistry , Cellulose/pharmacology , Cellulose/biosynthesis , Coculture Techniques , Microbial Sensitivity Tests , Leuconostoc/metabolism , Bacteriocins/pharmacology , Bacteriocins/chemistry
18.
Int J Biol Macromol ; 274(Pt 1): 133290, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908631

ABSTRACT

MccY is a novel, structurally stable microcin with antibacterial activity against Enterobacteriaceae. However, the bioavailability of orally administrated MccY is unknown. This study evaluated the effects of MccY as a antimicrobial on pre-digestion in vitro and its intake, digestion and gut metabolism in vivo. The result of pre-digestion results that MccY maintained its biological activity and was resistant to decomposition. The study established a safe threshold of 4.46-9.92 mg/kg for the MccY dosage-body weight relationship in BALB/c mice. Mice fed with MccY demonstrated improved body weight and intestinal barrier function, accompanied with increased IgM immunogenicity and decreased levels of TNF-α, IL-6, and IL-10 in the intestine. MccY significantly facilitates the growth and activity of probiotics including Lactobacillus, Prevotella, and Bacteroides, and leading to the production of SCFAs and MCFAs during bacterial interactions. Furthermore, MccY effectively protects against the inflammatory response caused by Salmonella Typhimurium infection and effectively clears the Salmonella bacteria from the gut. In conclusion, MccY is seen as a promising new therapeutic target drug for enhancing the intestinal microbe-barrier axis and preventing enteritis.


Subject(s)
Bacteriocins , Gastrointestinal Microbiome , Mice, Inbred BALB C , Probiotics , Animals , Probiotics/pharmacology , Mice , Bacteriocins/pharmacology , Bacteriocins/chemistry , Gastrointestinal Microbiome/drug effects , Salmonella typhimurium/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Salmonella Infections/drug therapy , Intestines/microbiology , Intestines/drug effects
19.
PLoS Biol ; 22(6): e3002616, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38865418

ABSTRACT

The gastrointestinal tract is densely colonized by a polymicrobial community known as the microbiota which serves as primary line of defence against pathogen invasion. The microbiota can limit gut-luminal pathogen growth at different stages of infection. This can be traced to specific commensal strains exhibiting direct or indirect protective functions. Although these mechanisms hold the potential to develop new approaches to combat enteric pathogens, they remain far from being completely described. In this study, we investigated how a mouse commensal Escherichia coli can outcompete Salmonella enterica serovar Typhimurium (S. Tm). Using a salmonellosis mouse model, we found that the commensal E. coli 8178 strain relies on a trojan horse trap strategy to limit S. Tm expansion in the inflamed gut. Combining mutants and reporter tools, we demonstrated that inflammation triggers the expression of the E. coli 8178 antimicrobial microcin H47 toxin which, when fused to salmochelin siderophores, can specifically alter S. Tm growth. This protective function was compromised upon disruption of the E. coli 8178 tonB-dependent catecholate siderophore uptake system, highlighting a previously unappreciated crosstalk between iron intake and microcin H47 activity. By identifying the genetic determinants mediating S. Tm competition, our work not only provides a better mechanistic understanding of the protective function displayed by members of the gut microbiota but also further expands the general contribution of microcins in bacterial antagonistic relationships. Ultimately, such insights can open new avenues for developing microbiota-based approaches to better control intestinal infections.


Subject(s)
Escherichia coli , Inflammation , Salmonella typhimurium , Siderophores , Animals , Escherichia coli/metabolism , Escherichia coli/genetics , Siderophores/metabolism , Mice , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/metabolism , Inflammation/metabolism , Inflammation/microbiology , Mice, Inbred C57BL , Bacteriocins/metabolism , Bacteriocins/pharmacology , Gastrointestinal Microbiome , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Salmonella Infections/microbiology , Salmonella Infections/metabolism , Female , Iron/metabolism , Symbiosis , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/metabolism
20.
Appl Microbiol Biotechnol ; 108(1): 384, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896287

ABSTRACT

Bacteriocins have the potential to effectively improve food-borne infections or gastrointestinal diseases and hold promise as viable alternatives to antibiotics. This study aimed to explore the antibacterial activity of three bacteriocins (nisin, enterocin Gr17, and plantaricin RX-8) and their ability to attenuate intestinal barrier dysfunction and inflammatory responses induced by Listeria monocytogenes, respectively. Bacteriocins have shown excellent antibacterial activity against L. monocytogenes without causing any cytotoxicity. Bacteriocins inhibited the adhesion and invasion of L. monocytogenes on Caco-2 cells, lactate dehydrogenase (LDH), trans-epithelial electrical resistance (TEER), and cell migration showed that bacteriocin improved the permeability of Caco-2 cells. These results were attributed to the promotion of tight junction proteins (TJP) assembly, specifically zonula occludens-1 (ZO-1), occludin, and claudin-1. Furthermore, bacteriocins could alleviate inflammation by inhibiting the mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) pathways and reducing the secretion of interleukin-6 (IL-6), interleukin-1 ß (IL-1ß) and tumor necrosis factor α (TNF-α). Among three bacteriocins, plantaricin RX-8 showed the best antibacterial activity against L. monocytogenes and the most pronounced protective effect on the intestinal barrier due to its unique structure. Based on our findings, we hypothesized that bacteriocins may inhibit the adhesion and invasion of L. monocytogenes by competing adhesion sites. Moreover, they may further enhance intestinal barrier function by inhibiting the expression of L. monocytogenes virulence factors, increasing the expression of TJP and decreasing the secretion of inflammatory factors. Therefore, bacteriocins will hopefully be an effective alternative to antibiotics, and this study provides valuable insights into food safety concerns. KEY POINTS: • Bacteriocins show excellent antibacterial activity against L. monocytogenes • Bacteriocins improve intestinal barrier damage and inflammatory response • Plantaricin RX-8 has the best protective effect on Caco-2 cells damage.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Listeria monocytogenes , Listeria monocytogenes/drug effects , Bacteriocins/pharmacology , Humans , Caco-2 Cells , Anti-Bacterial Agents/pharmacology , Inflammation , NF-kappa B/metabolism , Bacterial Adhesion/drug effects , Tight Junction Proteins/metabolism , Cytokines/metabolism , Listeriosis/microbiology , Listeriosis/drug therapy , Cell Movement/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL