Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.780
Filtrar
1.
Parasit Vectors ; 17(1): 222, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38745242

RESUMEN

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.


Asunto(s)
Aeromonas hydrophila , Bacteriófagos , Culex , Resistencia a los Insecticidas , Nitrilos , Piretrinas , Animales , Aeromonas hydrophila/virología , Aeromonas hydrophila/efectos de los fármacos , Culex/virología , Culex/microbiología , Bacteriófagos/fisiología , Bacteriófagos/aislamiento & purificación , Bacteriófagos/genética , Piretrinas/farmacología , Nitrilos/farmacología , Insecticidas/farmacología , Mosquitos Vectores/virología , Mosquitos Vectores/microbiología , Femenino
2.
J Med Microbiol ; 73(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38743467

RESUMEN

Introduction . Acinetobacter baumannii is a critical priority pathogen for novel antimicrobials (World Health Organization) because of the rise in nosocomial infections and its ability to evolve resistance to last resort antibiotics. A. baumannii is thus a priority target for phage therapeutics. Two strains of a novel, virulent bacteriophage (LemonAid and Tonic) able to infect carbapenem-resistant A. baumannii (strain NCTC 13420), were isolated from environmental water samples collected through a citizen science programme.Gap statement. Phage-host coevolution can lead to emergence of host resistance, with a concomitant reduction in the virulence of host bacteria; a potential benefit to phage therapy applications.Methodology. In vitro and in vivo assays, genomics and microscopy techniques were used to characterize the phages; determine mechanisms and impact of phage resistance on host virulence, and the efficacy of the phages against A. baumannii.Results. A. baumannii developed resistance to both viruses, LemonAid and Tonic. Resistance came at a cost to virulence, with the resistant variants causing significantly reduced mortality in a Galleria mellonella larval in vivo model. A replicated 8 bp insertion increased in frequency (~40 % higher frequency than in the wild-type) within phage-resistant A. baumannii mutants, putatively resulting in early truncation of a protein of unknown function. Evidence from comparative genomics and an adsorption assay suggests this protein acts as a novel phage receptor site in A. baumannii. We find no evidence linking resistance to changes in capsule structure, a known virulence factor. LemonAid efficiently suppressed growth of A. baumanni in vitro across a wide range of titres. However, in vivo, while survival of A. baumannii infected larvae significantly increased with both remedial and prophylactic treatment with LemonAid (107 p.f.u. ml-1), the effect was weak and not sufficient to save larvae from morbidity and mortality.Conclusion. While LemonAid and Tonic did not prove effective as a treatment in a Galleria larvae model, there is potential to harness their ability to attenuate virulence in drug-resistant A. baumannii.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Bacteriófagos , Acinetobacter baumannii/virología , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/patogenicidad , Acinetobacter baumannii/genética , Bacteriófagos/genética , Bacteriófagos/fisiología , Virulencia , Infecciones por Acinetobacter/microbiología , Animales , Mariposas Nocturnas/microbiología , Mariposas Nocturnas/virología , Terapia de Fagos , Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Larva/microbiología , Larva/virología
3.
Vet Res ; 55(1): 59, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715095

RESUMEN

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.


Asunto(s)
Bacteriófagos , Infecciones por Klebsiella , Klebsiella pneumoniae , Animales , Klebsiella pneumoniae/virología , Klebsiella pneumoniae/fisiología , Ratones , Infecciones por Klebsiella/terapia , Infecciones por Klebsiella/veterinaria , Infecciones por Klebsiella/microbiología , Bacteriófagos/fisiología , Modelos Animales de Enfermedad , Terapia de Fagos , Femenino , Glicósido Hidrolasas/metabolismo , Bovinos
4.
Cell Host Microbe ; 32(5): 634-636, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38723602

RESUMEN

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.


Asunto(s)
Profagos , Vibrio cholerae , Vibrio cholerae/genética , Profagos/genética , Profagos/fisiología , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Genoma Bacteriano , Bacteriófagos/genética , Bacteriófagos/fisiología , Regulación Bacteriana de la Expresión Génica , ARN Bacteriano/genética , ARN Bacteriano/metabolismo
5.
Nat Commun ; 15(1): 3715, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38698041

RESUMEN

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.


Asunto(s)
Bacteriófagos , Ribosomas , Ribosomas/metabolismo , Bacteriófagos/genética , Bacteriófagos/fisiología , Fitoplancton/virología , Fitoplancton/genética , Fitoplancton/metabolismo , Hibridación Fluorescente in Situ , Alphaproteobacteria/genética , Alphaproteobacteria/metabolismo , Ecosistema , Agua de Mar/microbiología , Agua de Mar/virología , Océanos y Mares
6.
BMC Microbiol ; 24(1): 155, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704526

RESUMEN

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.


Asunto(s)
Profagos , Recombinación Genética , Profagos/genética , Campylobacter/virología , Campylobacter/genética , Staphylococcus/virología , Staphylococcus/genética , Transferencia de Gen Horizontal , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/clasificación , Listeria/virología , Listeria/genética , Salmonella/virología , Salmonella/genética , Evolución Molecular , Bacterias/virología , Bacterias/genética
7.
Front Cell Infect Microbiol ; 14: 1382145, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38736748

RESUMEN

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.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Bacteriófagos , Carbapenémicos , Genoma Viral , Terapia de Fagos , Filogenia , Aguas del Alcantarillado , Acinetobacter baumannii/virología , Acinetobacter baumannii/efectos de los fármacos , Aguas del Alcantarillado/virología , Aguas del Alcantarillado/microbiología , Animales , Carbapenémicos/farmacología , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/clasificación , Bacteriófagos/aislamiento & purificación , Infecciones por Acinetobacter/microbiología , Ratones , Antibacterianos/farmacología , Sistemas de Lectura Abierta , Modelos Animales de Enfermedad , Mariposas Nocturnas/virología , Mariposas Nocturnas/microbiología , Composición de Base
8.
Microbiology (Reading) ; 170(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38739436

RESUMEN

Endolysins are bacteriophage (or phage)-encoded enzymes that catalyse the peptidoglycan breakdown in the bacterial cell wall. The exogenous action of recombinant phage endolysins against Gram-positive organisms has been extensively studied. However, the outer membrane acts as a physical barrier when considering the use of recombinant endolysins to combat Gram-negative bacteria. This study aimed to evaluate the antimicrobial activity of the SAR-endolysin LysKpV475 against Gram-negative bacteria as single or combined therapies, using an outer membrane permeabilizer (polymyxin B) and a phage, free or immobilized in a pullulan matrix. In the first step, the endolysin LysKpV475 in solution, alone and combined with polymyxin B, was tested in vitro and in vivo against ten Gram-negative bacteria, including highly virulent strains and multidrug-resistant isolates. In the second step, the lyophilized LysKpV475 endolysin was combined with the phage phSE-5 and investigated, free or immobilized in a pullulan matrix, against Salmonella enterica subsp. enterica serovar Typhimurium ATCC 13311. The bacteriostatic action of purified LysKpV475 varied between 8.125 µg ml-1 against Pseudomonas aeruginosa ATCC 27853, 16.25 µg ml-1 against S. enterica Typhimurium ATCC 13311, and 32.50 µg ml-1 against Klebsiella pneumoniae ATCC BAA-2146 and Enterobacter cloacae P2224. LysKpV475 showed bactericidal activity only for P. aeruginosa ATCC 27853 (32.50 µg ml-1) and P. aeruginosa P2307 (65.00 µg ml-1) at the tested concentrations. The effect of the LysKpV475 combined with polymyxin B increased against K. pneumoniae ATCC BAA-2146 [fractional inhibitory concentration index (FICI) 0.34; a value lower than 1.0 indicates an additive/combined effect] and S. enterica Typhimurium ATCC 13311 (FICI 0.93). A synergistic effect against S. enterica Typhimurium was also observed when the lyophilized LysKpV475 at ⅔ MIC was combined with the phage phSE-5 (m.o.i. of 100). The lyophilized LysKpV475 immobilized in a pullulan matrix maintained a significant Salmonella reduction of 2 logs after 6 h of treatment. These results demonstrate the potential of SAR-endolysins, alone or in combination with other treatments, in the free form or immobilized in solid matrices, which paves the way for their application in different areas, such as in biocontrol at the food processing stage, biosanitation of food contact surfaces and biopreservation of processed food in active food packing.


Asunto(s)
Antibacterianos , Endopeptidasas , Glucanos , Polimixina B , Fagos de Salmonella , Endopeptidasas/farmacología , Endopeptidasas/química , Endopeptidasas/metabolismo , Polimixina B/farmacología , Antibacterianos/farmacología , Antibacterianos/química , Fagos de Salmonella/genética , Fagos de Salmonella/fisiología , Fagos de Salmonella/química , Glucanos/química , Glucanos/farmacología , Animales , Pruebas de Sensibilidad Microbiana , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/virología , Ratones , Salmonella typhimurium/virología , Salmonella typhimurium/efectos de los fármacos , Bacteriófagos/fisiología , Bacteriófagos/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Virales/farmacología , Proteínas Virales/química
9.
Int J Mol Sci ; 25(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732145

RESUMEN

Bacteria and their phage adversaries are engaged in an ongoing arms race, resulting in the development of a broad antiphage arsenal and corresponding viral countermeasures. In recent years, the identification and utilization of CRISPR-Cas systems have driven a renewed interest in discovering and characterizing antiphage mechanisms, revealing a richer diversity than initially anticipated. Currently, these defense systems can be categorized based on the bacteria's strategy associated with the infection cycle stage. Thus, bacterial defense systems can degrade the invading genetic material, trigger an abortive infection, or inhibit genome replication. Understanding the molecular mechanisms of processes related to bacterial immunity has significant implications for phage-based therapies and the development of new biotechnological tools. This review aims to comprehensively cover these processes, with a focus on the most recent discoveries.


Asunto(s)
Bacterias , Bacteriófagos , Sistemas CRISPR-Cas , Bacterias/genética , Bacteriófagos/fisiología , Bacteriófagos/genética , Farmacorresistencia Bacteriana/genética , Humanos , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología
10.
Nat Microbiol ; 9(5): 1312-1324, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38565896

RESUMEN

Dormant prophages protect lysogenic cells by expressing diverse immune systems, which must avoid targeting their cognate prophages upon activation. Here we report that multiple Staphylococcus aureus prophages encode Tha (tail-activated, HEPN (higher eukaryotes and prokaryotes nucleotide-binding) domain-containing anti-phage system), a defence system activated by structural tail proteins of incoming phages. We demonstrate the function of two Tha systems, Tha-1 and Tha-2, activated by distinct tail proteins. Interestingly, Tha systems can also block reproduction of the induced tha-positive prophages. To prevent autoimmunity after prophage induction, these systems are inhibited by the product of a small overlapping antisense gene previously believed to encode an excisionase. This genetic organization, conserved in S. aureus prophages, allows Tha systems to protect prophages and their bacterial hosts against phage predation and to be turned off during prophage induction, balancing immunity and autoimmunity. Our results show that the fine regulation of these processes is essential for the correct development of prophages' life cycle.


Asunto(s)
Profagos , Staphylococcus aureus , Profagos/genética , Staphylococcus aureus/virología , Staphylococcus aureus/inmunología , Autoinmunidad , Lisogenia , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/inmunología , Fagos de Staphylococcus/fisiología , Proteínas de la Cola de los Virus/genética , Proteínas de la Cola de los Virus/metabolismo , Bacteriófagos/genética , Bacteriófagos/inmunología , Bacteriófagos/fisiología
12.
Proc Natl Acad Sci U S A ; 121(16): e2319790121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38593079

RESUMEN

Bacteriophages (phages) play critical roles in modulating microbial ecology. Within the human microbiome, the factors influencing the long-term coexistence of phages and bacteria remain poorly investigated. Saccharibacteria (formerly TM7) are ubiquitous members of the human oral microbiome. These ultrasmall bacteria form episymbiotic relationships with their host bacteria and impact their physiology. Here, we showed that during surface-associated growth, a human oral Saccharibacteria isolate (named TM7x) protects its host bacterium, a Schaalia odontolytica strain (named XH001) against lytic phage LC001 predation. RNA-Sequencing analysis identified in XH001 a gene cluster with predicted functions involved in the biogenesis of cell wall polysaccharides (CWP), whose expression is significantly down-regulated when forming a symbiosis with TM7x. Through genetic work, we experimentally demonstrated the impact of the expression of this CWP gene cluster on bacterial-phage interaction by affecting phage binding. In vitro coevolution experiments further showed that the heterogeneous populations of TM7x-associated and TM7x-free XH001, which display differential susceptibility to LC001 predation, promote bacteria and phage coexistence. Our study highlights the tripartite interaction between the bacterium, episymbiont, and phage. More importantly, we present a mechanism, i.e., episymbiont-mediated modulation of gene expression in host bacteria, which impacts their susceptibility to phage predation and contributes to the formation of "source-sink" dynamics between phage and bacteria in biofilm, promoting their long-term coexistence within the human microbiome.


Asunto(s)
Bacteriófagos , Humanos , Bacteriófagos/fisiología , Simbiosis , Bacterias/genética
13.
Viruses ; 16(4)2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38675852

RESUMEN

Fire blight, caused by the bacterium Erwinia amylovora, is a major threat to pear production worldwide. Bacteriophages, viruses that infect bacteria, are a promising alternative to antibiotics for controlling fire blight. In this study, we isolated a novel bacteriophage, RH-42-1, from Xinjiang, China. We characterized its biological properties, including host range, plaque morphology, infection dynamics, stability, and sensitivity to various chemicals. RH-42-1 infected several E. amylovora strains but not all. It produced clear, uniform plaques and exhibited optimal infectivity at a multiplicity of infection (MOI) of 1, reaching a high titer of 9.6 × 109 plaque-forming units (PFU)/mL. The bacteriophage had a short latent period (10 min), a burst size of 207 PFU/cell, and followed a sigmoidal one-step growth curve. It was stable at temperatures up to 60 °C but declined rapidly at higher temperatures. RH-42-1 remained viable within a pH range of 5 to 9 and was sensitive to extreme pH values. The bacteriophage demonstrates sustained activity upon exposure to ultraviolet radiation for 60 min, albeit with a marginal reduction. In our assays, it exhibited a certain level of resistance to 5% chloroform (CHCl3), 5% isopropanol (C3H8O), and 3% hydrogen peroxide (H2O2), which had little effect on its activity, whereas it showed sensitivity to 75% ethanol (C2H5OH). Electron microscopy revealed that RH-42-1 has a tadpole-shaped morphology. Its genome size is 14,942 bp with a GC content of 48.19%. Based on these characteristics, RH-42-1 was identified as a member of the Tectiviridae family, Alphatectivirus genus. This is the first report of a bacteriophage in this genus with activity against E. amylovora.


Asunto(s)
Bacteriófagos , Erwinia amylovora , Genoma Viral , Especificidad del Huésped , Microbiología del Suelo , Erwinia amylovora/virología , Erwinia amylovora/efectos de los fármacos , China , Bacteriófagos/aislamiento & purificación , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/clasificación , Enfermedades de las Plantas/microbiología , Filogenia , Pyrus/microbiología , Pyrus/virología , Concentración de Iones de Hidrógeno
14.
Viruses ; 16(4)2024 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-38675985

RESUMEN

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.


Asunto(s)
Bacteriólisis , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virología , Pseudomonas aeruginosa/fisiología , Pseudomonas aeruginosa/genética , Fagos Pseudomonas/fisiología , Fagos Pseudomonas/genética , Bacteriófagos/fisiología , Bacteriófagos/genética , Escherichia coli/virología , Escherichia coli/genética , Especificidad del Huésped , Muramidasa/metabolismo
15.
Sci Rep ; 14(1): 9218, 2024 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649444

RESUMEN

For reducing Campylobacter (C.) in the food production chain and thus the risk to the consumer, the combined application of different measures as a multiple-hurdle approach is currently under discussion. This is the first study to investigate possible synergistic activities in vivo, aiming at reducing intestinal C. jejuni counts by administering (i) bacteriophages (phages) in combination with a competitive exclusion (CE) product and (ii) carvacrol combined with organic acids. The combined application of the two selected phages (Fletchervirus phage NCTC 12673 and Firehammervirus phage vB_CcM-LmqsCPL1/1) and the CE product significantly reduced C. jejuni loads by 1.0 log10 in cecal and colonic contents as well as in cloacal swabs at the end of the trial (33 and 34 days post hatch). The proportion of bacterial isolates showing reduced phage susceptibility ranged from 10.9% (isolates from cecal content) to 47.8% (isolates from cloacal swabs 32 days post hatch) for the Fletchervirus phage, while all tested isolates remained susceptible to the Firehammervirus phage. The use of carvacrol combined with an organic acid blend (sorbic acid, benzoic acid, propionic acid, and acetic acid) significantly reduced Campylobacter counts by 1.0 log10 in cloacal swabs on day 30 only.


Asunto(s)
Bacteriófagos , Pollos , Cimenos , Cimenos/farmacología , Animales , Bacteriófagos/fisiología , Pollos/microbiología , Infecciones por Campylobacter/prevención & control , Infecciones por Campylobacter/microbiología , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Campylobacter jejuni/virología , Campylobacter jejuni/efectos de los fármacos , Campylobacter/efectos de los fármacos , Campylobacter/virología
16.
Sci Rep ; 14(1): 9399, 2024 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658654

RESUMEN

Edwardsiella piscicida causes significant economic losses to the aquaculture industry worldwide. Phage-based biocontrol methods are experiencing a renaissance because of the spread of drug-resistant genes and bacteria resulting from the heavy use of antibiotics. Here, we showed that the novel Edwardsiella phage EPP-1 could achieve comparable efficacy to florfenicol using a zebrafish model of Edwardsiella piscicida infection and could reduce the content of the floR resistance gene in zebrafish excreta. Specifically, phage EPP-1 inhibited bacterial growth in vitro and significantly improved the zebrafish survival rate in vivo (P = 0.0035), achieving an efficacy comparable to that of florfenicol (P = 0.2304). Notably, integrating the results of 16S rRNA sequencing, metagenomic sequencing, and qPCR, although the effects of phage EPP-1 converged with those of florfenicol in terms of the community composition and potential function of the zebrafish gut microbiota, it reduced the floR gene content in zebrafish excreta and aquaculture water. Overall, our study highlights the feasibility and safety of phage therapy for edwardsiellosis control, which has profound implications for the development of antibiotic alternatives to address the antibiotic crisis.


Asunto(s)
Antibacterianos , Bacteriófagos , Edwardsiella , Infecciones por Enterobacteriaceae , Tianfenicol/análogos & derivados , Pez Cebra , Animales , Pez Cebra/microbiología , Edwardsiella/genética , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/veterinaria , Infecciones por Enterobacteriaceae/terapia , Bacteriófagos/genética , Bacteriófagos/fisiología , Antibacterianos/farmacología , Farmacorresistencia Bacteriana/genética , Microbioma Gastrointestinal , Terapia de Fagos/métodos , ARN Ribosómico 16S/genética , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/terapia , Enfermedades de los Peces/prevención & control , Tianfenicol/farmacología , Acuicultura/métodos
17.
Nat Commun ; 15(1): 2993, 2024 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-38582763

RESUMEN

Bacteriophage therapy is a promising approach to address antimicrobial infections though questions remain regarding the impact of the immune response on clinical effectiveness. Here, we develop a mouse model to assess phage treatment using a cocktail of five phages from the Myoviridae and Siphoviridae families that target Vancomycin-Resistant Enterococcus gut colonization. Phage treatment significantly reduces fecal bacterial loads of Vancomycin-Resistant Enterococcus. We also characterize immune responses elicited following administration of the phage cocktail. While minimal innate responses are observed after phage administration, two rounds of treatment induces phage-specific neutralizing antibodies and accelerate phage clearance from tissues. Interestingly, the myophages in our cocktail induce a more robust neutralizing antibody response than the siphophages. This anti-phage immunity reduces the effectiveness of the phage cocktail in our murine model. Collectively, this study shows phage-specific immune responses may be an important consideration in the development of phage cocktails for therapeutic use.


Asunto(s)
Bacteriófagos , Enterococos Resistentes a la Vancomicina , Humanos , Animales , Ratones , Bacteriófagos/fisiología , Vancomicina/farmacología , Modelos Animales de Enfermedad , Myoviridae/fisiología , Antibacterianos/farmacología
18.
Cell Host Microbe ; 32(5): 727-738.e6, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38579715

RESUMEN

Many, if not all, bacteria use quorum sensing (QS) to control collective behaviors, and more recently, QS has also been discovered in bacteriophages (phages). Phages can produce communication molecules of their own, or "listen in" on the host's communication processes, to switch between lytic and lysogenic modes of infection. Here, we study the interaction of Vibrio cholerae with the lysogenic phage VP882, which is activated by the QS molecule DPO. We discover that induction of VP882 results in the binding of phage transcripts to the major RNA chaperone Hfq, which in turn outcompetes and downregulates host-encoded small RNAs (sRNAs). VP882 itself also encodes Hfq-binding sRNAs, and we demonstrate that one of these sRNAs, named VpdS, promotes phage replication by regulating host and phage mRNA levels. We further show that host-encoded sRNAs can antagonize phage replication by downregulating phage mRNA expression and thus might be part of the host's phage defense arsenal.


Asunto(s)
Bacteriófagos , Proteína de Factor 1 del Huésped , Percepción de Quorum , Vibrio cholerae , Vibrio cholerae/virología , Vibrio cholerae/genética , Percepción de Quorum/genética , Bacteriófagos/genética , Bacteriófagos/fisiología , Proteína de Factor 1 del Huésped/metabolismo , Proteína de Factor 1 del Huésped/genética , Replicación Viral , Lisogenia , ARN Viral/genética , ARN Viral/metabolismo , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Regulación Bacteriana de la Expresión Génica , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Mensajero/metabolismo , ARN Mensajero/genética , Interacciones Microbiota-Huesped/genética
19.
Cell Host Microbe ; 32(5): 768-778.e9, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38653241

RESUMEN

Microbiomes feature complex interactions between diverse bacteria and bacteriophages. Synthetic microbiomes offer a powerful way to study these interactions; however, a major challenge is obtaining a representative bacteriophage population during the bacterial isolation process. We demonstrate that colony isolation reliably excludes virulent viruses from sample sources with low virion-to-bacteria ratios such as feces, creating "virulent virus-free" controls. When the virulent dsDNA virome is reintroduced to a 73-strain synthetic gut microbiome in a bioreactor model of the human colon, virulent viruses target susceptible strains without significantly altering community structure or metabolism. In addition, we detected signals of prophage induction that associate with virulent predation. Overall, our findings indicate that dilution-based isolation methods generate synthetic gut microbiomes that are heavily depleted, if not devoid, of virulent viruses and that such viruses, if reintroduced, have a targeted effect on community assembly, metabolism, and prophage replication.


Asunto(s)
Bacterias , Bacteriófagos , Heces , Microbioma Gastrointestinal , Bacteriófagos/genética , Bacteriófagos/fisiología , Humanos , Heces/microbiología , Heces/virología , Bacterias/virología , Bacterias/genética , Profagos/genética , Profagos/fisiología , Viroma , Reactores Biológicos/microbiología , Reactores Biológicos/virología , Colon/microbiología , Colon/virología , Microbiota , Virulencia
20.
PLoS Biol ; 22(4): e3002346, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38648198

RESUMEN

Where there are bacteria, there will be bacteriophages. These viruses are known to be important players in shaping the wider microbial community in which they are embedded, with potential implications for human health. On the other hand, bacteria possess a range of distinct immune mechanisms that provide protection against bacteriophages, including the mutation or complete loss of the phage receptor, and CRISPR-Cas adaptive immunity. While our previous work showed how a microbial community may impact phage resistance evolution, little is known about the inverse, namely how interactions between phages and these different phage resistance mechanisms affect the wider microbial community in which they are embedded. Here, we conducted a 10-day, fully factorial evolution experiment to examine how phage impact the structure and dynamics of an artificial four-species bacterial community that includes either Pseudomonas aeruginosa wild-type or an isogenic mutant unable to evolve phage resistance through CRISPR-Cas. Additionally, we used mathematical modelling to explore the ecological interactions underlying full community behaviour, as well as to identify general principles governing the impacts of phage on community dynamics. Our results show that the microbial community structure is drastically altered by the addition of phage, with Acinetobacter baumannii becoming the dominant species and P. aeruginosa being driven nearly extinct, whereas P. aeruginosa outcompetes the other species in the absence of phage. Moreover, we find that a P. aeruginosa strain with the ability to evolve CRISPR-based resistance generally does better when in the presence of A. baumannii, but that this benefit is largely lost over time as phage is driven extinct. Finally, we show that pairwise data alone is insufficient when modelling our microbial community, both with and without phage, highlighting the importance of higher order interactions in governing multispecies dynamics in complex communities. Combined, our data clearly illustrate how phage targeting a dominant species allows for the competitive release of the strongest competitor while also contributing to community diversity maintenance and potentially preventing the reinvasion of the target species, and underline the importance of mapping community composition before therapeutically applying phage.


Asunto(s)
Bacteriófagos , Sistemas CRISPR-Cas , Microbiota , Pseudomonas aeruginosa , Bacteriófagos/fisiología , Bacteriófagos/genética , Pseudomonas aeruginosa/virología , Acinetobacter baumannii/virología , Mutación , Bacterias/virología , Bacterias/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...