Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 380
Filter
Add more filters

Publication year range
1.
Nature ; 580(7805): 658-662, 2020 04.
Article in English | MEDLINE | ID: mdl-32350467

ABSTRACT

R-type bacteriocins are minimal contractile nanomachines that hold promise as precision antibiotics1-4. Each bactericidal complex uses a collar to bridge a hollow tube with a contractile sheath loaded in a metastable state by a baseplate scaffold1,2. Fine-tuning of such nucleic acid-free protein machines for precision medicine calls for an atomic description of the entire complex and contraction mechanism, which is not available from baseplate structures of the (DNA-containing) T4 bacteriophage5. Here we report the atomic model of the complete R2 pyocin in its pre-contraction and post-contraction states, each containing 384 subunits of 11 unique atomic models of 10 gene products. Comparison of these structures suggests the following sequence of events during pyocin contraction: tail fibres trigger lateral dissociation of baseplate triplexes; the dissociation then initiates a cascade of events leading to sheath contraction; and this contraction converts chemical energy into mechanical force to drive the iron-tipped tube across the bacterial cell surface, killing the bacterium.


Subject(s)
Pseudomonas aeruginosa , Pyocins/chemistry , Pyocins/metabolism , Bacteriophage T4/chemistry , Bacteriophage T4/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Genes, Bacterial/genetics , Models, Molecular , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Substrate Specificity , Type VI Secretion Systems/chemistry , Type VI Secretion Systems/metabolism
2.
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
3.
Biochem J ; 480(14): 1035-1049, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37399084

ABSTRACT

Pseudomonas aeruginosa is a common cause of serious hospital-acquired infections, the leading proven cause of mortality in people with cystic fibrosis and is associated with high levels of antimicrobial resistance. Pyocins are narrow-spectrum protein antibiotics produced by P. aeruginosa that kill strains of the same species and have the potential to be developed as therapeutics targeting multi-drug resistant isolates. We have identified two novel pyocins designated SX1 and SX2. Pyocin SX1 is a metal-dependent DNase while pyocin SX2 kills cells through inhibition of protein synthesis. Mapping the uptake pathways of SX1 and SX2 shows these pyocins utilize a combination of the common polysaccharide antigen (CPA) and a previously uncharacterized TonB-dependent transporter (TBDT) PA0434 to traverse the outer membrane. In addition, TonB1 and FtsH are required by both pyocins to energize their transport into cells and catalyze their translocation across the inner membrane, respectively. Expression of PA0434 was found to be specifically regulated by copper availability and we have designated PA0434 as Copper Responsive Transporter A, or CrtA. To our knowledge these are the first S-type pyocins described that utilize a TBDT that is not involved in iron uptake.


Subject(s)
Cystic Fibrosis , Pyocins , Humans , Pyocins/metabolism , Pyocins/pharmacology , Copper/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Pseudomonas aeruginosa/metabolism
4.
J Bacteriol ; 205(6): e0002923, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37260386

ABSTRACT

Most Pseudomonas aeruginosa strains produce bacteriocins derived from contractile or noncontractile phage tails known as R- and F-type pyocins, respectively. These bacteriocins possess strain-specific bactericidal activity against P. aeruginosa and likely increase evolutionary fitness through intraspecies competition. R-type pyocins have been studied extensively and show promise as alternatives to antibiotics. Although they have similar therapeutic potential, experimental studies on F-type pyocins are limited. Here, we provide a bioinformatic and experimental investigation of F-type pyocins. We introduce a systematic naming scheme for genes found in R- and F-type pyocin operons and identify 15 genes invariably found in strains producing F-type pyocins. Five proteins encoded at the 3' end of the F-type pyocin cluster are divergent in sequence and likely determine bactericidal specificity. We use sequence similarities among these proteins to define eleven distinct F-type pyocin groups, five of which had not been previously described. The five genes encoding the variable proteins associate in two modules that have clearly reassorted independently during the evolution of these operons. These proteins are considerably more diverse than the specificity-determining tail fibers of R-type pyocins, suggesting that F-type pyocins may have emerged earlier. Experimental studies on six F-type pyocin groups show that each displays a distinct spectrum of bactericidal activity. This activity is strongly influenced by the lipopolysaccharide O-antigen type, but other factors also play a role. F-type pyocins appear to kill as efficiently as R-type pyocins. These studies set the stage for the development of F-type pyocins as antibacterial therapeutics. IMPORTANCE Pseudomonas aeruginosa is an opportunistic pathogen that causes antibiotic-resistant infections with high mortality rates, particularly in immunocompromised individuals and cystic fibrosis patients. Due to the increasing frequency of multidrug-resistant P. aeruginosa infections, there is great need for the development of alternative therapeutics. In this study, we investigate one such potential therapeutic: F-type pyocins, which are bacteriocins naturally produced by P. aeruginosa that resemble noncontractile phage tails. We show that they are potent killers of P. aeruginosa and identify their probable bactericidal specificity determinants, which opens up the possibility of engineering them to precisely target strains of pathogenic bacteria. The resemblance of F-type pyocins to well-characterized phage tails will greatly facilitate their development into effective antibacterials.


Subject(s)
Bacteriocins , Bacteriophages , Humans , Pyocins/pharmacology , Pseudomonas aeruginosa/metabolism , Bacteriocins/genetics , Bacteriocins/pharmacology , Bacteriocins/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacteriophages/metabolism
5.
Environ Microbiol ; 24(3): 1294-1307, 2022 03.
Article in English | MEDLINE | ID: mdl-34735036

ABSTRACT

Interactions within bacterial communities are frequently mediated by the production of antimicrobial agents. Despite the increasing interest in research of new antimicrobials, studies describing antagonistic interactions among cold-adapted microorganisms are still rare. Our study assessed the antimicrobial interactions of 36 Antarctic Pseudomonas spp. and described the genetic background of these interactions in selected strains. The overall bacteriocinogeny was greater compared to mesophilic Pseudomonas non-aeruginosa species. R-type tailocins were detected on transmission electron micrographs in 16 strains (44.4%); phylogenetic analysis of the corresponding gene clusters revealed that the P. prosekii CCM 8878 tailocin was related to the Rp3 group, whereas the tailocin in Pseudomonas sp. CCM 8880 to the Rp4 group. Soluble antimicrobials were produced by eight strains (22.-2%); gene mining found pyocin L homologues in the genomes of P. prosekii CCM 8881 and CCM 8879 and pyocin S9-like homologues in P. prosekii CCM 8881 and Pseudomonas sp. CCM 8880. Analysis of secretomes confirmed the production of all S- and L-type pyocin genes. Our results suggest that bacteriocin-based inhibition plays an important role in interactions among Antarctic soil bacteria, and these native, cold-adapted microorganisms could be a promising source of new antimicrobials.


Subject(s)
Bacteriocins , Pyocins , Antarctic Regions , Bacteriocins/genetics , Phylogeny , Pseudomonas , Pseudomonas aeruginosa/genetics
6.
Infect Immun ; 89(2)2021 01 19.
Article in English | MEDLINE | ID: mdl-33168590

ABSTRACT

In Pseudomonas aeruginosa, the second messenger cyclic-di-GMP and Gac/Rsm signaling pathways are associated with the transition from acute to chronic infection. Therefore, identification of the molecular mechanisms that govern lifestyle choice in bacteria is very important. Here, we identified a novel cyclic-di-GMP modulator, PrtR, which was shown to repress pyocin production by inhibition of PrtN and activate the type III secretion system (T3SS) through PtrB. Compared to a wild-type strain or a prtN mutant, the prtR prtN double mutant exhibited a wrinkly colony and hyperbiofilm phenotype, as well as an increase in intracellular c-di-GMP levels. Interestingly, a diguanylate cyclase (DGC) gene, siaD, was repressed by PrtR. Further experiments revealed that PrtR directly interacts with SiaD and facilitates the accumulation of c-di-GMP in cells. We also demonstrated that PrtR regulates the activity of the Gac/Rsm system, thus affecting expression of the T3SS and type VI secretion system (T6SS) and the formation of biofilm. Taken together, the present findings indicate that PrtR, as a c-di-GMP modulator, plays key roles in the adaptation to opportunistic infection of P. aeruginosa Additionally, this study revealed a novel mechanism for PrtR-mediated regulation of the lifestyle transition via the Gac/Rsm and c-di-GMP signaling networks.


Subject(s)
Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/metabolism , Pyocins/metabolism , Signal Transduction/immunology , Virulence/genetics , Virulence/immunology , Gene Expression Regulation, Bacterial , Humans , Pseudomonas Infections/genetics , Pseudomonas Infections/immunology , Pseudomonas Infections/physiopathology , Pyocins/immunology , Signal Transduction/genetics
7.
J Antimicrob Chemother ; 76(9): 2317-2324, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34142136

ABSTRACT

BACKGROUND: Bloodstream infections with antibiotic-resistant Pseudomonas aeruginosa are common and increasingly difficult to treat. Pyocins are naturally occurring protein antibiotics produced by P. aeruginosa that have potential for human use. OBJECTIVES: To determine if pyocin treatment is effective in a murine model of sepsis with P. aeruginosa. METHODS: Recombinant pyocins S5 and AP41 were purified and tested for efficacy in a Galleria mellonella infection model and a murine model of P. aeruginosa sepsis. RESULTS: Both pyocins produced no adverse effects when injected alone into mice and showed good in vitro antipseudomonal activity. In an invertebrate model of sepsis using G. mellonella, both pyocins significantly prolonged survival from 1/10 (10%) survival in controls to 80%-100% survival among groups of 10 pyocin-treated larvae. Following injection into mice, both showed extensive distribution into different organs. When administered 5 h after infection, pyocin S5 significantly increased survival from 33% (2/6) to 83% (5/6) in a murine model of sepsis (difference significant by log-rank test, P < 0.05). CONCLUSIONS: Pyocins S5 and AP41 show in vivo biological activity and can improve survival in two models of P. aeruginosa infection. They hold promise as novel antimicrobial agents for treatment of MDR infections with this microbe.


Subject(s)
Pseudomonas Infections , Sepsis , Animals , Disease Models, Animal , Mice , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa , Pyocins , Sepsis/drug therapy
8.
J Appl Microbiol ; 131(6): 2780-2792, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33977611

ABSTRACT

AIM: This study is aimed at characterization of both antimicrobial and anti-biofilm activity of R-pyocin from clinical Pseudomonas aeruginosa against Gram-positive pathogens including Staphylococcus aureus. METHODS AND RESULTS: Pyocinogenic P. aeruginosa was detected using reverse-side method, and pyocinogeny typing was confirmed using revised-spotting method. Transmission electron microscopy (TEM) was used for morphological characterization of R-pyocin and for detection of changes in membrane of R-pyocin-treated S. aureus. SDS-PAGE analysis was used for detection of the molecular weight of R-pyocin protein-subunits and Poisson-killing-distribution assay for burst-size calculation. Lipotechoic-acid (LTA) adsorption-assay was used to confirm whether LTA in Gram-positive bacteria served as R-pyocin receptor. Moreover, R-pyocin production at 10-60°C was assessed herein. Host-range of activity of R-pyocin was tested against antimicrobial resistant (AMR) pathogens. The anti-biofilm activity of R-pyocin was detected against sensitive bacterial strains. Chemical, enzymatic, pH and thermo-stability of R-pyocin were evaluated. TEM micrographs revealed a typical morphology of myotailocins indicating the production of R-pyocin designated as RPU15. TEM revealed pores formation in S. aureus membrane, and bacteriophage-like plaques were obvious on plates of R-pyocin-treated S. aureus. R-pyocin activity was neutralized by LTA of S. aureus and Listeria monocytogenes. Pseudomonas aeruginosa PU15 produced ˜428 non-inducible R-pyocin particles. RPU15 sheath and tube protein-subunits exhibited a molecular weight of 38 and 23 kDa, respectively. RPU15 possessed activity against S. aureus, L. monocytogenes, Bacillus cereus and Candida albicans and reduced biofilm-biomasses of tested AMR strains. CONCLUSION: Our results show the potential therapeutic use of R-pyocin due to its effectiveness on tested bacterial biofilms. SIGNIFICANCE AND IMPACT OF THE STUDY: This is the first study that investigates antimicrobial and anti-biofilm activity of R-pyocin activity against S. aureus. R-pyocin shows new phenomenon of bacteriophage-like plaques. Our findings represent a future therapeutic agent targeting both methicillin-resistant and vancomycin-resistant S. aureus.


Subject(s)
Anti-Bacterial Agents , Pyocins/pharmacology , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Biofilms , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects
9.
Can J Microbiol ; 67(12): 919-932, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34437812

ABSTRACT

Pseudomonas aeruginosa, a gram-negative opportunistic pathogen, is one of the major species isolated from infected chronic wounds. The multidrug resistance exhibited by P. aeruginosa and its ability to form biofilms that are difficult to eradicate, along with the rising cost of producing new antibiotics, has necessitated the search for alternatives to standard antibiotics. Pyocins are antimicrobial compounds produced by P. aeruginosa that protect themselves from their competitors. We synthesized and purified recombinant P. aeruginosa R2 pyocin and used it in an aqueous solution (rR2P) or formulated in polyethylene glycol (rR2PC) to treat P. aeruginosa-infected wounds. Clinical strains of P. aeruginosa were found to be sensitive (completely), partially sensitive, or resistant to rR2P. In the in vitro biofilm model, rR2P inhibited biofilm development by rR2P-sensitive isolates, while rR2PC eliminated partial biofilms formed by these strains in an in vitro wound biofilm model. In the murine model of excision wounds, and at 24 h post-infection, rR2PC application significantly reduced the bioburden of the clinical isolate BPI86. Application of rR2PC containing two glycoside hydrolase antibiofilm agents eliminated BPI86 from infected wounds. These results suggest that the topical application of rR2PC is an effective therapy for treating wounds infected with R2P-senstive P. aeruginosa strains.


Subject(s)
Pseudomonas Infections , Wound Infection , Animals , Biofilms , Mice , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa , Pyocins , Wound Infection/drug therapy
10.
J Bacteriol ; 202(21)2020 10 08.
Article in English | MEDLINE | ID: mdl-32817098

ABSTRACT

Multidrug resistance (MDR) is a serious threat to public health, making the development of new antimicrobials an urgent necessity. Pyocins are protein antibiotics produced by Pseudomonas aeruginosa strains to kill closely related cells during intraspecific competition. Here, we report an in-depth biochemical, microbicidal, and structural characterization of a new S-type pyocin, named S8. Initially, we described the domain organization and secondary structure of S8. Subsequently, we observed that a recombinant S8 composed of the killing subunit in complex with the immunity (ImS8) protein killed the strain PAO1. Furthermore, mutation of a highly conserved glutamic acid to alanine (Glu100Ala) completely inhibited this antimicrobial activity. The integrity of the H-N-H motif is probably essential in the killing activity of S8, as Glu100 is a highly conserved residue of this motif. Next, we observed that S8 is a metal-dependent endonuclease, as EDTA treatment abolished its ability to cleave supercoiled pUC18 plasmid. Supplementation of apo S8 with Ni2+ strongly induced this DNase activity, whereas Mn2+ and Mg2+ exhibited moderate effects and Zn2+ was inhibitory. Additionally, S8 bound Zn2+ with a higher affinity than Ni2+ and the Glu100Ala mutation decreased the affinity of S8 for these metals, as shown by isothermal titration calorimetry (ITC). Finally, we describe the crystal structure of the Glu100Ala S8 DNase-ImS8 complex at 1.38 Å, which gave us new insights into the endonuclease activity of S8. Our results reinforce the possibility of using pyocin S8 as an alternative therapy for infections caused by MDR strains, while leaving commensal human microbiota intact.IMPORTANCE Pyocins are proteins produced by Pseudomonas aeruginosa strains that participate in intraspecific competition and host-pathogen interactions. They were first described in the 1950s and since then have gained attention as possible new antibiotics. However, there is still only scarce information about the molecular mechanisms by which these molecules induce cell death. Here, we show that the metal-dependent endonuclease activity of pyocin S8 is involved with its antimicrobial action against strain PAO1. We also describe that this killing activity is dependent on a conserved Glu residue within the H-N-H motif. The potency and selectivity of pyocin S8 toward a narrow spectrum of P. aeruginosa strains make this protein an attractive antimicrobial alternative for combatting MDR strains, while leaving commensal human microbiota intact.


Subject(s)
Anti-Bacterial Agents/chemistry , Deoxyribonuclease I/chemistry , Pseudomonas aeruginosa/metabolism , Pyocins/chemistry , Amino Acid Motifs , Glutamic Acid/chemistry , Structure-Activity Relationship
11.
J Bacteriol ; 202(11)2020 05 11.
Article in English | MEDLINE | ID: mdl-32205461

ABSTRACT

Factor for inversion stimulation (Fis) is a versatile DNA binding protein that plays an important role in coordinating bacterial global gene expression in response to growth phases and environmental stresses. Previously, we demonstrated that Fis regulates the type III secretion system (T3SS) in Pseudomonas aeruginosa In this study, we explored the role of Fis in the antibiotic resistance of P. aeruginosa and found that mutation of the fis gene increases the bacterial susceptibility to ciprofloxacin. We further demonstrated that genes related to pyocin biosynthesis are upregulated in the fis mutant. The pyocins are produced in response to genotoxic agents, including ciprofloxacin, and the release of pyocins results in lysis of the producer cell. Thus, pyocin biosynthesis genes sensitize P. aeruginosa to ciprofloxacin. We found that PrtN, the positive regulator of the pyocin biosynthesis genes, is upregulated in the fis mutant. Genetic experiments and electrophoretic mobility shift assays revealed that Fis directly binds to the promoter region of prtN and represses its expression. Therefore, our results revealed novel Fis-mediated regulation on pyocin production and bacterial resistance to ciprofloxacin in P. aeruginosaIMPORTANCEPseudomonas aeruginosa is an important opportunistic pathogenic bacterium that causes various acute and chronic infections in human, especially in patients with compromised immunity, cystic fibrosis (CF), and/or severe burn wounds. About 60% of cystic fibrosis patients have a chronic respiratory infection caused by P. aeruginosa The bacterium is intrinsically highly resistant to antibiotics, which greatly increases difficulties in clinical treatment. Therefore, it is critical to understand the mechanisms and the regulatory pathways that are involved in antibiotic resistance. In this study, we elucidated a novel regulatory pathway that controls the bacterial resistance to fluoroquinolone antibiotics, which enhances our understanding of how P. aeruginosa responds to ciprofloxacin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Ciprofloxacin/pharmacology , Drug Resistance, Bacterial , Factor For Inversion Stimulation Protein/metabolism , Pseudomonas aeruginosa/drug effects , Pyocins/biosynthesis , Bacterial Proteins/genetics , Factor For Inversion Stimulation Protein/genetics , Humans , Microbial Sensitivity Tests , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/genetics
12.
Proc Biol Sci ; 287(1938): 20201706, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33143580

ABSTRACT

Nearly all bacteria produce narrow-spectrum antibiotics called bacteriocins. Studies have shown that bacteriocins can mediate microbial interactions, but the mechanisms underlying patterns of inhibition are less well understood. We assembled a spatially structured collection of isolates of Pseudomonas aeruginosa from bathroom and kitchen sink drains in nine households. Growth inhibition of these P. aeruginosa by bacteriocins, known as pyocins in this species, was measured using pairwise inhibition assays. Carbon source usage of these isolates was measured, and genetic distance was estimated using multilocus sequencing. We found that as the distance between sites of isolation increased, there was a significantly higher probability of inhibition, and that pyocin inhibition and susceptibility vary greatly among isolates collected from different houses. We also detected support for other mechanisms influencing diversity: inhibition outcomes were influenced by the type of drain from which isolates were collected, and while we found no indication that carbon source utilization influences inhibition, inhibition was favoured at an intermediate genetic distance. Overall, these results suggest that the combined effects of dispersal limitation among sites and competitive exclusion within them maintain diversity in pyocin inhibition and susceptibility phenotypes, and that additional processes such as local adaptation and effects of phylogenetic distance could further contribute to spatial variability.


Subject(s)
Anti-Bacterial Agents/toxicity , Pseudomonas aeruginosa/physiology , Pyocins/toxicity , Bacteriocins , Microbial Interactions , Phenotype , Phylogeny
13.
J Bacteriol ; 201(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-30988031

ABSTRACT

Certain Pseudomonas aeruginosa strains produce a homolog of colicin M, namely, PaeM, that specifically inhibits peptidoglycan biosynthesis of susceptible P. aeruginosa strains by hydrolyzing the lipid II intermediate precursor. Two variants of this pyocin were identified whose sequences mainly differed in the N-terminal protein moiety, i.e., the region involved in the binding to the FiuA outer membrane receptor and translocation into the periplasm. The antibacterial activity of these two variants, PaeM1 and PaeM2, was tested against various P. aeruginosa strains comprising reference strains PAO1 and PA14, PaeM-producing strains, and 60 clinical isolates. Seven of these strains, including PAO1, were susceptible to only one variant (2 to PaeM1 and 5 to PaeM2), and 11 were affected by both. The remaining strains, including PA14 and four PaeM1 producers, were resistant to both variants. The differences in the antibacterial spectra of the two PaeM homologs prompted us to investigate the molecular determinants allowing their internalization into P. aeruginosa cells, taking the PAO1 strain that is susceptible to PaeM2 but resistant to PaeM1 as the indicator strain. Heterologous expression of fiuA gene orthologs from different strains into PAO1, site-directed mutagenesis experiments, and construction of PaeM chimeric proteins provided evidence that the cell susceptibility and discrimination differences between the PaeM variants resulted from a polymorphism of both the pyocin and the outer membrane receptor FiuA. Moreover, we found that a third component, TonB1, a protein involved in iron transport in P. aeruginosa, working together with FiuA and the ExbB/ExbD complex, was directly implicated in this discrimination.IMPORTANCE Bacterial antibiotic resistance constitutes a threat to human health, imposing the need for identification of new targets and development of new strategies to fight multiresistant pathogens. Bacteriocins and other weapons that bacteria have themselves developed to kill competitors are therefore of great interest and a valuable source of inspiration for us. Attention was paid here to two variants of a colicin M homolog (PaeM) produced by certain strains of P. aeruginosa that inhibit the growth of their congeners by blocking cell wall peptidoglycan synthesis. Molecular determinants allowing recognition of these pyocins by the outer membrane receptor FiuA were identified, and a receptor polymorphism affecting the susceptibility of P. aeruginosa clinical strains was highlighted, providing new insights into the potential use of these pyocins as an alternative to antibiotics.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Drug Resistance, Bacterial , Polymorphism, Genetic , Pseudomonas aeruginosa/genetics , Pyocins/pharmacology , Anti-Bacterial Agents/pharmacology , Cell Wall/chemistry , Mutagenesis, Site-Directed , Peptidoglycan/chemistry , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Receptors, Cell Surface
14.
Mol Microbiol ; 108(1): 6-15, 2018 04.
Article in English | MEDLINE | ID: mdl-29405518

ABSTRACT

Contractile tail bacteriophages, or myobacteriophages, use a sophisticated biomolecular structure to inject their genome into the bacterial host cell. This structure consists of a contractile sheath enveloping a rigid tube that is sharpened by a spike-shaped protein complex at its tip. The spike complex forms the centerpiece of a baseplate complex that terminates the sheath and the tube. The baseplate anchors the tail to the target cell membrane with the help of fibrous proteins emanating from it and triggers contraction of the sheath. The contracting sheath drives the tube with its spiky tip through the target cell membrane. Subsequently, the bacteriophage genome is injected through the tube. The structural transformation of the bacteriophage T4 baseplate upon binding to the host cell has been recently described in near-atomic detail. In this review we discuss structural elements and features of this mechanism that are likely to be conserved in all contractile injection systems (systems evolutionary and structurally related to contractile bacteriophage tails). These include the type VI secretion system (T6SS), which is used by bacteria to transfer effectors into other bacteria and into eukaryotic cells, and tailocins, a large family of contractile bacteriophage tail-like compounds that includes the P. aeruginosa R-type pyocins.


Subject(s)
Bacteriophage T4/chemistry , Bacteriophage T4/physiology , Viral Tail Proteins/chemistry , Viral Tail Proteins/physiology , Bacteriophage T4/genetics , Biological Evolution , Cell Membrane/chemistry , Cell Membrane/metabolism , Genome, Viral , Gram-Negative Bacteria/chemistry , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/physiology , Pyocins/chemistry , Pyocins/metabolism , Type VI Secretion Systems/chemistry , Type VI Secretion Systems/genetics , Type VI Secretion Systems/physiology , Viral Tail Proteins/genetics , X-Ray Diffraction
15.
Article in English | MEDLINE | ID: mdl-30962344

ABSTRACT

The prevalence of multidrug-resistant Pseudomonas aeruginosa has stimulated development of alternative therapeutics. Bacteriophage peptidoglycan hydrolases, termed lysins, represent an emerging antimicrobial option for targeting Gram-positive bacteria. However, lysins against Gram-negatives are generally deterred by the outer membrane and their inability to work in serum. One solution involves exploiting evolved delivery systems used by colicin-like bacteriocins (e.g., S-type pyocins of P. aeruginosa) to translocate through the outer membrane. Following surface receptor binding, colicin-like bacteriocins form Tol- or TonB-dependent translocons to actively import bactericidal domains through outer membrane protein channels. With this understanding, we developed lysocins, which are bioengineered lysin-bacteriocin fusion molecules capable of periplasmic import. In our proof-of-concept studies, components from the P. aeruginosa bacteriocin pyocin S2 (PyS2) responsible for surface receptor binding and outer membrane translocation were fused to the GN4 lysin to generate the PyS2-GN4 lysocin. PyS2-GN4 delivered the GN4 lysin to the periplasm to induce peptidoglycan cleavage and log-fold killing of P. aeruginosa with minimal endotoxin release. While displaying narrow-spectrum antipseudomonal activity in human serum, PyS2-GN4 also efficiently disrupted biofilms, outperformed standard-of-care antibiotics, exhibited no cytotoxicity toward eukaryotic cells, and protected mice from P. aeruginosa challenge in a bacteremia model. In addition to targeting P. aeruginosa, lysocins can be constructed to target other prominent Gram-negative bacterial pathogens.


Subject(s)
Anti-Infective Agents/pharmacology , Bacterial Outer Membrane/metabolism , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/metabolism , Peptidoglycan/pharmacology , Animals , Bacteriocins/metabolism , Bacteriophages/metabolism , Cell Line, Tumor , Colicins/metabolism , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/metabolism , HL-60 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Periplasm/metabolism , Pseudomonas aeruginosa/drug effects , Pyocins/metabolism
16.
J Antimicrob Chemother ; 73(10): 2770-2776, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30052973

ABSTRACT

Background: The appearance and dissemination of MDR among pathogenic bacteria has forced the search for new antimicrobials. Bacteriocins have been proposed as potential alternatives for the treatment of infections due to multiresistant strains. Objectives: To analyse the activity of R-pyocins against clinical isolates of Pseudomonas aeruginosa from patients with cystic fibrosis and other sources and evaluate them as a potential adjuvant or alternative to the current antibiotic treatment. Methods: The activity of R-pyocins against 150 strains of P. aeruginosa isolated from patients with cystic fibrosis or bacteraemia was studied through spot assay. Interactions between R-pyocins and antipseudomonal agents were quantitatively studied by the chequerboard method. Results: The proportion of P. aeruginosa isolates susceptible to R-pyocins was found to be higher in cystic fibrosis isolates compared with bacteraemia isolates (79.41% versus 50%). Moreover, no interactions were found between common antipseudomonal agents and R-pyocin susceptibility, except for the ST175 high-risk clone. Conclusions: Our results highlight the possibility of using R-pyocins as therapeutic agents, alone or as adjuvants, against P. aeruginosa in cystic fibrosis.


Subject(s)
Anti-Bacterial Agents/metabolism , Cystic Fibrosis/complications , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Pyocins/metabolism , Drug Interactions , Humans , Pseudomonas aeruginosa/isolation & purification
17.
Appl Environ Microbiol ; 84(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29980560

ABSTRACT

Pyocins are bacteriocins secreted by Pseudomonas aeruginosa, and they assist in the colonization of different niches. A major subset of these antibacterial proteins adopt a modular organization characteristic of polymorphic toxins. They include a receptor-binding domain, a segment enabling membrane passage, and a toxin module at the carboxy terminus, which eventually kills the target cells. To protect themselves from their own products, bacteriocin-producing strains express an immunity gene concomitantly with the bacteriocin. We show here that a pyocin equipped with a phylogenetically distinct ColM toxin domain, PaeM4, mediates antagonism against a large set of P. aeruginosa isolates. Immunity to PaeM4 is provided by the inner membrane protein PmiC, which is equipped with a transmembrane topology not previously described for the ColM family. Given that strains lacking a pmiC gene are killed by PaeM4, the presence of such an immunity partner likely is a key criterion for escaping cellular death mediated by PaeM4. The presence of a TonB box in PaeM4 and enhanced bacteriocin activity under iron-poor conditions strongly suggested the targeting of a TonB-dependent receptor. Evaluation of PaeM4 activities against TonB-dependent receptor knockout mutants in P. aeruginosa PAO1 revealed that the heme receptor HxuC (PA1302) serves as a PaeM4 target at the cellular surface. Because other ColM-type pyocins may target the ferrichrome receptor FiuA, our results illustrate the versatility in target recognition conferred by the polymorphic nature of ColM-type bacteriocins.IMPORTANCE The antimicrobial armamentarium of a bacterium is a major asset for colonizing competitive environments. Bacteriocins comprise a subset of these compounds. Pyocins are an example of such antibacterial proteins produced by Pseudomonas aeruginosa, killing other P. aeruginosa strains. A large group of these molecules show a modular protein architecture that includes a receptor-binding domain for initial target cell attachment and a killer domain. In this study, we have shown that a novel modular pyocin (PaeM4) that kills target bacteria via interference with peptidoglycan assembly takes advantage of the HxuC heme receptor. Cells can protect themselves from killing by the presence of a dedicated immunity partner, an integral inner membrane protein that adopts a transmembrane topology distinct from that of proteins currently known to provide immunity against such toxin activity. Understanding the receptors with which pyocins interact and how immunity to pyocins is achieved is a pivotal step toward the rational design of bacteriocin cocktails for the treatment of P. aeruginosa infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Drug Resistance, Bacterial , Pseudomonas aeruginosa/drug effects , Pyocins/pharmacology , Receptors, Cell Surface/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/genetics , Genome, Bacterial , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/metabolism , Pyocins/chemistry , Pyocins/metabolism , Receptors, Cell Surface/genetics
18.
PLoS Biol ; 13(7): e1002191, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26158271

ABSTRACT

Bacteria form dense surface-associated communities known as biofilms that are central to their persistence and how they affect us. Biofilm formation is commonly viewed as a cooperative enterprise, where strains and species work together for a common goal. Here we explore an alternative model: biofilm formation is a response to ecological competition. We co-cultured a diverse collection of natural isolates of the opportunistic pathogen Pseudomonas aeruginosa and studied the effect on biofilm formation. We show that strain mixing reliably increases biofilm formation compared to unmixed conditions. Importantly, strain mixing leads to strong competition: one strain dominates and largely excludes the other from the biofilm. Furthermore, we show that pyocins, narrow-spectrum antibiotics made by other P. aeruginosa strains, can stimulate biofilm formation by increasing the attachment of cells. Side-by-side comparisons using microfluidic assays suggest that the increase in biofilm occurs due to a general response to cellular damage: a comparable biofilm response occurs for pyocins that disrupt membranes as for commercial antibiotics that damage DNA, inhibit protein synthesis or transcription. Our data show that bacteria increase biofilm formation in response to ecological competition that is detected by antibiotic stress. This is inconsistent with the idea that sub-lethal concentrations of antibiotics are cooperative signals that coordinate microbial communities, as is often concluded. Instead, our work is consistent with competition sensing where low-levels of antibiotics are used to detect and respond to the competing genotypes that produce them.


Subject(s)
Antibiosis , Biofilms/growth & development , Pseudomonas aeruginosa/growth & development , Pyocins/pharmacology , Anti-Bacterial Agents , Biofilms/drug effects , Coculture Techniques , Microfluidics
19.
Appl Microbiol Biotechnol ; 102(24): 10393-10408, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30338356

ABSTRACT

Antimicrobial peptides (AMPs) from prokaryotic source also known as bacteriocins are ribosomally synthesized by bacteria belonging to different eubacterial taxonomic branches. Most of these AMPs are low molecular weight cationic membrane active peptides that disrupt membrane by forming pores in target cell membranes resulting in cell death. While these peptides known to exhibit broad-spectrum antimicrobial activity, including antibacterial and antifungal, they displayed minimal cytotoxicity to the host cells. Their antimicrobial efficacy has been demonstrated in vivo using diverse animal infection models. Therefore, we have discussed some of the promising peptides for their ability towards potential therapeutic applications. Further, some of these bacteriocins have also been reported to exhibit significant biological activity against various types of cancer cells in different experimental studies. In fact, differential cytotoxicity towards cancer cells as compared to normal cells by certain bacteriocins directs for a much focused research to utilize these compounds as novel therapeutic agents. In this review, bacteriocins that demonstrated antitumor activity against diverse cancer cell lines have been discussed emphasizing their biochemical features, selectivity against extra targets and molecular mechanisms of action.


Subject(s)
Antineoplastic Agents/pharmacology , Bacteriocins/genetics , Bacteriocins/pharmacology , Azurin/pharmacology , Bacteriocins/chemistry , Cations , Cell Membrane/drug effects , Humans , Nisin/pharmacology , Pediocins/pharmacology , Protein Engineering/methods , Pyocins/chemistry , Pyocins/pharmacology
20.
Article in English | MEDLINE | ID: mdl-28242657

ABSTRACT

A novel transposon belonging to the Tn3-like family was identified on the chromosome of a commensal strain of Pseudomonas aeruginosa sequence type 2343 (ET02). Tn6350 is 7,367 bp long and harbors eight open reading frames (ORFs), an ATPase (IS481 family), a transposase (DDE catalytic type), a Tn3 resolvase, three hypothetical proteins, and genes encoding the new pyocin S8 with its immunity protein. We show that pyocin S8 displays activity against carbapenemase-producing P. aeruginosa, including IMP-1, SPM-1, VIM-1, GES-5, and KPC-2 producers.


Subject(s)
DNA Transposable Elements/genetics , Pseudomonas aeruginosa/genetics , Pyocins/metabolism , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial/genetics , Humans , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/isolation & purification , beta-Lactamases/biosynthesis , beta-Lactamases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL