Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
1.
Viruses ; 16(7)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-39066163

ABSTRACT

The Gram-negative ESKAPE bacterium Pseudomonas aeruginosa has become a pathogen of serious concern due its extensive multi-drug resistance (MDR) profile, widespread incidences of hospital-acquired infections throughout the United States, and high occurrence in wound infections suffered by warfighters serving abroad. Bacteriophage (phage) therapy has received renewed attention as an alternative therapeutic option against recalcitrant bacterial infections, both as multi-phage cocktails and in combination with antibiotics as synergistic pairings. Environmental screening and phage enrichment has yielded three lytic viruses capable of infecting the MDR P. aeruginosa strain PAO1. Co-administration of each phage with the carbapenem antibiotics ertapenem, imipenem, and meropenem generated enhanced overall killing of bacteria beyond either phage or drug treatments alone. A combination cocktail of all three phages was completely inhibitory to growth, even without antibiotics. The same 3× phage cocktail also disrupted PAO1 biofilms, reducing biomass by over 75% compared to untreated biofilms. Further, the phage cocktail demonstrated broad efficacy as well, capable of infecting 33 out of 100 diverse clinical isolate strains of P. aeruginosa. Together, these results indicate a promising approach for designing layered medical countermeasures to potentiate antibiotic activity and possibly overcome resistance against recalcitrant, MDR bacteria such as P. aeruginosa. Combination therapy, either by synergistic phage-antibiotic pairings, or by phage cocktails, presents a means of controlling mutations that can allow for bacteria to gain a competitive edge.


Subject(s)
Anti-Bacterial Agents , Carbapenems , Drug Resistance, Multiple, Bacterial , Phage Therapy , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/drug effects , Carbapenems/pharmacology , Anti-Bacterial Agents/pharmacology , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Biofilms/drug effects , Bacteriophages/physiology , Microbial Sensitivity Tests , Humans , Pseudomonas Phages/physiology , Imipenem/pharmacology
2.
Viruses ; 16(7)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39066214

ABSTRACT

Antimicrobial resistance poses a serious risk to contemporary healthcare since it reduces the number of bacterial illnesses that may be treated with antibiotics, particularly for patients with long-term conditions like cystic fibrosis (CF). People with a genetic predisposition to CF often have recurrent bacterial infections in their lungs due to a buildup of sticky mucus, necessitating long-term antibiotic treatment. Pseudomonas aeruginosa infections are a major cause of CF lung illness, and P. aeruginosa airway isolates are frequently resistant to many antibiotics. Bacteriophages (also known as phages), viruses that infect bacteria, are a viable substitute for antimicrobials to treat P. aeruginosa infections in individuals with CF. Here, we reviewed the utilization of P. aeruginosa bacteriophages both in vivo and in vitro, as well as in the treatment of illnesses and diseases, and the outcomes of the latter.


Subject(s)
Cystic Fibrosis , Phage Therapy , Pseudomonas Infections , Pseudomonas Phages , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virology , Humans , Pseudomonas Infections/microbiology , Pseudomonas Infections/therapy , Cystic Fibrosis/microbiology , Pseudomonas Phages/genetics , Pseudomonas Phages/physiology , Animals , Bacteriophages/physiology , Bacteriophages/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
3.
BMC Microbiol ; 24(1): 207, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858621

ABSTRACT

BACKGROUND: Quorum sensing (QS) is a cell density-based intercellular communication system that controls virulence gene expression and biofilm formation. In Pseudomonas aeruginosa (P. aeruginosa), the LasR system sits at the top of the QS hierarchy and coordinates the expression of a series of important traits. However, the role of lasR in phage infection remains unclear. This study aims to investigate the role of lasR QS in phage infection. METHODS: The P. aeruginosa phage was isolated from sewage, and its biological characteristics and whole genome were analyzed. The adsorption receptor was identified via a phage adsorption assay. Following lasR gene knockout, the adsorption rate and bactericidal activity of phage were analyzed. Finally, real-time quantitative polymerase chain reaction (RT-qPCR) was conducted to explore how lasR promoting phage infection. RESULTS: The lytic phage vB_Pae_PLY was isolated and lipopolysaccharide (LPS) was identified as its adsorption receptor. The adsorption rate and bactericidal activity of vB_Pae_PLY were reduced after lasR knockout. RT-qPCR results showed that the expression of galU, a key gene involved in LPS synthesis, was down-regulated, and several genes related to type IV pili (T4P) were also down-regulated in the lasR mutant PaΔlasR. CONCLUSIONS: The study showed that QS lasR may promote phage vB_Pae_PLY infection by involving in the synthesis of LPS and T4P. This study provides an example of QS in promoting phage infection and deepens the understanding of phage-bacteria interactions.


Subject(s)
Bacterial Proteins , Pseudomonas aeruginosa , Quorum Sensing , Trans-Activators , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/genetics , Quorum Sensing/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Pseudomonas Phages/genetics , Pseudomonas Phages/physiology , Sewage/virology , Sewage/microbiology , Gene Expression Regulation, Bacterial , Lipopolysaccharides/metabolism , Gene Knockout Techniques
4.
mSphere ; 9(7): e0070723, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38934592

ABSTRACT

Phage therapy is increasing in relevance as an alternative treatment to combat antibiotic resistant bacteria. Phage cocktails are the state-of-the-art method of administering phages in clinical settings, preferred over monophage treatment because of their ability to eliminate multiple bacterial strains and reduce resistance formation. In our study, we compare monophage applications and phage cocktails to our chosen method of phage sequential treatments. To do so, we isolated four novel bacteriophages capable of infecting Pseudomonas alcaligenes T3, a close relative of P. aeruginosa, and characterized them using sequencing and transmission electron microscopy. While investigating monophage treatments, we observed that different phage concentrations had a strong impact on the timing and amount of resistance formation. When using phage cocktails, we observed that P. alcaligenes were capable of forming resistance in the same timespan it took them to become resistant to single phages. We isolated mutants resistant to each single phage as well as mutants exposed to phage cocktails, resulting in bacteria resistant to all four phages at once. Sequencing these mutants showed that different treatments yielded unique single nucleotide polymorphism mutation patterns. In order to combat resistance formation, we added phages one by one in intervals of 24 h, thus managing to delay resistance development and keeping bacterial growth significantly lower compared to phage cocktails.IMPORTANCEWHO declared antimicrobial resistance a top threat to global health; while antibiotics have stood at the forefront in the fight against bacterial infection, the increasing number of multidrug-resistant bacteria highlights a need to branch out in order to address the threat of antimicrobial resistance. Bacteriophages, viruses solely infecting bacteria, could present a solution due to their abundance, versatility, and adaptability. For this study, we isolated new phages infecting a fast-mutating Pseudomonas alcaligenes strain capable of forming resistance within 30 h. By using a sequential treatment approach of adding one phage after another, we were able to curb bacterial growth significantly more compared to state-of-the-art phage cocktails.


Subject(s)
Phage Therapy , Pseudomonas Phages , Pseudomonas , Phage Therapy/methods , Pseudomonas Phages/genetics , Pseudomonas Phages/physiology , Pseudomonas/virology , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/virology , Mutation , Anti-Bacterial Agents/pharmacology , Bacteriophages/genetics , Bacteriophages/physiology , Bacteriophages/classification , Drug Resistance, Bacterial , Drug Resistance, Multiple, Bacterial
5.
Microb Biotechnol ; 17(6): e14489, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38864499

ABSTRACT

Treating plant bacterial diseases is notoriously difficult because of the lack of available antimicrobials. Pseudomonas syringae pathovar syringae (Pss) is a major pathogen of cherry (Prunus avium) causing bacterial canker of the stem, leaf and fruit, impacting productivity and leading to a loss of trees. In an attempt to find a treatment for this disease, naturally occurring bacteriophage (phage) that specifically target Pss is being investigated as a biocontrol strategy. However, before using them as a biocontrol treatment, it is important to both understand their efficacy in reducing the bacterial population and determine if the bacterial pathogens can evolve resistance to evade phage infection. To investigate this, killing curve assays of five MR phages targeting Pss showed that phage resistance rapidly emerges in vitro, even when using a cocktail of the five phages together. To gain insight to the changes occurring, Pss colonies were collected three times during a 66-h killing curve assay and separately, Pss and phage were also coevolved over 10 generations, enabling the measurement of genomic and fitness changes in bacterial populations. Pss evolved resistance to phages through modifications in lipopolysaccharide (LPS) synthesis pathways. Bacterial fitness (growth) and virulence were affected in only a few mutants. Deletion of LPS-associated genes suggested that LPS was the main target receptor for all five MR phages. Later generations of coevolved phages from the coevolution experiment were more potent at reducing the bacterial density and when used with wild-type phages could reduce the emergence of phage-resistant mutants. This study shows that understanding the genetic mechanisms of bacterial pathogen resistance to phages is important for helping to design a more effective approach to kill the bacteria while minimizing the opportunity for phage resistance to manifest.


Subject(s)
Plant Diseases , Pseudomonas syringae , Pseudomonas syringae/virology , Pseudomonas syringae/genetics , Plant Diseases/microbiology , Pseudomonas Phages/genetics , Pseudomonas Phages/physiology , Bacteriophages/genetics , Bacteriophages/physiology
6.
Nat Microbiol ; 9(7): 1828-1841, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38886583

ABSTRACT

Bacteriophages have evolved diverse strategies to overcome host defence mechanisms and to redirect host metabolism to ensure successful propagation. Here we identify a phage protein named Dap1 from Pseudomonas aeruginosa phage PaoP5 that both modulates bacterial host behaviour and contributes to phage fitness. We show that expression of Dap1 in P. aeruginosa reduces bacterial motility and promotes biofilm formation through interference with DipA, a c-di-GMP phosphodiesterase, which causes an increase in c-di-GMP levels that trigger phenotypic changes. Results also show that deletion of dap1 in PaoP5 significantly reduces genome packaging. In this case, Dap1 directly binds to phage HNH endonuclease, prohibiting host Lon-mediated HNH degradation and promoting phage genome packaging. Moreover, PaoP5Δdap1 fails to rescue P. aeruginosa-infected mice, implying the significance of dap1 in phage therapy. Overall, these results highlight remarkable dual functionality in a phage protein, enabling the modulation of host behaviours and ensuring phage fitness.


Subject(s)
Phage Therapy , Pseudomonas Infections , Pseudomonas Phages , Pseudomonas aeruginosa , Viral Proteins , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/genetics , Animals , Mice , Pseudomonas Phages/genetics , Pseudomonas Phages/physiology , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Pseudomonas Infections/immunology , Virulence , Viral Proteins/genetics , Viral Proteins/metabolism , Biofilms/growth & development , Cyclic GMP/metabolism , Cyclic GMP/analogs & derivatives , Female , Bacteriophages/physiology , Bacteriophages/genetics
7.
Microbiol Spectr ; 12(8): e0352023, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38912817

ABSTRACT

Pseudomonas aeruginosa infections are getting increasingly serious as antimicrobial resistance spreads. Phage therapy may be a solution to the problem, especially if improved by current advances on phage-host studies. As a mucosal pathogen, we hypothesize that P. aeruginosa and its phages are linked to the bacteriophage adherence to mucus (BAM) model. This means that phage-host interactions could be influenced by mucin presence, impacting the success of phage infections on the P. aeruginosa host and consequently leading to the protection of the metazoan host. By using a group of four different phages, we tested three important phenotypes associated with the BAM model: phage binding to mucin, phage growth in mucin-exposed hosts, and the influence of mucin on CRISPR immunity of the bacterium. Three of the tested phages significantly bound to mucin, while two had improved growth rates in mucin-exposed hosts. Improved phage growth was likely the result of phage exploitation of mucin-induced physiological changes in the host. We could not detect CRISPR activity in our system but identified two putative anti-CRISPR proteins coded by the phage. Overall, the differential responses seen for the phages tested show that the same bacterial species can be targeted by mucosal-associated phages or by phages not affected by mucus presence. In conclusion, the BAM model is relevant for phage-bacterium interactions in P. aeruginosa, opening new possibilities to improve phage therapy against this important pathogen by considering mucosal interaction dynamics.IMPORTANCESome bacteriophages are involved in a symbiotic relationship with animals, in which phages held in mucosal surfaces protect them from invading bacteria. Pseudomonas aeruginosa is one of the many bacterial pathogens threatening humankind during the current antimicrobial resistance crisis. Here, we have tested whether P. aeruginosa and its phages are affected by mucosal conditions. We discovered by using a collection of four phages that, indeed, mucosal interaction dynamics can be seen in this model. Three of the tested phages significantly bound to mucin, while two had improved growth rates in mucin-exposed hosts. These results link P. aeruginosa and its phages to the bacteriophage adherence to the mucus model and open opportunities to explore this to improve phage therapy, be it by exploiting the phenotypes detected or by actively selecting mucosal-adapted phages for treatment.


Subject(s)
Mucins , Mucus , Pseudomonas Infections , Pseudomonas Phages , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/physiology , Mucus/microbiology , Mucus/virology , Pseudomonas Phages/physiology , Pseudomonas Phages/genetics , Mucins/metabolism , Pseudomonas Infections/microbiology , Pseudomonas Infections/therapy , Humans , Phage Therapy , Animals , Virus Attachment , Clustered Regularly Interspaced Short Palindromic Repeats
8.
Arch Microbiol ; 206(6): 283, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38806864

ABSTRACT

The objective of this study was to investigate the effectiveness of a phage cocktail against Pseudomonas fluorescens group and its effect on the microbial, physical and chemical properties of raw milk during different storage conditions. A phage cocktail consisting of Pseudomonas fluorescens, Pseudomonas tolaasii, and Pseudomonas libanensis phages was prepared. As a result, reductions in fluorescent Pseudomonas counts of up to 3.44 log units for the storage at 4 °C and 2.38 log units for the storage at 25 °C were achieved. Following the phage application, it is found that there was no significant difference in the total mesophilic aerobic bacteria and Enterobacteriaceae counts. However, it was observed that the number of lactic acid bacteria was higher in phage-treated groups. The results also showed that pH values in the phage added groups were lower than the others and the highest titratable acidity was obtained only in the bacteria-inoculated group. As a future perspective, this study suggests that, while keeping the number of target microorganisms under control in the milk with the use of phages during storage, the microbiota and accordingly the quality parameters of the milk can be affected. This work contributes to the development of effective strategies for maintaining the quality and extending the shelf life of milk and dairy products.


Subject(s)
Milk , Pseudomonas Phages , Pseudomonas fluorescens , Milk/microbiology , Pseudomonas fluorescens/virology , Animals , Pseudomonas Phages/physiology , Pseudomonas Phages/isolation & purification , Food Microbiology , Hydrogen-Ion Concentration , Bacteriophages/physiology , Bacteriophages/isolation & purification
9.
J Bacteriol ; 206(5): e0040223, 2024 05 23.
Article in English | MEDLINE | ID: mdl-38687034

ABSTRACT

Pseudomonas aeruginosa is an opportunistic bacterial pathogen that commonly causes medical hardware, wound, and respiratory infections. Temperate filamentous Pf phages that infect P. aeruginosa impact numerous virulence phenotypes. Most work on Pf phages has focused on Pf4 and its host P. aeruginosa PAO1. Expanding from Pf4 and PAO1, this study explores diverse Pf phages infecting P. aeruginosa clinical isolates. We describe a simple technique targeting the Pf lysogeny maintenance gene, pflM (PA0718), that enables the effective elimination of Pf prophages from diverse P. aeruginosa hosts. The pflM gene shows diversity among different Pf phage isolates; however, all examined pflM alleles encode the DUF5447 domain. We demonstrate that pflM deletion results in prophage excision but not replication, leading to total prophage loss, indicating a role for lysis/lysogeny decisions for the DUF5447 domain. This study also assesses the effects different Pf phages have on host quorum sensing, biofilm formation, pigment production, and virulence against the bacterivorous nematode Caenorhabditis elegans. We find that Pf phages have strain-specific impacts on quorum sensing and biofilm formation, but nearly all suppress pigment production and increase C. elegans avoidance behavior. Collectively, this research not only introduces a valuable tool for Pf prophage elimination from diverse P. aeruginosa isolates but also advances our understanding of the complex relationship between P. aeruginosa and filamentous Pf phages.IMPORTANCEPseudomonas aeruginosa is an opportunistic bacterial pathogen that is frequently infected by filamentous Pf phages (viruses) that integrate into its chromosome, affecting behavior. Although prior work has focused on Pf4 and PAO1, this study investigates diverse Pf in clinical isolates. A simple method targeting the deletion of the Pf lysogeny maintenance gene pflM (PA0718) effectively eliminates Pf prophages from clinical isolates. The research evaluates the impact Pf prophages have on bacterial quorum sensing, biofilm formation, and virulence phenotypes. This work introduces a valuable tool to eliminate Pf prophages from clinical isolates and advances our understanding of P. aeruginosa and filamentous Pf phage interactions.


Subject(s)
Prophages , Pseudomonas aeruginosa , Quorum Sensing , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/physiology , Prophages/genetics , Prophages/physiology , Virulence , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/virology , Biofilms/growth & development , Animals , Lysogeny , Pseudomonas Phages/genetics , Pseudomonas Phages/physiology , Pseudomonas Infections/microbiology
10.
Sci Rep ; 14(1): 9354, 2024 04 23.
Article in English | MEDLINE | ID: mdl-38653744

ABSTRACT

Phage-antibiotic combinations to treat bacterial infections are gaining increased attention due to the synergistic effects often observed when applying both components together. Most studies however focus on a single pathogen, although in many clinical cases multiple species are present at the site of infection. The aim of this study was to investigate the anti-biofilm activity of phage-antibiotic/antifungal combinations on single- and dual-species biofilms formed by P. aeruginosa and the fungal pathogen Candida albicans. The Pseudomonas phage Motto in combination with ciprofloxacin had significant anti-biofilm activity. We then compared biofilms formed by P. aeruginosa alone with the dual-species biofilms formed by bacteria and C. albicans. Here, we found that the phage together with the antifungal fluconazole was active against 6-h-old dual-species biofilms but showed only negligible activity against 24-h-old biofilms. This study lays the first foundation for potential therapeutic approaches to treat co-infections caused by bacteria and fungi using phage-antibiotic combinations.


Subject(s)
Anti-Bacterial Agents , Antifungal Agents , Biofilms , Candida albicans , Ciprofloxacin , Pseudomonas Phages , Pseudomonas aeruginosa , Biofilms/drug effects , Biofilms/growth & development , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Pseudomonas aeruginosa/virology , Antifungal Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Pseudomonas Phages/physiology , Candida albicans/drug effects , Candida albicans/physiology , Ciprofloxacin/pharmacology , Fluconazole/pharmacology , Microbial Sensitivity Tests
11.
Science ; 384(6691): eadl0635, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38574145

ABSTRACT

The retractile type IV pilus (T4P) is important for virulence of the opportunistic human pathogen Pseudomonas aeruginosa. The single-stranded RNA (ssRNA) phage PP7 binds to T4P and is brought to the cell surface through pilus retraction. Using fluorescence microscopy, we discovered that PP7 detaches T4P, which impairs cell motility and restricts the pathogen's virulence. Using cryo-electron microscopy, mutagenesis, optical trapping, and Langevin dynamics simulation, we resolved the structure of PP7, T4P, and the PP7/T4P complex and showed that T4P detachment is driven by the affinity between the phage maturation protein and its bound pilin, plus the pilus retraction force and speed, and pilus bending. Pilus detachment may be widespread among other ssRNA phages and their retractile pilus systems and offers new prospects for antibacterial prophylaxis and therapeutics.


Subject(s)
Fimbriae, Bacterial , Pseudomonas Phages , Pseudomonas aeruginosa , RNA Viruses , Virus Internalization , Humans , Cryoelectron Microscopy , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/virology , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/virology , RNA Viruses/chemistry , RNA Viruses/physiology , Pseudomonas Phages/chemistry , Pseudomonas Phages/physiology , Viral Proteins/metabolism
12.
Appl Environ Microbiol ; 90(5): e0028624, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38624196

ABSTRACT

Host-parasite interactions are highly susceptible to changes in temperature due to mismatches in species thermal responses. In nature, parasites often exist in communities, and responses to temperature are expected to vary between host-parasite pairs. Temperature change thus has consequences for both host-parasite dynamics and parasite-parasite interactions. Here, we investigate the impact of warming (37°C, 40°C, and 42°C) on parasite life-history traits and competition using the opportunistic bacterial pathogen Pseudomonas aeruginosa (host) and a panel of three genetically diverse lytic bacteriophages (parasites). We show that phages vary in their responses to temperature. While 37°C and 40°C did not have a major effect on phage infectivity, infection by two phages was restricted at 42°C. This outcome was attributed to disruption of different phage life-history traits including host attachment and replication inside hosts. Furthermore, we show that temperature mediates competition between phages by altering their competitiveness. These results highlight phage trait variation across thermal regimes with the potential to drive community dynamics. Our results have important implications for eukaryotic viromes and the design of phage cocktail therapies.IMPORTANCEMammalian hosts often elevate their body temperatures through fevers to restrict the growth of bacterial infections. However, the extent to which fever temperatures affect the communities of phages with the ability to parasitize those bacteria remains unclear. In this study, we investigate the impact of warming across a fever temperature range (37°C, 40°C, and 42°C) on phage life-history traits and competition using a bacterium (host) and bacteriophage (parasite) system. We show that phages vary in their responses to temperature due to disruption of different phage life-history traits. Furthermore, we show that temperature can alter phage competitiveness and shape phage-phage competition outcomes. These results suggest that fever temperatures have the potential to restrict phage infectivity and drive phage community dynamics. We discuss implications for the role of temperature in shaping host-parasite interactions more widely.


Subject(s)
Pseudomonas aeruginosa , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/physiology , Bacteriophages/physiology , Hot Temperature , Pseudomonas Phages/physiology , Pseudomonas Phages/growth & development , Life History Traits , Temperature
13.
Viruses ; 16(4)2024 04 21.
Article in English | MEDLINE | ID: mdl-38675985

ABSTRACT

The phage PRR1 belongs to the Leviviridae family, a group of ssRNA bacteriophages that infect Gram-negative bacteria. The variety of host cells is determined by the specificity of PRR1 to a pilus encoded by a broad host range of IncP-type plasmids that confer multiple types of antibiotic resistance to the host. Using P. aeruginosa strain PAO1 as a host, we analyzed the PRR1 infection cycle, focusing on cell lysis. PRR1 infection renders P. aeruginosa cells sensitive to lysozyme approximately 20 min before the start of a drop in suspension turbidity. At the same time, infected cells start to accumulate lipophilic anions. The on-line monitoring of the entire infection cycle showed that single-gene-mediated lysis strongly depends on the host cells' physiological state. The blockage of respiration or a reduction in the intracellular ATP concentration during the infection resulted in the inhibition of lysis. The same effect was observed when the synthesis of PRR1 lysis protein was induced in an E. coli expression system. In addition, lysis was strongly dependent on the level of aeration. Dissolved oxygen concentrations sufficient to support cell growth did not ensure efficient lysis, and a coupling between cell lysis initiation and aeration level was observed. However, the duration of the drop in suspension turbidity did not depend on the level of aeration.


Subject(s)
Bacteriolysis , Pseudomonas Phages , Pseudomonas aeruginosa , Escherichia coli/genetics , Host Specificity , Muramidase/metabolism , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/physiology , Pseudomonas aeruginosa/genetics , Pseudomonas Phages/physiology , Pseudomonas Phages/genetics
14.
Int Microbiol ; 27(4): 1333-1344, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38206524

ABSTRACT

Pseudomonas spp., such as P. fluorescens group, P. fragi, and P. putida, are the major psychrophilic spoilage bacteria in the food industry. Bacteriophages (phages) are a promising tool for controlling food-spoilage and food-poisoning bacteria; however, there are few reports on phages effective on food-spoilage bacteria such as Pseudomonas spp. In this study, 12 Pseudomonas phages were isolated from chicken and soil samples. Based on the host range and lytic activity at 30 °C and 4 °C and various combinations of phages, phages vB_PflP-PCS4 and vB_PflP-PCW2 were selected to prepare phage cocktails to control Pseudomonas spp. The phage cocktail consisting of vB_PflP-PCS4 and vB_PflP-PCW2 showed the strongest lytic activity and retarded regrowth of P. fluorescens and P. putida at 30 °C, 8 °C, and 4 °C at a multiplicity of infection of 100. Nucleotide sequence analysis of the genomic DNA indicated that vB_PflP-PCS4 and vB_PflP-PCW2 phages were lytic phages of the Podoviridae family and lacked tRNA, toxin, or virulence genes. A novel endolysin gene was found in the genomic DNA of phage vB_PflP-PCS4. The results of this study suggest that the phage cocktail consisting of vB_PflP-PCS4 and vB_PflP-PCW2 is a promising tool for the biocontrol of psychrophilic food-spoilage pseudomonads during cold storage and distribution.


Subject(s)
Chickens , Food Microbiology , Host Specificity , Animals , Soil Microbiology , Pseudomonas Phages/physiology , Pseudomonas Phages/genetics , Pseudomonas/virology , Genome, Viral , Podoviridae/physiology , Podoviridae/genetics , Podoviridae/isolation & purification , Podoviridae/classification , Biological Control Agents , DNA, Viral/genetics , Bacteriophages/physiology , Bacteriophages/genetics , Bacteriophages/isolation & purification , Bacteriophages/classification
15.
Viruses ; 15(11)2023 Nov 03.
Article in English | MEDLINE | ID: mdl-38005888

ABSTRACT

Therapeutic bacteriophages (phages) are primarily chosen based on their in vitro bacteriolytic activity. Although anti-phage antibodies are known to inhibit phage infection, the influence of other immune system components is less well known. An important anti-bacterial and anti-viral innate immune system that may interact with phages is the complement system, a cascade of proteases that recognizes and targets invading microorganisms. In this research, we aimed to study the effects of serum components such as complement on the infectivity of different phages targeting Pseudomonas aeruginosa. We used a fluorescence-based assay to monitor the killing of P. aeruginosa by phages of different morphotypes in the presence of human serum. Our results reveal that several myophages are inhibited by serum in a concentration-dependent way, while the activity of four podophages and one siphophage tested in this study is not affected by serum. By using specific nanobodies blocking different components of the complement cascade, we showed that activation of the classical complement pathway is a driver of phage inhibition. To determine the mechanism of inhibition, we produced bioorthogonally labeled fluorescent phages to study their binding by means of microscopy and flow cytometry. We show that phage adsorption is hampered in the presence of active complement. Our results indicate that interactions with complement may affect the in vivo activity of therapeutically administered phages. A better understanding of this phenomenon is essential to optimize the design and application of therapeutic phage cocktails.


Subject(s)
Bacteriophages , Pseudomonas Infections , Pseudomonas Phages , Humans , Pseudomonas aeruginosa/physiology , Pseudomonas Phages/physiology , Bacteriolysis , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology
16.
Appl Microbiol Biotechnol ; 107(2-3): 897-913, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36625915

ABSTRACT

The implementation of non-traditional antibacterials is currently one of the most intensively explored areas of modern medical and biological sciences. One of the most promising alternative strategies to combat bacterial infections is the application of lytic phages combined with established and new antibacterials. The presented study investigates the potential of agarose-based biocomposites containing lytic Pseudomonas phages (KT28, KTN4, and LUZ19), cupric ions (Cu2+), strawberry furanone (HDMF), and gentamicin (GE) as antibacterials and anti-virulent compounds for novel wound dressings. Phages (KT28, KTN4, LUZ19, and triple-phage cocktail) alone and in combination with a triple-chemical mixture (Cu + GE + HDMF) when applied as the liquid formulation caused a significant bacterial count reduction and biofilm production inhibition of clinical P. aeruginosa strains. The immobilization in the agarose scaffold significantly impaired the bioavailability and diffusion of phage particles, depending on virion morphology and targeted receptor specificity. The antibacterial potential of chemicals was also reduced by the agarose scaffold. Moreover, the Cu + GE + HDMF mixture impaired the lytic activity of phages depending on viral particles' susceptibility to cupric ion toxicity. Therefore, three administration types were tested and the optimal turned out to be the one separating antibacterials both physically and temporally. Taken together, the additive effect of phages combined with chemicals makes biocomposite a good solution for designing new wound dressings. Nevertheless, the phage utilization should involve an application of aqueous cocktails directly onto the wound, followed by chemicals immobilized in hydrogel dressings which allow for taking advantage of the antibacterial and anti-virulent effects of all components. KEY POINTS: • The immobilization in the agarose impairs the bioavailability of phage particles and the Cu + GE + HDMF mixture. • The cupric ions are toxic to phages and are sequestrated on phage particles and agarose matrix. • The elaborated TIME-SHIFT administration effectively separates antibacterials both physically and temporally.


Subject(s)
Bacteriophages , Pseudomonas Infections , Pseudomonas Phages , Humans , Bacteriophages/physiology , Pseudomonas aeruginosa , Sepharose , Pseudomonas Phages/physiology , Anti-Bacterial Agents/pharmacology , Pseudomonas Infections/microbiology
17.
J Virol ; 96(5): e0176921, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35020473

ABSTRACT

There is a continuously expanding gap between predicted phage gene sequences and their corresponding functions, which has largely hampered the development of phage therapy. Previous studies reported several phage proteins that could interfere with the intracellular processes of the host to obtain efficient infection. But few phage proteins that protect host against phage infection have been identified and characterized in detail. Here, we isolate a phage, vB_Pae_QDWS, capable of infecting Pseudomonas aeruginosa PAO1 and report that its encoded Gp21 protein protects PAO1 against phage infection. Expression of Gp21 regulates bacterial quorum sensing with an inhibitory effect in low cell density and an activation effect in high cell density. By testing the type IV pilus (TFP)-mediated twitching motility and transmission electron microscopy analysis, Gp21 was found to decrease the pilus synthesis. Further, by constructing the TFP synthesis gene pilB mutant and performing adsorption and phage resistance assay, we demonstrated that the Gp21 protein could block phage infection via decreasing the TFP-mediated phage adsorption. Gp21 is a novel protein that inhibits phage efficacy against bacteria. The study deepens our understanding of phage-host interactions. IMPORTANCE The majority of the annotated phage genes are currently deposited as "hypothetical protein" with unknown function. Research has revealed that some phage proteins serve to inhibit or redirect the host intracellular processes for phage infection. Conversely, we report a phage encoded protein Gp21 that protects the host against phage infection. The pathways that Gp21 involved in antiphage defense in Pseudomonas aeruginosa PAO1 interfere with quorum sensing and decrease type IV pilus-mediated phage adsorption. Gp21 is a novel protein with a low sequence homology with other reported twitching inhibitory proteins. As a lytic phage-derived protein, Gp21 expression protects P. aeruginosa PAO1 from reinfection by phage vB_Pae_QDWS, which may explain the well-known pseudolysogeny caused by virulent phages. Our discoveries provide valuable new insight into phage-host evolutionary dynamics.


Subject(s)
Pseudomonas Phages , Pseudomonas aeruginosa , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Gene Expression , Pseudomonas Phages/physiology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/virology , Quorum Sensing
18.
Viruses ; 13(11)2021 11 14.
Article in English | MEDLINE | ID: mdl-34835081

ABSTRACT

Bacteriophages are viruses that specifically infect target bacteria. Recently, bacteriophages have been considered potential biological control agents for bacterial pathogens due to their host specificity. Pseudomonas syringae pv. actinidiae (Psa) is a reemerging pathogen that causes bacterial canker of kiwifruit (Actinidia sp.). The economic impact of this pest and the development of resistance to antibiotics and copper sprays in Psa and other pathovars have led to investigation of alternative management strategies. Phage therapy may be a useful alternative to conventional treatments for controlling Psa infections. Although the efficacy of bacteriophage φ6 was evaluated for the control of Psa, the characteristics of other DNA bacteriophages infecting Psa remain unclear. In this study, the PHB09 lytic bacteriophage specific to Psa was isolated from kiwifruit orchard soil. Extensive host range testing using Psa isolated from kiwifruit orchards and other Pseudomonas strains showed PHB09 has a narrow host range. It remained stable over a wide range of temperatures (4-50 °C) and pH values (pH 3-11) and maintained stability for 50 min under ultraviolet irradiation. Complete genome sequence analysis indicated PHB09 might belong to a new myovirus genus in Caudoviricetes. Its genome contains a total of 94,844 bp and 186 predicted genes associated with phage structure, packaging, host lysis, DNA manipulation, transcription, and additional functions. The isolation and identification of PHB09 enrich the research on Pseudomonas phages and provide a promising biocontrol agent against kiwifruit bacterial canker.


Subject(s)
Host Specificity , Podoviridae/isolation & purification , Pseudomonas Phages/isolation & purification , Pseudomonas syringae/virology , Virion/ultrastructure , Actinidia/microbiology , DNA, Viral/chemistry , DNA, Viral/genetics , Plant Diseases/microbiology , Podoviridae/genetics , Podoviridae/physiology , Podoviridae/ultrastructure , Pseudomonas Phages/genetics , Pseudomonas Phages/physiology , Pseudomonas Phages/ultrastructure
19.
Nucleic Acids Res ; 49(18): 10770-10784, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34520554

ABSTRACT

H-NS family proteins, bacterial xenogeneic silencers, play central roles in genome organization and in the regulation of foreign genes. It is thought that gene repression is directly dependent on the DNA binding modes of H-NS family proteins. These proteins form lateral protofilaments along DNA. Under specific environmental conditions they switch to bridging two DNA duplexes. This switching is a direct effect of environmental conditions on electrostatic interactions between the oppositely charged DNA binding and N-terminal domains of H-NS proteins. The Pseudomonas lytic phage LUZ24 encodes the protein gp4, which modulates the DNA binding and function of the H-NS family protein MvaT of Pseudomonas aeruginosa. However, the mechanism by which gp4 affects MvaT activity remains elusive. In this study, we show that gp4 specifically interferes with the formation and stability of the bridged MvaT-DNA complex. Structural investigations suggest that gp4 acts as an 'electrostatic zipper' between the oppositely charged domains of MvaT protomers, and stabilizes a structure resembling their 'half-open' conformation, resulting in relief of gene silencing and adverse effects on P. aeruginosa growth. The ability to control H-NS conformation and thereby its impact on global gene regulation and growth might open new avenues to fight Pseudomonas multidrug resistance.


Subject(s)
Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , Pseudomonas Phages/physiology , Trans-Activators/metabolism , Viral Proteins/metabolism , Bacterial Proteins/chemistry , DNA/metabolism , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Bacterial , Gene Silencing , Models, Molecular , Protein Binding , Pseudomonas/genetics , Pseudomonas/growth & development , Pseudomonas/virology , Trans-Activators/chemistry , Viral Proteins/chemistry
20.
Sci Rep ; 11(1): 19393, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34588479

ABSTRACT

In recent years, the use of bacteriophages (or 'phages') against multidrug-resistant (MDR) bacteria including Pseudomonas aeruginosa has drawn considerable attention, globally. In this work, we report the isolation and detailed characterization of a highly lytic Pseudomonasphage DRL-P1 isolated from wastewater. Under TEM, DRL-P1 appeared as a member of the phage family Myoviridae. DRL-P1 featured rapid adsorption (~ 5 min), short-latency (~ 30 min), and large burst size (~ 100 PFU per infected cell). DRL-P1 can withstand a wide temperature range (4 °C to 40 °C) and pH (5.0 to 10.0) conditions. The 66,243 bp DRL-P1 genome (MN564818) encodes at least 93 ORFs, of which 36 were functionally annotated based on homology with similar phage proteins available in the databases. Comparative analyses of related genomes suggest an independent evolutionary history and discrete taxonomic position of DRL-P1 within genus Pbunavirus. No toxin or antibiotic resistance genes was identified. DRL-P1 is tolerant to lyophilization and encapsulation techniques and retained lytic activity even after 18 months of storage. We also demonstrated decontaminating potentials of DRL-P1 in vitro, on an artificially contaminated cover-slip model. To the best of our knowledge, this is the first Pbunavirus to be reported from India. Our study suggests DRL-P1 as a potential candidate for various applications.


Subject(s)
Myoviridae , Pseudomonas Phages , Pseudomonas aeruginosa/virology , Wastewater , DNA, Viral , Drug Resistance, Multiple, Bacterial/genetics , Genome, Viral , Myoviridae/classification , Myoviridae/isolation & purification , Myoviridae/physiology , Pseudomonas Phages/classification , Pseudomonas Phages/isolation & purification , Pseudomonas Phages/physiology , Wastewater/microbiology , Wastewater/virology
SELECTION OF CITATIONS
SEARCH DETAIL