Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 316
Filtrar
1.
Nat Commun ; 15(1): 8572, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39362854

RESUMEN

Spread of multidrug-resistant Pseudomonas aeruginosa strains threatens to render currently available antibiotics obsolete, with limited prospects for the development of new antibiotics. Lytic bacteriophages, the viruses of bacteria, represent a path to combat this threat. In vitro-directed evolution is traditionally applied to expand the bacteriophage host range or increase bacterial suppression in planktonic cultures. However, while up to 80% of human microbial infections are biofilm-associated, research towards targeted improvement of bacteriophages' ability to combat biofilms remains scarce. This study aims at an in vitro biofilm evolution assay to improve multiple bacteriophage parameters in parallel and the optimisation of bacteriophage cocktail design by exploiting a bacterial bacteriophage resistance trade-off. The evolved bacteriophages show an expanded host spectrum, improved antimicrobial efficacy and enhanced antibiofilm performance, as assessed by isothermal microcalorimetry and quantitative polymerase chain reaction, respectively. Our two-phage cocktail reveals further improved antimicrobial efficacy without incurring dual-bacteriophage-resistance in treated bacteria. We anticipate this assay will allow a better understanding of phenotypic-genomic relationships in bacteriophages and enable the training of bacteriophages against other desired pathogens. This, in turn, will strengthen bacteriophage therapy as a treatment adjunct to improve clinical outcomes of multidrug-resistant bacterial infections.


Asunto(s)
Antibacterianos , Bacteriófagos , Biopelículas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virología , Pseudomonas aeruginosa/fisiología , Pseudomonas aeruginosa/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Bacteriófagos/fisiología , Bacteriófagos/genética , Antibacterianos/farmacología , Fagos Pseudomonas/fisiología , Fagos Pseudomonas/genética , Humanos , Terapia de Fagos/métodos , Farmacorresistencia Bacteriana Múltiple , Infecciones por Pseudomonas/terapia , Infecciones por Pseudomonas/microbiología , Especificidad del Huésped , Pruebas de Sensibilidad Microbiana
2.
Nat Commun ; 15(1): 8482, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39353939

RESUMEN

DEV is an obligatory lytic Pseudomonas phage of the N4-like genus, recently reclassified as Schitoviridae. The DEV genome encodes 91 ORFs, including a 3398 amino acid virion-associated RNA polymerase (vRNAP). Here, we describe the complete architecture of DEV, determined using a combination of cryo-electron microscopy localized reconstruction, biochemical methods, and genetic knockouts. We built de novo structures of all capsid factors and tail components involved in host attachment. We demonstrate that DEV long tail fibers are essential for infection of Pseudomonas aeruginosa but dispensable for infecting mutants with a truncated lipopolysaccharide devoid of the O-antigen. We determine that DEV vRNAP is part of a three-gene operon conserved in 191 Schitoviridae genomes. We propose these three proteins are ejected into the host to form a genome ejection motor spanning the cell envelope. We posit that the design principles of the DEV ejection apparatus are conserved in all Schitoviridae.


Asunto(s)
Microscopía por Crioelectrón , Genoma Viral , Fagos Pseudomonas , Pseudomonas aeruginosa , Fagos Pseudomonas/genética , Fagos Pseudomonas/ultraestructura , Genoma Viral/genética , Pseudomonas aeruginosa/virología , Pseudomonas aeruginosa/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , Virión/ultraestructura , Virión/genética , Sistemas de Lectura Abierta/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Virales/química , Operón/genética , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Cápside/metabolismo , Cápside/ultraestructura
3.
Mol Genet Genomics ; 299(1): 97, 2024 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-39395039

RESUMEN

The food industry has incurred substantial losses from contamination by Pseudomonas fluorescens, emphasizing the critical importance of implementing effective control strategies. Phages are potential sterilizers due to their specific killing abilities and the difficulty bacteria face in developing resistance. However, a significant barrier to their development is the lack of diversity among phage types. In this study, we characterized a novel lytic P. fluorescens phage, named vB_PF_Y1-MI. Phage vB_PF_Y1-MI displayed a latent period of nearly 10 min and a high burst size of 1493 PFU/cell. This phage showed good activity over a wide range of temperature (up to 70 °C) and pH (3-12). The genome of phage vB_PF_Y1-MI spans 93,233 bp with a GC content of 45%. It encompasses 174 open-reading frames and 19 tRNA genes, while no lysogeny or virulence-associated genes were detected. Phylogenetic analysis positions it as a novel unassigned evolutionary lineage within the Caudoviricetes class among related dsDNA phages. Our study provides foundational insights into vB_PF_Y1-MI and emphasizes its potential as an effective biological control agent against P. fluorescens. This research offers crucial theoretical groundwork and technical support for subsequent efforts in preventing and controlling P. fluorescens contamination.


Asunto(s)
Genoma Viral , Leche , Filogenia , Pseudomonas fluorescens , Pseudomonas fluorescens/virología , Pseudomonas fluorescens/genética , Leche/microbiología , Leche/virología , Animales , Genoma Viral/genética , Fagos Pseudomonas/genética , Fagos Pseudomonas/aislamiento & purificación , Composición de Base/genética , Contaminación de Alimentos/prevención & control , Microbiología de Alimentos , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Bacteriófagos/clasificación , Sistemas de Lectura Abierta/genética
4.
mSystems ; 9(9): e0080124, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39166874

RESUMEN

The opportunistic human pathogen Pseudomonas aeruginosa is naturally infected by a large class of temperate, transposable, Mu-like phages. We examined the genotypic and phenotypic diversity of P. aeruginosa PA14 lysogen populations as they resolve clustered regularly interspaced short palindromic repeat (CRISPR) autoimmunity, mediated by an imperfect CRISPR match to the Mu-like DMS3 prophage. After 12 days of evolution, we measured a decrease in spontaneous induction in both exponential and stationary phase growth. Co-existing variation in spontaneous induction rates in the exponential phase depended on the way the coexisting strains resolved genetic conflict. Multiple mutational modes to resolve genetic conflict between host and phage resulted in coexistence in evolved populations of single lysogens that maintained CRISPR immunity to other phages and polylysogens that lost immunity completely. This work highlights a new dimension of the role of lysogenic phages in the evolution of their hosts.IMPORTANCEThe chronic opportunistic multi-drug-resistant pathogen Pseudomonas aeruginosa is persistently infected by temperate phages. We assess the contribution of temperate phage infection to the evolution of the clinically relevant strain UCBPP-PA14. We found that a low level of clustered regularly interspaced short palindromic repeat (CRISPR)-mediated self-targeting resulted in polylysogeny evolution and large genome rearrangements in lysogens; we also found extensive diversification in CRISPR spacers and cas genes. These genomic modifications resulted in decreased spontaneous induction in both exponential and stationary phase growth, increasing lysogen fitness. This work shows the importance of considering latent phage infection in characterizing the evolution of bacterial populations.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Lisogenia , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virología , Pseudomonas aeruginosa/genética , Lisogenia/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Fagos Pseudomonas/genética , Evolución Molecular , Bacteriófagos/genética , Profagos/genética
5.
EMBO J ; 43(19): 4384-4405, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39143239

RESUMEN

Bacteriophages are the most abundant biological entities on Earth, but our understanding of many aspects of their lifecycles is still incomplete. Here, we have structurally analysed the infection cycle of the siphophage Casadabanvirus JBD30. Using its baseplate, JBD30 attaches to Pseudomonas aeruginosa via the bacterial type IV pilus, whose subsequent retraction brings the phage to the bacterial cell surface. Cryo-electron microscopy structures of the baseplate-pilus complex show that the tripod of baseplate receptor-binding proteins attaches to the outer bacterial membrane. The tripod and baseplate then open to release three copies of the tape-measure protein, an event that is followed by DNA ejection. JBD30 major capsid proteins assemble into procapsids, which expand by 7% in diameter upon filling with phage dsDNA. The DNA-filled heads are finally joined with 180-nm-long tails, which bend easily because flexible loops mediate contacts between the successive discs of major tail proteins. It is likely that the structural features and replication mechanisms described here are conserved among siphophages that utilize the type IV pili for initial cell attachment.


Asunto(s)
Microscopía por Crioelectrón , Fagos Pseudomonas , Pseudomonas aeruginosa , Replicación Viral , Pseudomonas aeruginosa/virología , Pseudomonas aeruginosa/metabolismo , Fagos Pseudomonas/ultraestructura , Fagos Pseudomonas/genética , Fagos Pseudomonas/metabolismo , Fagos Pseudomonas/fisiología , Fimbrias Bacterianas/metabolismo , Fimbrias Bacterianas/ultraestructura , Fimbrias Bacterianas/virología , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , ADN Viral/metabolismo , ADN Viral/genética , Siphoviridae/genética , Siphoviridae/ultraestructura , Siphoviridae/fisiología , Siphoviridae/metabolismo
6.
Nat Commun ; 15(1): 7244, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39174532

RESUMEN

The filamentous 'Pf' bacteriophages of Pseudomonas aeruginosa play roles in biofilm formation and virulence, but mechanisms governing Pf prophage activation in biofilms are unclear. Here, we identify a prophage regulatory module, KKP (kinase-kinase-phosphatase), that controls virion production of co-resident Pf prophages and mediates host defense against diverse lytic phages. KKP consists of Ser/Thr kinases PfkA and PfkB, and phosphatase PfpC. The kinases have multiple host targets, one of which is MvaU, a host nucleoid-binding protein and known prophage-silencing factor. Characterization of KKP deletion and overexpression strains with transcriptional, protein-level and prophage-based approaches indicates that shifts in the balance between kinase and phosphatase activities regulate phage production by controlling MvaU phosphorylation. In addition, KKP acts as a tripartite toxin-antitoxin system that provides defense against some lytic phages. A conserved lytic phage replication protein inhibits the KKP phosphatase PfpC, stimulating toxic kinase activity and blocking lytic phage production. Thus, KKP represents a phosphorylation-based mechanism for prophage regulation and antiphage defense. The conservation of KKP gene clusters in >1000 diverse temperate prophages suggests that integrated control of temperate and lytic phage infection by KKP-like regulatory modules may play a widespread role in shaping host cell physiology.


Asunto(s)
Lisogenia , Profagos , Pseudomonas aeruginosa , Lisogenia/genética , Pseudomonas aeruginosa/virología , Pseudomonas aeruginosa/genética , Profagos/genética , Profagos/fisiología , Fosforilación , Monoéster Fosfórico Hidrolasas/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Proteínas Virales/metabolismo , Proteínas Virales/genética , Fagos Pseudomonas/genética , Fagos Pseudomonas/metabolismo , Biopelículas/crecimiento & desarrollo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Regulación Viral de la Expresión Génica
7.
Science ; 385(6704): 105-112, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38963841

RESUMEN

Introns containing homing endonucleases are widespread in nature and have long been assumed to be selfish elements that provide no benefit to the host organism. These genetic elements are common in viruses, but whether they confer a selective advantage is unclear. In this work, we studied intron-encoded homing endonuclease gp210 in bacteriophage ΦPA3 and found that it contributes to viral competition by interfering with the replication of a coinfecting phage, ΦKZ. We show that gp210 targets a specific sequence in ΦKZ, which prevents the assembly of progeny viruses. This work demonstrates how a homing endonuclease can be deployed in interference competition among viruses and provide a relative fitness advantage. Given the ubiquity of homing endonucleases, this selective advantage likely has widespread evolutionary implications in diverse plasmid and viral competition as well as virus-host interactions.


Asunto(s)
Endonucleasas , Intrones , Fagos Pseudomonas , Pseudomonas aeruginosa , Interferencia Viral , Proteínas Virales , Endonucleasas/metabolismo , Endonucleasas/genética , Interferencia Viral/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Ensamble de Virus , Replicación Viral , Fagos Pseudomonas/enzimología , Fagos Pseudomonas/genética , Pseudomonas aeruginosa/virología
8.
J Mol Biol ; 436(18): 168713, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39029888

RESUMEN

Bacteriophage ΦKZ (PhiKZ) is the founding member of a family of giant bacterial viruses. It has potential as a therapeutic as its host, Pseudomonas aeruginosa, kills tens of thousands of people worldwide each year. ΦKZ infection is independent of the host transcriptional apparatus; the virus forms a "nucleus", producing a proteinaceous barrier around the ΦKZ genome that excludes the host immune systems. It expresses its own non-canonical multi-subunit non-virion RNA polymerase (nvRNAP), which is imported into its "nucleus" to transcribe viral genes. The ΦKZ nvRNAP is formed by four polypeptides representing homologues of the eubacterial ß/ß' subunits, and a fifth that is likely to have evolved from an ancestral homologue to σ-factor. We have resolved the structure of the ΦKZ nvRNAP initiating transcription from its cognate promoter, p119L, including previously disordered regions. Our results shed light on the similarities and differences between ΦKZ nvRNAP mechanisms of transcription and those of canonical eubacterial RNAPs and the related non-canonical nvRNAP of bacteriophage AR9.


Asunto(s)
ARN Polimerasas Dirigidas por ADN , Regiones Promotoras Genéticas , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/química , Modelos Moleculares , Bacteriófagos/genética , Bacteriófagos/enzimología , Transcripción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Virales/química , Fagos Pseudomonas/genética , Fagos Pseudomonas/enzimología , Conformación Proteica , Pseudomonas aeruginosa/virología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/enzimología , Cristalografía por Rayos X
9.
Viruses ; 16(7)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-39066214

RESUMEN

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.


Asunto(s)
Fibrosis Quística , Terapia de Fagos , Infecciones por Pseudomonas , Fagos Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virología , Humanos , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/terapia , Fibrosis Quística/microbiología , Fagos Pseudomonas/genética , Fagos Pseudomonas/fisiología , Animales , Bacteriófagos/fisiología , Bacteriófagos/genética , Antibacterianos/farmacología , Antibacterianos/uso terapéutico
10.
Microbiol Spectr ; 12(8): e0387523, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38949386

RESUMEN

Bacteriophages (hereafter "phages") are ubiquitous predators of bacteria in the natural world, but interest is growing in their development into antibacterial therapy as complement or replacement for antibiotics. However, bacteria have evolved a huge variety of antiphage defense systems allowing them to resist phage lysis to a greater or lesser extent. In addition to dedicated phage defense systems, some aspects of the general stress response also impact phage susceptibility, but the details of this are not well known. In order to elucidate these factors in the opportunistic pathogen Pseudomonas aeruginosa, we used the laboratory-conditioned strain PAO1 as host for phage infection experiments as it is naturally poor in dedicated phage defense systems. Screening by transposon insertion sequencing indicated that the uncharacterized operon PA3040-PA3042 was potentially associated with resistance to lytic phages. However, we found that its primary role appeared to be in regulating biofilm formation, particularly in a clinical isolate of P. aeruginosa in which it also altered tobramycin resistance. Its expression was highly growth-phase dependent and responsive to phage infection and cell envelope stress. Our results suggest that this operon may be a cryptic but important locus for P. aeruginosa stress tolerance. IMPORTANCE: An important category of bacterial stress response systems is bacteriophage defense, where systems are triggered by bacteriophage infection and activate a response which may either destroy the phage genome or destroy the infected cell so that the rest of the population survives. In some bacteria, the cell envelope stress response is activated by bacteriophage infection, but it is unknown whether this contributes to the survival of the infection. We have found that a conserved uncharacterized operon (PA3040-PA3042) of the cell envelope stress regulon in Pseudomonas aeruginosa, which has very few dedicated phage defense systems, responds to phage infection and stationary phase as well as envelope stress and is important for growth and biofilm formation in a clinical isolate of P. aeruginosa, even in the absence of phages. As homologs of these genes are found in other bacteria, they may be a novel component of the general stress response.


Asunto(s)
Antibacterianos , Biopelículas , Farmacorresistencia Bacteriana , Operón , Pseudomonas aeruginosa , Tobramicina , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/virología , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Antibacterianos/farmacología , Tobramicina/farmacología , Farmacorresistencia Bacteriana/genética , Humanos , Infecciones por Pseudomonas/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bacteriófagos/genética , Bacteriófagos/fisiología , Regulación Bacteriana de la Expresión Génica , Estrés Fisiológico , Pared Celular/metabolismo , Pared Celular/efectos de los fármacos , Pared Celular/genética , Fagos Pseudomonas/genética , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos
11.
Microb Biotechnol ; 17(6): e14489, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38864499

RESUMEN

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.


Asunto(s)
Enfermedades de las Plantas , Pseudomonas syringae , Pseudomonas syringae/virología , Pseudomonas syringae/genética , Enfermedades de las Plantas/microbiología , Fagos Pseudomonas/genética , Fagos Pseudomonas/fisiología , Bacteriófagos/genética , Bacteriófagos/fisiología
12.
J Gen Virol ; 105(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38833289

RESUMEN

Relatively few phages that infect plant pathogens have been isolated and investigated. The Pseudomonas syringae species complex is present in various environments, including plants. It can cause major crop diseases, such as bacterial canker on apricot trees. This study presents a collection of 25 unique phage genomes that infect P. syringae. These phages were isolated from apricot orchards with bacterial canker symptoms after enrichment with 21 strains of P. syringae. This collection comprises mostly virulent phages, with only three being temperate. They belong to 14 genera, 11 of which are newly discovered, and 18 new species, revealing great genetic diversity within this collection. Novel DNA packaging systems have been identified bioinformatically in one of the new phage species, but experimental confirmation is required to define the precise mechanism. Additionally, many phage genomes contain numerous potential auxiliary metabolic genes with diversified putative functions. At least three phages encode genes involved in bacterial tellurite resistance, a toxic metalloid. This suggests that viruses could play a role in bacterial stress tolerance. This research emphasizes the significance of continuing the search for new phages in the agricultural ecosystem to unravel novel ecological diversity and new gene functions. This work contributes to the foundation for future fundamental and applied research on phages infecting phytopathogenic bacteria.


Asunto(s)
Genoma Viral , Enfermedades de las Plantas , Fagos Pseudomonas , Pseudomonas syringae , Pseudomonas syringae/virología , Pseudomonas syringae/genética , Enfermedades de las Plantas/microbiología , Enfermedades de las Plantas/virología , Fagos Pseudomonas/genética , Filogenia , Variación Genética
13.
Science ; 384(6701): eado0713, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38870284

RESUMEN

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.


Asunto(s)
Bacteriocinas , Fagos Pseudomonas , Pseudomonas , Proteínas de la Cola de los Virus , Antibiosis , Membrana Externa Bacteriana/metabolismo , Bacteriocinas/genética , Bacteriocinas/metabolismo , Variación Genética , Genoma Bacteriano , Polisacáridos Bacterianos/metabolismo , Pseudomonas/metabolismo , Pseudomonas/virología , Fagos Pseudomonas/genética , Fagos Pseudomonas/metabolismo , Proteínas de la Cola de los Virus/metabolismo , Proteínas de la Cola de los Virus/genética , Terapia de Fagos/métodos
14.
BMC Microbiol ; 24(1): 207, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858621

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas , Pseudomonas aeruginosa , Percepción de Quorum , Transactivadores , Pseudomonas aeruginosa/virología , Pseudomonas aeruginosa/genética , Percepción de Quorum/genética , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Fagos Pseudomonas/genética , Fagos Pseudomonas/fisiología , Aguas del Alcantarillado/virología , Aguas del Alcantarillado/microbiología , Regulación Bacteriana de la Expresión Génica , Lipopolisacáridos/metabolismo , Técnicas de Inactivación de Genes
15.
Cell Host Microbe ; 32(7): 1050-1058.e7, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38870941

RESUMEN

Viral genomes are most vulnerable to cellular defenses at the start of the infection. A family of jumbo phages related to phage ΦKZ, which infects Pseudomonas aeruginosa, assembles a protein-based phage nucleus to protect replicating phage DNA, but how it is protected prior to phage nucleus assembly is unclear. We find that host proteins related to membrane and lipid biology interact with injected phage protein, clustering in an early phage infection (EPI) vesicle. The injected virion RNA polymerase (vRNAP) executes early gene expression until phage genome separation from the vRNAP and the EPI vesicle, moving into the nascent proteinaceous phage nucleus. Enzymes involved in DNA replication and CRISPR/restriction immune nucleases are excluded by the EPI vesicle. We propose that the EPI vesicle is rapidly constructed with injected phage proteins, phage DNA, host lipids, and host membrane proteins to enable genome protection, early transcription, localized translation, and to ensure faithful genome transfer to the proteinaceous nucleus.


Asunto(s)
ADN Viral , Genoma Viral , Fagos Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virología , Fagos Pseudomonas/genética , Fagos Pseudomonas/metabolismo , ADN Viral/genética , Proteínas Virales/metabolismo , Proteínas Virales/genética , Bacteriófagos/genética , Bacteriófagos/fisiología , Virión/metabolismo , Replicación Viral , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , Lípidos , Replicación del ADN
16.
mSphere ; 9(7): e0070723, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38934592

RESUMEN

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.


Asunto(s)
Terapia de Fagos , Fagos Pseudomonas , Pseudomonas , Terapia de Fagos/métodos , Fagos Pseudomonas/genética , Fagos Pseudomonas/fisiología , Pseudomonas/virología , Infecciones por Pseudomonas/terapia , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/virología , Mutación , Antibacterianos/farmacología , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/clasificación , Farmacorresistencia Bacteriana , Farmacorresistencia Bacteriana Múltiple
17.
Microbiol Spectr ; 12(8): e0352023, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38912817

RESUMEN

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.


Asunto(s)
Mucinas , Moco , Infecciones por Pseudomonas , Fagos Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virología , Pseudomonas aeruginosa/fisiología , Moco/microbiología , Moco/virología , Fagos Pseudomonas/fisiología , Fagos Pseudomonas/genética , Mucinas/metabolismo , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/terapia , Humanos , Terapia de Fagos , Animales , Acoplamiento Viral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas
18.
Nat Microbiol ; 9(7): 1828-1841, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38886583

RESUMEN

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.


Asunto(s)
Terapia de Fagos , Infecciones por Pseudomonas , Fagos Pseudomonas , Pseudomonas aeruginosa , Proteínas Virales , Pseudomonas aeruginosa/virología , Pseudomonas aeruginosa/patogenicidad , Pseudomonas aeruginosa/genética , Animales , Ratones , Fagos Pseudomonas/genética , Fagos Pseudomonas/fisiología , Infecciones por Pseudomonas/terapia , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/inmunología , Virulencia , Proteínas Virales/genética , Proteínas Virales/metabolismo , Biopelículas/crecimiento & desarrollo , GMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , Femenino , Bacteriófagos/fisiología , Bacteriófagos/genética
19.
Environ Microbiol ; 26(6): e16671, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38863081

RESUMEN

The environmental bacterium, Pseudomonas putida, possesses a broad spectrum of metabolic pathways. This makes it highly promising for use in biotechnological production as a cell factory, as well as in bioremediation strategies to degrade various aromatic pollutants. For P. putida to flourish in its environment, it must withstand the continuous threats posed by bacteriophages. Interestingly, until now, only a handful of phages have been isolated for the commonly used laboratory strain, P. putida KT2440, and no phage defence mechanisms have been characterized. In this study, we present a new Collection of Environmental P. putida Phages from Estonia, or CEPEST. This collection comprises 67 double-stranded DNA phages, which belong to 22 phage species and 9 phage genera. Our findings reveal that most phages in the CEPEST collection are more infectious at lower temperatures, have a narrow host range, and require an intact lipopolysaccharide for P. putida infection. Furthermore, we show that cryptic prophages present in the P. putida chromosome provide strong protection against the infection of many phages. However, the chromosomal toxin-antitoxin systems do not play a role in the phage defence of P. putida. This research provides valuable insights into the interactions between P. putida and bacteriophages, which could have significant implications for biotechnological and environmental applications.


Asunto(s)
Especificidad del Huésped , Pseudomonas putida , Pseudomonas putida/virología , Pseudomonas putida/genética , Pseudomonas putida/metabolismo , Profagos/genética , Fagos Pseudomonas/genética , Fagos Pseudomonas/aislamiento & purificación , Estonia , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación
20.
J Bacteriol ; 206(5): e0040223, 2024 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-38687034

RESUMEN

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.


Asunto(s)
Profagos , Pseudomonas aeruginosa , Percepción de Quorum , Biopelículas/crecimiento & desarrollo , Caenorhabditis elegans/microbiología , Caenorhabditis elegans/virología , Lisogenia , Profagos/genética , Profagos/fisiología , Pseudomonas aeruginosa/virología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Pseudomonas aeruginosa/fisiología , Infecciones por Pseudomonas/microbiología , Fagos Pseudomonas/genética , Fagos Pseudomonas/fisiología , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA