Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.825
Filter
3.
Nat Commun ; 15(1): 4704, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830845

ABSTRACT

Metabolic syndrome encompasses amongst other conditions like obesity and type-2 diabetes and is associated with gut microbiome (GM) dysbiosis. Fecal microbiota transplantation (FMT) has been explored to treat metabolic syndrome by restoring the GM; however, concerns on accidentally transferring pathogenic microbes remain. As a safer alternative, fecal virome transplantation (FVT, sterile-filtrated feces) has the advantage over FMT in that mainly bacteriophages are transferred. FVT from lean male donors have shown promise in alleviating the metabolic effects of high-fat diet in a preclinical mouse study. However, FVT still carries the risk of eukaryotic viral infections. To address this, recently developed methods are applied for removing or inactivating eukaryotic viruses in the viral component of FVT. Modified FVTs are compared with unmodified FVT and saline in a diet-induced obesity model on male C57BL/6 N mice. Contrasted with obese control, mice administered a modified FVT (nearly depleted for eukaryotic viruses) exhibits enhanced blood glucose clearance but not weight loss. The unmodified FVT improves liver pathology and reduces the proportions of immune cells in the adipose tissue with a non-uniform response. GM analysis suggests that bacteriophage-mediated GM modulation influences outcomes. Optimizing these approaches could lead to the development of safe bacteriophage-based therapies targeting metabolic syndrome through GM restoration.


Subject(s)
Diet, High-Fat , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Metabolic Syndrome , Mice, Inbred C57BL , Mice, Obese , Obesity , Virome , Animals , Male , Metabolic Syndrome/therapy , Obesity/therapy , Mice , Diet, High-Fat/adverse effects , Dysbiosis/therapy , Feces/virology , Feces/microbiology , Bacteriophages/physiology , Blood Glucose/metabolism , Disease Models, Animal , Liver/pathology , Liver/metabolism , Adipose Tissue
4.
Arch Virol ; 169(7): 142, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851653

ABSTRACT

Uropathogenic Escherichia coli (UPEC) is the most common causative agent of urinary tract infections, and strains that are resistant to antibiotics are a major problem in treating these infections. Phage therapy is a promising alternative approach that can be used to treat infections caused by polyresistant bacterial strains. In the present study, 16 bacteriophages isolated from sewage and surface water were investigated. Phage host specificity was tested on a collection of 77 UPEC strains. The phages infected 2-44 strains, and 80% of the strains were infected by at least one phage. The susceptible E. coli strains belonged predominantly to the B2 phylogenetic group, including strains of two clones, CC131 and CC73, that have a worldwide distribution. All of the phages belonged to class Caudoviricetes and were identified as members of the families Straboviridae, Autographiviridae, and Drexlerviridae and the genera Kagunavirus, Justusliebigvirus, and Murrayvirus. A phage cocktail composed of six phages - four members of the family Straboviridae and two members of the family Autographiviridae - was prepared, and its antibacterial activity was tested in liquid medium. Complete suppression of bacterial growth was observed after 5-22 hours of cultivation, followed by partial regrowth. At 24 hours postinfection, the cocktail suppressed bacterial growth to 43-92% of control values. Similar results were obtained when testing the activity of the phage cocktail in LB and in artificial urine medium. The results indicate that our phage cocktail has potential to inhibit bacterial growth during infection, and they will therefore be preserved in the national phage bank, serving as valuable resources for therapeutic applications.


Subject(s)
Drug Resistance, Multiple, Bacterial , Host Specificity , Phylogeny , Uropathogenic Escherichia coli , Uropathogenic Escherichia coli/virology , Uropathogenic Escherichia coli/drug effects , Bacteriophages/classification , Bacteriophages/physiology , Bacteriophages/genetics , Bacteriophages/isolation & purification , Sewage/virology , Phage Therapy/methods , Humans , Anti-Bacterial Agents/pharmacology , Escherichia coli Infections/microbiology , Escherichia coli Infections/therapy
5.
Nat Microbiol ; 9(6): 1434-1453, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38834776

ABSTRACT

In contrast to the many reports of successful real-world cases of personalized bacteriophage therapy (BT), randomized controlled trials of non-personalized bacteriophage products have not produced the expected results. Here we present the outcomes of a retrospective observational analysis of the first 100 consecutive cases of personalized BT of difficult-to-treat infections facilitated by a Belgian consortium in 35 hospitals, 29 cities and 12 countries during the period from 1 January 2008 to 30 April 2022. We assessed how often personalized BT produced a positive clinical outcome (general efficacy) and performed a regression analysis to identify functional relationships. The most common indications were lower respiratory tract, skin and soft tissue, and bone infections, and involved combinations of 26 bacteriophages and 6 defined bacteriophage cocktails, individually selected and sometimes pre-adapted to target the causative bacterial pathogens. Clinical improvement and eradication of the targeted bacteria were reported for 77.2% and 61.3% of infections, respectively. In our dataset of 100 cases, eradication was 70% less probable when no concomitant antibiotics were used (odds ratio = 0.3; 95% confidence interval = 0.127-0.749). In vivo selection of bacteriophage resistance and in vitro bacteriophage-antibiotic synergy were documented in 43.8% (7/16 patients) and 90% (9/10) of evaluated patients, respectively. We observed a combination of antibiotic re-sensitization and reduced virulence in bacteriophage-resistant bacterial isolates that emerged during BT. Bacteriophage immune neutralization was observed in 38.5% (5/13) of screened patients. Fifteen adverse events were reported, including seven non-serious adverse drug reactions suspected to be linked to BT. While our analysis is limited by the uncontrolled nature of these data, it indicates that BT can be effective in combination with antibiotics and can inform the design of future controlled clinical trials. BT100 study, ClinicalTrials.gov registration: NCT05498363 .


Subject(s)
Anti-Bacterial Agents , Bacterial Infections , Bacteriophages , Phage Therapy , Humans , Retrospective Studies , Phage Therapy/methods , Bacteriophages/physiology , Bacteriophages/genetics , Female , Male , Middle Aged , Anti-Bacterial Agents/therapeutic use , Adult , Bacterial Infections/therapy , Treatment Outcome , Aged , Precision Medicine/methods , Adolescent , Young Adult , Bacteria/virology , Bacteria/genetics , Child , Aged, 80 and over , Child, Preschool , Belgium , Infant
6.
Curr Microbiol ; 81(7): 215, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849666

ABSTRACT

Non-tailed icosahedral phages belonging to families Fiersviridae (phages MS2 and Qbeta), Tectiviridae (PRD1) and Microviridae (phiX174) have not been considered in detail so far as potential antibacterial agents. The aim of the study was to examine various aspects of the applicability of these phages as antibacterial agents. Antibacterial potential of four phages was investigated via bacterial growth and biofilm formation inhibition, lytic spectra determination, and phage safety examination. The phage phiX174 was combined with different classes of antibiotics to evaluate potential synergistic interactions. In addition, the incidence of phiX174-insensitive mutants was analyzed. The results showed that only phiX174 out of four phages tested against their corresponding hosts inhibited bacterial growth for > 90% at different multiplicity of infection and that only this phage considerably prevented biofilm formation. Although all phages show the absence of potentially undesirable genes, they also have extremely narrow lytic spectra. The synergism was determined between phage phiX174 and ceftazidime, ceftriaxone, ciprofloxacin, macrolides, and chloramphenicol. It was shown that the simultaneous application of agents is more effective than successive treatment, where one agent is applied first. The analysis of the appearance of phiX174 bacteriophage-insensitive mutants showed that mutations occur with a frequency of 10-3. The examined non-tailed phages have a limited potential for use as antibacterial agents, primarily due to a very narrow lytic spectrum and the high frequency of resistant mutants appearance, but Microviridae can be considered in the future as biocontrol agents against susceptible strains of E. coli in combinations with conventional antimicrobial agents.


Subject(s)
Anti-Bacterial Agents , Biofilms , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Bacteriophages/genetics , Bacteriophages/physiology , Escherichia coli/virology , Escherichia coli/drug effects , Bacteriophage phi X 174/drug effects , Bacteriophage phi X 174/genetics , Bacteria/drug effects , Bacteria/virology , Mutation
7.
Science ; 384(6701): eado0713, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38870284

ABSTRACT

Bacteria can repurpose their own bacteriophage viruses (phage) to kill competing bacteria. Phage-derived elements are frequently strain specific in their killing activity, although there is limited evidence that this specificity drives bacterial population dynamics. Here, we identified intact phage and their derived elements in a metapopulation of wild plant-associated Pseudomonas genomes. We discovered that the most abundant viral cluster encodes a phage remnant resembling a phage tail called a tailocin, which bacteria have co-opted to kill bacterial competitors. Each pathogenic Pseudomonas strain carries one of a few distinct tailocin variants that target the variable polysaccharides in the outer membrane of co-occurring pathogenic Pseudomonas strains. Analysis of herbarium samples from the past 170 years revealed that the same tailocin and bacterial receptor variants have persisted in Pseudomonas populations. These results suggest that tailocin genetic diversity can be mined to develop targeted "tailocin cocktails" for microbial control.


Subject(s)
Bacteriocins , Pseudomonas , Bacteriocins/pharmacology , Bacteriocins/metabolism , Bacteriocins/genetics , Pseudomonas/metabolism , Pseudomonas Phages/genetics , Pseudomonas Phages/metabolism , Bacteriophages/genetics , Bacteriophages/physiology , Genetic Variation , Viral Tail Proteins/metabolism , Viral Tail Proteins/genetics , Bacterial Outer Membrane/metabolism , Genome, Bacterial , Polysaccharides, Bacterial/metabolism , Antibiosis
8.
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
9.
BMC Microbiol ; 24(1): 211, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877452

ABSTRACT

BACKGROUND: This study investigates the effectiveness of the bacteriophage KZag1 against drug-resistant Klebsiella pneumoniae, aiming to assess its potential as a therapeutic agent. The novelty lies in the characterization of KZag1, a Myovirus with specific efficacy against multidrug-resistant K. pneumoniae strains. This highlights the significance of exploring alternative strategies, particularly phage therapy, in addressing biofilm-associated infections. METHODS: KZag1, characterized by a typical Myovirus structure with a 75 ± 5 nm diameter icosahedral head and a 15 ± 5 nm short tail, was evaluated in experimental trials against 15 strains of K. pneumoniae. The infection cycle duration was determined to be 50 min, resulting in an estimated burst size of approximately 83 plaque-forming units per colony-forming unit (PFU/CFU). Stability assessments were conducted within a pH range of 4 to 12 and temperatures ranging from 45°C to 60°C. Biofilm biomass reduction was observed, particularly at a multiplicity of infection (MOI) of 10. RESULTS: KZag1 demonstrated infection efficacy against 12 out of 15 tested K. pneumoniae strains. The phage exhibited stability across a broad pH range and at elevated temperatures. Notably, treatment with KZag1 significantly reduced K. pneumoniae biofilm biomass, emphasizing its potential in combating biofilm formation. Genomic analysis revealed a complete genome of 157,089 base pairs with a GC content of 46.38%, encompassing 203 open reading frames (ORFs) and a cysteine-specific tRNA sequence. Comparison with phage GP4 highlighted similarities, with KZag1 having a longer genome by approximately 4829 base pairs and a higher GC content by approximately 0.93%. Phylogenetic analysis classified KZag1 within the Myoviridae family. CONCLUSION: The efficacy of KZag1 against K. pneumoniae biofilm suggests its potential as a therapeutic candidate, especially for drug-resistant infections. Further clinical research is warranted to explore its synergy with other treatments, elucidate genomic traits, compare with Myoviridae phages, and understand its host interactions. These findings underscore the promising role of KZag1 in addressing drug-resistant bacterial infections.


Subject(s)
Bacteriophages , Biofilms , Genome, Viral , Klebsiella pneumoniae , Klebsiella pneumoniae/virology , Klebsiella pneumoniae/genetics , Biofilms/growth & development , Bacteriophages/genetics , Bacteriophages/physiology , Bacteriophages/classification , Bacteriophages/isolation & purification , Myoviridae/genetics , Myoviridae/physiology , Myoviridae/classification , Drug Resistance, Multiple, Bacterial/genetics , Phylogeny , DNA, Viral/genetics , Base Composition , Phage Therapy
10.
Microb Biotechnol ; 17(6): e14507, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38884488

ABSTRACT

Pathogens resistant to classical control strategies pose a significant threat to crop yield, with seeds being a major transmission route. Bacteriophages, viruses targeting bacteria, offer an environmentally sustainable biocontrol solution. In this study, we isolated and characterized two novel phages, Athelas and Alfirin, which infect Pseudomonas syringae and Agrobacterium fabrum, respectively, and included the recently published Pfeifenkraut phage infecting Xanthomonas translucens. Using a simple immersion method, phages coated onto seeds successfully lysed bacteria post air-drying. The seed coat mucilage (SCM), a polysaccharide-polymer matrix exuded by seeds, plays a critical role in phage binding. Seeds with removed mucilage formed five to 10 times less lysis zones compared to those with mucilage. The podovirus Athelas showed the highest mucilage dependency. Phages from the Autographiviridae family also depended on mucilage for seed adhesion. Comparative analysis of Arabidopsis SCM mutants suggested the diffusible cellulose as a key component for phage binding. Long-term activity tests demonstrated high phage stability on seed surfaces and significantly increasing seedling survival rates in the presence of pathogens. Using non-virulent host strains enhanced phage presence on seeds but also has potential limitations. These findings highlight phage-based interventions as promising, sustainable strategies for combating pathogen resistance and improving crop yield.


Subject(s)
Arabidopsis , Bacteriophages , Plant Diseases , Pseudomonas syringae , Seeds , Seeds/microbiology , Seeds/virology , Pseudomonas syringae/virology , Plant Diseases/microbiology , Plant Diseases/prevention & control , Plant Diseases/virology , Bacteriophages/physiology , Bacteriophages/genetics , Arabidopsis/virology , Arabidopsis/microbiology , Xanthomonas/virology , Plant Mucilage/metabolism , Plant Mucilage/chemistry , Biological Control Agents , Virus Attachment
11.
BMC Microbiol ; 24(1): 155, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38704526

ABSTRACT

BACKGROUND: The in-depth understanding of the role of lateral genetic transfer (LGT) in phage-prophage interactions is essential to rationalizing phage applications for human and animal therapy, as well as for food and environmental safety. This in silico study aimed to detect LGT between phages of potential industrial importance and their hosts. METHODS: A large array of genetic recombination detection algorithms, implemented in SplitsTree and RDP4, was applied to detect LGT between various Escherichia, Listeria, Salmonella, Campylobacter, Staphylococcus, Pseudomonas, and Vibrio phages and their hosts. PHASTER and RAST were employed respectively to identify prophages across the host genome and to annotate LGT-affected genes with unknown functions. PhageAI was used to gain deeper insights into the life cycle history of recombined phages. RESULTS: The split decomposition inferences (bootstrap values: 91.3-100; fit: 91.433-100), coupled with the Phi (0.0-2.836E-12) and RDP4 (P being well below 0.05) statistics, provided strong evidence for LGT between certain Escherichia, Listeria, Salmonella, and Campylobacter virulent phages and prophages of their hosts. The LGT events entailed mainly the phage genes encoding for hypothetical proteins, while some of these genetic loci appeared to have been affected even by intergeneric recombination in specific E. coli and S. enterica virulent phages when interacting with their host prophages. Moreover, it is shown that certain L. monocytogenes virulent phages could serve at least as the donors of the gene loci, involved in encoding for the basal promoter specificity factor, for L. monocytogenes. In contrast, the large genetic clusters were determined to have been simultaneously exchanged by many S. aureus prophages and some Staphylococcus temperate phages proposed earlier as potential therapeutic candidates (in their native or modified state). The above genetic clusters were found to encompass multiple genes encoding for various proteins, such as e.g., phage tail proteins, the capsid and scaffold proteins, holins, and transcriptional terminator proteins. CONCLUSIONS: It is suggested that phage-prophage interactions, mediated by LGT (including intergeneric recombination), can have a far-reaching impact on the co-evolutionary trajectories of industrial phages and their hosts especially when excessively present across microbially rich environments.


Subject(s)
Prophages , Recombination, Genetic , Prophages/genetics , Campylobacter/virology , Campylobacter/genetics , Staphylococcus/virology , Staphylococcus/genetics , Gene Transfer, Horizontal , Bacteriophages/genetics , Bacteriophages/physiology , Bacteriophages/classification , Listeria/virology , Listeria/genetics , Salmonella/virology , Salmonella/genetics , Evolution, Molecular , Bacteria/virology , Bacteria/genetics
12.
Nat Commun ; 15(1): 3715, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698041

ABSTRACT

Phages play an essential role in controlling bacterial populations. Those infecting Pelagibacterales (SAR11), the dominant bacteria in surface oceans, have been studied in silico and by cultivation attempts. However, little is known about the quantity of phage-infected cells in the environment. Using fluorescence in situ hybridization techniques, we here show pelagiphage-infected SAR11 cells across multiple global ecosystems and present evidence for tight community control of pelagiphages on the SAR11 hosts in a case study. Up to 19% of SAR11 cells were phage-infected during a phytoplankton bloom, coinciding with a ~90% reduction in SAR11 cell abundance within 5 days. Frequently, a fraction of the infected SAR11 cells were devoid of detectable ribosomes, which appear to be a yet undescribed possible stage during pelagiphage infection. We dubbed such cells zombies and propose, among other possible explanations, a mechanism in which ribosomal RNA is used as a resource for the synthesis of new phage genomes. On a global scale, we detected phage-infected SAR11 and zombie cells in the Atlantic, Pacific, and Southern Oceans. Our findings illuminate the important impact of pelagiphages on SAR11 populations and unveil the presence of ribosome-deprived zombie cells as part of the infection cycle.


Subject(s)
Bacteriophages , Ribosomes , Ribosomes/metabolism , Bacteriophages/genetics , Bacteriophages/physiology , Phytoplankton/virology , Phytoplankton/genetics , Phytoplankton/metabolism , In Situ Hybridization, Fluorescence , Alphaproteobacteria/genetics , Alphaproteobacteria/metabolism , Ecosystem , Seawater/microbiology , Seawater/virology , Oceans and Seas
13.
Vet Res ; 55(1): 59, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715095

ABSTRACT

Klebsiella pneumoniae has become one of the most intractable gram-negative pathogens infecting humans and animals due to its severe antibiotic resistance. Bacteriophages and protein products derived from them are receiving increasing amounts of attention as potential alternatives to antibiotics. In this study, we isolated and investigated the characteristics of a new lytic phage, P1011, which lyses K5 K. pneumoniae specifically among 26 serotypes. The K5-specific capsular polysaccharide-degrading depolymerase dep1011 was identified and expressed. By establishing murine infection models using bovine strain B16 (capable of supporting phage proliferation) and human strain KP181 (incapable of sustaining phage expansion), we explored the safety and efficacy of phage and dep1011 treatments against K5 K. pneumoniae. Phage P1011 resulted in a 60% survival rate of the mice challenged with K. pneumoniae supporting phage multiplication, concurrently lowering the bacterial burden in their blood, liver, and lungs. Unexpectedly, even when confronted with bacteria impervious to phage multiplication, phage therapy markedly decreased the number of viable organisms. The protective efficacy of the depolymerase was significantly better than that of the phage. The depolymerase achieved 100% survival in both treatment groups regardless of phage propagation compatibility. These findings indicated that P1011 and dep1011 might be used as potential antibacterial agents to control K5 K. pneumoniae infection.


Subject(s)
Bacteriophages , Klebsiella Infections , Klebsiella pneumoniae , Animals , Klebsiella pneumoniae/virology , Klebsiella pneumoniae/physiology , Mice , Klebsiella Infections/therapy , Klebsiella Infections/veterinary , Klebsiella Infections/microbiology , Bacteriophages/physiology , Disease Models, Animal , Phage Therapy , Female , Glycoside Hydrolases/metabolism , Cattle
14.
Front Immunol ; 15: 1398652, 2024.
Article in English | MEDLINE | ID: mdl-38779682

ABSTRACT

In the advancement of Inflammatory Bowel Disease (IBD) treatment, existing therapeutic methods exhibit limitations; they do not offer a complete cure for IBD and can trigger adverse side effects. Consequently, the exploration of novel therapies and multifaceted treatment strategies provides patients with a broader range of options. Within the framework of IBD, gut microbiota plays a pivotal role in disease onset through diverse mechanisms. Bacteriophages, as natural microbial regulators, demonstrate remarkable specificity by accurately identifying and eliminating specific pathogens, thus holding therapeutic promise. Although clinical trials have affirmed the safety of phage therapy, its efficacy is prone to external influences during storage and transport, which may affect its infectivity and regulatory roles within the microbiota. Improving the stability and precise dosage control of bacteriophages-ensuring robustness in storage and transport, consistent dosing, and targeted delivery to infection sites-is crucial. This review thoroughly explores the latest developments in IBD treatment and its inherent challenges, focusing on the interaction between the microbiota and bacteriophages. It highlights bacteriophages' potential as microbiome modulators in IBD treatment, offering detailed insights into research on bacteriophage encapsulation and targeted delivery mechanisms. Particular attention is paid to the functionality of various carrier systems, especially regarding their protective properties and ability for colon-specific delivery. This review aims to provide a theoretical foundation for using bacteriophages as microbiome modulators in IBD treatment, paving the way for enhanced regulation of the intestinal microbiota.


Subject(s)
Bacteriophages , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Phage Therapy , Humans , Phage Therapy/methods , Inflammatory Bowel Diseases/therapy , Bacteriophages/physiology , Animals
15.
Cell Host Microbe ; 32(5): 634-636, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38723602

ABSTRACT

Bacterial genomes are littered with exogenous: competing DNA elements. Here, Sprenger et al. demonstrate that the Vibrio cholerae prophage VP882 modulates host functions via production of regulatory sRNAs to promote phage development. Alternatively, host sRNAs inhibit the VP882 lytic phase by specifically regulating phage genes.


Subject(s)
Prophages , Vibrio cholerae , Vibrio cholerae/genetics , Prophages/genetics , Prophages/physiology , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Genome, Bacterial , Bacteriophages/genetics , Bacteriophages/physiology , Gene Expression Regulation, Bacterial , RNA, Bacterial/genetics , RNA, Bacterial/metabolism
16.
Front Cell Infect Microbiol ; 14: 1382145, 2024.
Article in English | MEDLINE | ID: mdl-38736748

ABSTRACT

Carbapenem-resistant Acinetobacter baumannii (CRAB) has become a new threat in recent years, owing to its rapidly increasing resistance to antibiotics and new effective therapies are needed to combat this pathogen. Phage therapy is considered to be the most promising alternative for treating CRAB infections. In this study, a novel phage, Ab_WF01, which can lyse clinical CRAB, was isolated and characterized from hospital sewage. The multiplicity of infection, morphology, one-step growth curve, stability, sensitivity, and lytic activity of the phage were also investigated. The genome of phage Ab_WF01 was 41, 317 bp in size with a GC content of 39.12% and encoded 51 open reading frames (ORFs). tRNA, virulence, and antibiotic resistance genes were not detected in the phage genome. Comparative genomic and phylogenetic analyses suggest that phage Ab_WF01 is a novel species of the genus Friunavirus, subfamily Beijerinckvirinae, and family Autographiviridae. The in vivo results showed that phage Ab_WF01 significantly increased the survival rate of CRAB-infected Galleria mellonella (from 0% to 70% at 48 h) and mice (from 0% to 60% for 7 days). Moreover, after day 3 post-infection, phage Ab_WF01 reduced inflammatory response, with strongly ameliorated histological damage and bacterial clearance in infected tissue organs (lungs, liver, and spleen) in mouse CRAB infection model. Taken together, these results show that phage Ab_WF01 holds great promise as a potential alternative agent with excellent stability for against CRAB infections.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Bacteriophages , Carbapenems , Genome, Viral , Phage Therapy , Phylogeny , Sewage , Acinetobacter baumannii/virology , Acinetobacter baumannii/drug effects , Sewage/virology , Sewage/microbiology , Animals , Carbapenems/pharmacology , Bacteriophages/genetics , Bacteriophages/physiology , Bacteriophages/classification , Bacteriophages/isolation & purification , Acinetobacter Infections/microbiology , Mice , Anti-Bacterial Agents/pharmacology , Open Reading Frames , Disease Models, Animal , Moths/virology , Moths/microbiology , Base Composition
17.
Parasit Vectors ; 17(1): 222, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38745242

ABSTRACT

BACKGROUND: Culex pipiens pallens is a well-known mosquito vector for several diseases. Deltamethrin, a commonly used pyrethroid insecticide, has been frequently applied to manage adult Cx. pipiens pallens. However, mosquitoes can develop resistance to these insecticides as a result of insecticide misuse and, therefore, it is crucial to identify novel methods to control insecticide resistance. The relationship between commensal bacteria and vector resistance has been recently recognized. Bacteriophages (= phages) are effective tools by which to control insect commensal bacteria, but there have as yet been no studies using phages on adult mosquitoes. In this study, we isolated an Aeromonas phage vB AhM-LH that specifically targets resistance-associated symbiotic bacteria in mosquitoes. We investigated the impact of Aeromonas phage vB AhM-LH in an abundance of Aeromonas hydrophila in the gut of Cx. pipiens pallens and its effect on the status of deltamethrin resistance. METHODS: Phages were isolated on double-layer agar plates and their biological properties analyzed. Phage morphology was observed by transmission electron microscopy (TEM) after negative staining. The phage was then introduced into the mosquito intestines via oral feeding. The inhibitory effect of Aeromonas phage vB AhM-LH on Aeromonas hydrophila in mosquito intestines was assessed through quantitative real-time PCR analysis. Deltamethrin resistance of mosquitoes was assessed using WHO bottle bioassays. RESULTS: An Aeromonas phage vB AhM-LH was isolated from sewage and identified as belonging to the Myoviridae family in the order Caudovirales using TEM. Based on biological characteristics analysis and in vitro antibacterial experiments, Aeromonas phage vB AhM-LH was observed to exhibit excellent stability and effective bactericidal activity. Sequencing revealed that the Aeromonas phage vB AhM-LH genome comprises 43,663 bp (51.6% CG content) with 81 predicted open reading frames. No integrase-related gene was detected in the vB AH-LH genome, which marked it as a potential biological antibacterial. Finally, we found that Aeromonas phage vB AhM-LH could significantly reduce deltamethrin resistance in Cx. pipiens pallens, in both the laboratory and field settings, by decreasing the abundance of Aeromonas hydrophila in their midgut. CONCLUSIONS: Our findings demonstrate that Aeromonas phage vB AhM-LH could effectively modulate commensal bacteria Aeromonas hydrophila in adult mosquitoes, thus representing a promising strategy to mitigate mosquito vector resistance.


Subject(s)
Aeromonas hydrophila , Bacteriophages , Culex , Insecticide Resistance , Nitriles , Pyrethrins , Animals , Aeromonas hydrophila/virology , Aeromonas hydrophila/drug effects , Culex/virology , Culex/microbiology , Bacteriophages/physiology , Bacteriophages/isolation & purification , Bacteriophages/genetics , Pyrethrins/pharmacology , Nitriles/pharmacology , Insecticides/pharmacology , Mosquito Vectors/virology , Mosquito Vectors/microbiology , Female
18.
Commun Biol ; 7(1): 590, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755280

ABSTRACT

Infection of bacteria by phages is a complex multi-step process that includes specific recognition of the host cell, creation of a temporary breach in the host envelope, and ejection of viral DNA into the bacterial cytoplasm. These steps must be perfectly regulated to ensure efficient infection. Here we report the dual function of the tail completion protein gp16.1 of bacteriophage SPP1. First, gp16.1 has an auxiliary role in assembly of the tail interface that binds to the capsid connector. Second, gp16.1 is necessary to ensure correct routing of phage DNA to the bacterial cytoplasm. Viral particles assembled without gp16.1 are indistinguishable from wild-type virions and eject DNA normally in vitro. However, they release their DNA to the extracellular space upon interaction with the host bacterium. The study shows that a highly conserved tail completion protein has distinct functions at two essential steps of the virus life cycle in long-tailed phages.


Subject(s)
Viral Tail Proteins , Viral Tail Proteins/metabolism , Viral Tail Proteins/genetics , Bacteriophages/genetics , Bacteriophages/physiology , Bacteriophages/metabolism , DNA, Viral/metabolism , DNA, Viral/genetics , Virion/metabolism
19.
BMC Infect Dis ; 24(1): 497, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755537

ABSTRACT

BACKGROUND: In recent years, there has been a growing interest in phage therapy as an effective therapeutic tool against colibacillosis caused by avian pathogenic Escherichia coli (APEC) which resulted from the increasing number of multidrug resistant (MDR) APEC strains. METHODS: In the present study, we reported the characterization of a new lytic bacteriophage (Escherichia phage AG- MK-2022. Basu) isolated from poultry slaughterhouse wastewater. In addition, the in vitro bacteriolytic activity of the newly isolated phage (Escherichia phage AG- MK-2022. Basu) and the Escherichia phage VaT-2019a isolate PE17 (GenBank: MK353636.1) were assessed against MDR- APEC strains (n = 100) isolated from broiler chickens with clinical signs of colibacillosis. RESULTS: Escherichia phage AG- MK-2022. Basu belongs to the Myoviridae family and exhibits a broad host range. Furthermore, the phage showed stability under a wide range of temperatures, pH values and different concentrations of NaCl. Genome analysis of the Escherichia phage AG- MK-2022. Basu revealed that the phage possesses no antibiotic resistance genes (ARGs), mobile genetic elements (MGEs), and any E. coli virulence associated genes. In vitro bacterial challenge tests demonstrated that two phages, the Escherichia phage VaT-2019a isolate PE17 and the Escherichia phage AG- MK-2022. Basu exhibited high bactericidal activity against APEC strains and lysed 95% of the tested APEC strains. CONCLUSIONS: The current study findings indicate that both phages could be suggested as safe biocontrol agents and alternatives to antibiotics for controlling MDR-APEC strains isolated from broilers.


Subject(s)
Chickens , Drug Resistance, Multiple, Bacterial , Escherichia coli Infections , Escherichia coli , Phage Therapy , Poultry Diseases , Animals , Escherichia coli/virology , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli Infections/microbiology , Escherichia coli Infections/veterinary , Chickens/microbiology , Poultry Diseases/microbiology , Coliphages/genetics , Coliphages/physiology , Host Specificity , Genome, Viral , Wastewater/microbiology , Wastewater/virology , Myoviridae/genetics , Myoviridae/isolation & purification , Myoviridae/physiology , Myoviridae/classification , Bacteriophages/genetics , Bacteriophages/physiology , Bacteriophages/isolation & purification
20.
Nat Commun ; 15(1): 3474, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750067

ABSTRACT

Evidence from the International Space Station suggests microbial populations are rapidly adapting to the spacecraft environment; however, the mechanism of this adaptation is not understood. Bacteriophages are prolific mediators of bacterial adaptation on Earth. Here we survey 245 genomes sequenced from bacterial strains isolated on the International Space Station for dormant (lysogenic) bacteriophages. Our analysis indicates phage-associated genes are significantly different between spaceflight strains and their terrestrial counterparts. In addition, we identify 283 complete prophages, those that could initiate bacterial lysis and infect additional hosts, of which 21% are novel. These prophage regions encode functions that correlate with increased persistence in extreme environments, such as spaceflight, to include antimicrobial resistance and virulence, DNA damage repair, and dormancy. Our results correlate microbial adaptation in spaceflight to bacteriophage-encoded functions that may impact human health in spaceflight.


Subject(s)
Adaptation, Physiological , Bacteria , Bacteriophages , Space Flight , Bacteria/virology , Bacteria/genetics , Bacteriophages/genetics , Bacteriophages/physiology , Prophages/genetics , Prophages/physiology , Humans , Virulence/genetics , Genome, Bacterial/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...