Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.019
Filtrar
1.
Mol Cell ; 84(17): 3172-3174, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39241751

RESUMEN

Deploying anti-CRISPR proteins is a potent strategy used by phages to inhibit bacterial CRISPR-Cas defense. In a new Nature paper, Trost et al.1 discover and characterize an exciting anti-CRISPR mechanism with possible implications beyond this microscopic arms race.


Asunto(s)
Bacterias , Bacteriófagos , Sistemas CRISPR-Cas , Bacteriófagos/genética , Bacterias/genética , Bacterias/inmunología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
2.
Front Cell Infect Microbiol ; 14: 1442062, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39224703

RESUMEN

Background: Klebsiella pneumoniae is a major cause of hospital-acquired infections (HAIs), primarily spread through environmental contamination in hospitals. The effectiveness of current chemical disinfectants is waning due to emerging resistance, which poses environmental hazards and fosters new resistance in pathogens. Developing environmentally friendly and effective disinfectants against multidrug-resistant organisms is increasingly important. Methods: This study developed a bacteriophage cocktail targeting two common carbapenem-resistant Klebsiella pneumoniae (CRKP) strains, ST11 KL47 and ST11 KL64. The cocktail was used as an adjunctive disinfectant in a hospital's respiratory intensive care unit (RICU) via ultrasonic nebulization. Digital PCR was used to quantify CRKP levels post-intervention. The microbial community composition was analyzed via 16S rRNA sequencing to assess the intervention's impact on overall diversity. Results: The phage cocktail significantly reduced CRKP levels within the first 24 hours post-treatment. While a slight increase in pathogen levels was observed after 24 hours, they remained significantly lower than those treated with conventional disinfectants. 16S rRNA sequencing showed a decrease in the target pathogens' relative abundance, while overall species diversity remained stable, confirming that phages selectively target CRKP without disrupting ecological balance. Discussion: The findings highlight the efficacy and safety of phage-based biocleaners as a sustainable alternative to conventional disinfectants. Phages selectively reduce multidrug-resistant pathogens while preserving microbial diversity, making them a promising tool for infection control.


Asunto(s)
Bacteriófagos , Descontaminación , Unidades de Cuidados Intensivos , Klebsiella pneumoniae , ARN Ribosómico 16S , ARN Ribosómico 16S/genética , Klebsiella pneumoniae/virología , Klebsiella pneumoniae/genética , Descontaminación/métodos , Bacteriófagos/genética , Humanos , Reacción en Cadena de la Polimerasa/métodos , Infección Hospitalaria/prevención & control , Infección Hospitalaria/microbiología , Desinfectantes/farmacología , Infecciones por Klebsiella/prevención & control , Infecciones por Klebsiella/microbiología , Análisis de Secuencia de ADN
3.
Front Cell Infect Microbiol ; 14: 1421724, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39268483

RESUMEN

The increase of antibiotic-resistant bacteria has become a global health emergency and the need to explore alternative therapeutic options arises. Phage therapy uses bacteriophages to target specific bacterial strains. Phages are highly specific and can target resistant bacteria. Currently, research in this regard is focused on ensuring reliability and safety to bring this tool into clinical practice. The first step is to conduct comprehensive preclinical research. In this work, we present two novel bacteriophages vB_Kpn_F13 and vB_Kpn_F14 isolated against clinical carbapenem-resistant Klebsiella pneumoniae strains obtained from hospital sewage. Multiple studies in vitro were conducted, such as sequencing, electron microscopy, stability, host range infectivity, planktonic effect and biofilm inhibition in order to discover their ability to be used against carbapenem-resistant K. pneumoniae pathogens causing difficult-to-treat infections.


Asunto(s)
Bacteriófagos , Biopelículas , Enterobacteriaceae Resistentes a los Carbapenémicos , Carbapenémicos , Infecciones por Klebsiella , Klebsiella pneumoniae , Terapia de Fagos , Klebsiella pneumoniae/virología , Klebsiella pneumoniae/efectos de los fármacos , Bacteriófagos/aislamiento & purificación , Bacteriófagos/fisiología , Bacteriófagos/genética , Enterobacteriaceae Resistentes a los Carbapenémicos/aislamiento & purificación , Enterobacteriaceae Resistentes a los Carbapenémicos/virología , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/terapia , Carbapenémicos/farmacología , Biopelículas/crecimiento & desarrollo , Biopelículas/efectos de los fármacos , Humanos , Especificidad del Huésped , Aguas del Alcantarillado/virología , Aguas del Alcantarillado/microbiología , Antibacterianos/farmacología , Genoma Viral , Pruebas de Sensibilidad Microbiana
4.
Front Cell Infect Microbiol ; 14: 1397675, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39268487

RESUMEN

Background: The infant gut microbiome's establishment is pivotal for health and immune development. Understanding it unveils insights into growth, development, and maternal microbial interactions. Research often emphasizes gut bacteria, neglecting the phageome. Methods: To investigate the influence of geographic or maternal factors (mode of delivery, mode of breastfeeding, gestational diabetes mellitus) on the gut microbiota and phages of newborns, we collected fecal samples from 34 pairs of mothers and their infants within 24 hours of delivery from three regions (9 pairs from Enshi, 7 pairs from Hohhot, and 18 pairs from Hulunbuir) using sterile containers. Gut microbiota analysis by Shotgun sequencing was subsequently performed. Results: Our results showed that geographic location affects maternal gut microbiology (P < 0.05), while the effect on infant gut microbiology was not significant (P = 0.184). Among the maternal factors, mode of delivery had a significant (P < 0.05) effect on the newborn. Specific bacteria (e.g., Bacteroides, Escherichia spp., Phocaeicola vulgatus, Escherichia coli, Staphylococcus hominis, Veillonella spp.), predicted active metabolites, and bacteriophage vOTUs varied with delivery mode. Phocaeicola vulgatus significantly correlated with some metabolites and bacteriophages in the early infant gut (P < 0.05). In the GD group, a strong negative correlation of phage diversity between mother and infants was observed (R = -0.58, P=0.04). Conclusion: In conclusion, neonatal early gut microbiome (including bacteria and bacteriophages) colonization is profoundly affected by the mode of delivery, and maternal gestational diabetes mellitus. The key bacteria may interact with bacteriophages to influence the levels of specific metabolites. Our study provides new evidence for the study of the infant microbiome, fills a gap in the analysis of the infant gut microbiota regarding the virome, and emphasizes the importance of maternal health for the infant initial gut virome.


Asunto(s)
Bacterias , Diabetes Gestacional , Heces , Microbioma Gastrointestinal , Humanos , Diabetes Gestacional/microbiología , Embarazo , Femenino , Recién Nacido , Heces/microbiología , Heces/virología , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Adulto , Bacteriófagos/genética , Parto Obstétrico , Lactancia Materna
5.
F1000Res ; 13: 380, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39233781

RESUMEN

Bacteria of the genus Aeromonas, especially A. hydrophila and A. veronii are recognized as important fish pathogens that cause significant economic losses in aquaculture. Environmentally friendly bacteriophage-based solutions for the treatment of fish and for the reduction of colonization by pathogenic bacteria in production facilities are currently in high demand. The bacteriophage Gekk3-15 was isolated during a search for novel phage strains potentially suitable for Aeromonas biocontrol applications. Genome sequencing revealed that this virus is a relatively small myovirus with a 64847 bp long dsDNA genome, which is consistent with virion electron microscopy data. Bacteriophage Gekk3-15 is distinct in its nucleotide and encoded aa sequences from all other sequenced bacteriophage genomes, and may represent a new viral taxon at the genus or subfamily level.


Asunto(s)
Aeromonas , Bacteriófagos , Genoma Viral , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Aeromonas/virología , Aeromonas/genética , Secuenciación Completa del Genoma/métodos
6.
PLoS Genet ; 20(9): e1011384, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39231196

RESUMEN

Lytic bacteriophages hold substantial promise in medical and biotechnological applications. Therefore a comprehensive understanding of phage infection mechanisms is crucial. CRISPR-Cas systems offer a way to explore these mechanisms via site-specific phage mutagenesis. However, phages can resist Cas-mediated cleavage through extensive DNA modifications like cytosine glycosylation, hindering mutagenesis efficiency. Our study utilizes the eukaryotic enzyme NgTET to temporarily reduce phage DNA modifications, facilitating Cas nuclease cleavage and enhancing mutagenesis efficiency. This approach enables precise DNA targeting and seamless point mutation integration, exemplified by deactivating specific ADP-ribosyltransferases crucial for phage infection. Furthermore, by temporally removing DNA modifications, we elucidated the effects of these modifications on T4 phage infections without necessitating gene deletions. Our results present a strategy enabling the investigation of phage epigenome functions and streamlining the engineering of phages with cytosine DNA modifications. The described temporal modulation of the phage epigenome is valuable for synthetic biology and fundamental research to comprehend phage infection mechanisms through the generation of mutants.


Asunto(s)
Bacteriófagos , Sistemas CRISPR-Cas , ADN Viral , Epigenoma , ADN Viral/genética , Bacteriófagos/genética , Ingeniería Genética/métodos , Bacteriófago T4/genética , Mutagénesis Sitio-Dirigida/métodos , Escherichia coli/genética , Escherichia coli/virología , Genoma Viral
7.
Int J Mol Sci ; 25(17)2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39273250

RESUMEN

Tuberculosis (TB), caused by Mycobacterium tuberculosis, is an infectious disease that seriously affects human life and health. Despite centuries of efforts to control it, in recent years, the emergence of multidrug-resistant bacterial pathogens of M. tuberculosis due to various factors has exacerbated the disease, posing a serious threat to global health. Therefore, a new method to control M. tuberculosis is urgently needed. Phages, viruses that specifically infect bacteria, have emerged as potential biocontrol agents for bacterial pathogens due to their host specificity. In this study, a mycobacterium phage, Henu3, was isolated from soil around a hospital. The particle morphology, biological characteristics, genomics and phylogeny of Henu3 were characterized. Additionally, to explore the balance between phage resistance and stress response, phage Henu3-resistant strains 0G10 and 2E1 were screened by sequence passage and bidirectional validation methods, which significantly improved the sensitivity of phage to antibiotics (cefotaxime and kanamycin). By whole-genome re-sequencing of strains 0G10 and 2E1, 12 genes involved in cell-wall synthesis, transporter-encoded genes, two-component regulatory proteins and transcriptional regulatory factor-encoded genes were found to have mutations. These results suggest that phage Henu3 has the potential to control M. tuberculosis pathogens, and phage Henu3 has the potential to be a new potential solution for the treatment of M. tuberculosis infection.


Asunto(s)
Mycobacterium tuberculosis , Mycobacterium tuberculosis/virología , Mycobacterium tuberculosis/genética , Filogenia , Genoma Viral , Bacteriófagos/genética , Bacteriófagos/fisiología , Humanos , Micobacteriófagos/genética , Micobacteriófagos/fisiología , Secuenciación Completa del Genoma , Aptitud Genética
8.
Int J Mol Sci ; 25(17)2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39273543

RESUMEN

The rise of carbapenem-resistant Klebsiella pneumoniae (CRKP) presents a significant global challenge in clinical and healthcare settings, severely limiting treatment options. This study aimed to utilize a bacteriophage as an alternative therapy against carbapenem-resistant K. pneumoniae. A novel lytic N4-like Klebsiella phage, vB_kpnP_KPYAP-1 (KPYAP-1), was isolated from sewage. It demonstrated efficacy against the K62 serotype polysaccharide capsule of blaOXA-48-producing K. pneumoniae. KPYAP-1 forms small, clear plaques, has a latent period of 20 min, and reaches a growth plateau at 35 min, with a burst size of 473 plaque-forming units (PFUs) per infected cell. Phylogenetic analysis places KPYAP-1 in the Schitoviridae family, Enquatrovirinae subfamily, and Kaypoctavirus genus. KPYAP-1 employs an N4-like direct terminal repeat mechanism for genome packaging and encodes a large virion-encapsulated RNA polymerase. It lacks integrase or repressor genes, antibiotic resistance genes, bacterial virulence factors, and toxins, ensuring its safety for therapeutic use. Comparative genome analysis revealed that the KPYAP-1 genome is most similar to the KP8 genome, yet differs in tail fiber protein, indicating variations in host recognition. In a zebrafish infection model, KPYAP-1 significantly improved the survival rate of infected fish by 92% at a multiplicity of infection (MOI) of 10, demonstrating its potential for in vivo treatment. These results highlight KPYAP-1 as a promising candidate for developing phage-based therapies targeting carbapenemase-producing K. pneumoniae.


Asunto(s)
Bacteriófagos , Infecciones por Klebsiella , Klebsiella pneumoniae , Pez Cebra , Klebsiella pneumoniae/virología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Animales , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/aislamiento & purificación , Infecciones por Klebsiella/terapia , Infecciones por Klebsiella/microbiología , Filogenia , Genoma Viral , Carbapenémicos/farmacología , Enterobacteriaceae Resistentes a los Carbapenémicos/genética , Enterobacteriaceae Resistentes a los Carbapenémicos/efectos de los fármacos , Antibacterianos/farmacología , Terapia de Fagos
9.
BMC Microbiol ; 24(1): 338, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261757

RESUMEN

Currently, phage biocontrol is increasingly used as a green and natural technology for treating Salmonella and other infections, but phages exhibit instability and activity loss during storage. Therefore, in this study, the effects of lyophilization on the activity and stability of phage cocktails for the control of multidrug-resistant Salmonella in broiler chickens were determined. Eight serotypes of Salmonella were isolated and identified from broiler chicken farms, and bacteriophages against multidrug-resistant Salmonella enterica subsp. enterica serovar Kentucky, Salmonella enterica subsp. enterica serovar Typhimrium and Salmonella enterica subsp. enterica serovar Enteritidis were isolated. The bacteriophage cocktail was prepared and lyophilized, and it was subjected to in vitro and in vivo examinations. A reconstituted lyophilized bacteriophage cocktail was used for the oral treatment of chicks before and after challenge with multidrug-resistant S. Kentucky. The colonization of cecum by S. Kentucky was detected by using real-time PCR, and the serum levels of IgM, IgA and IL-4 and pathological changes in the different groups were detected. Three Caudovirales phages families were identified including Autographiviridae, Straboviridae and Drexlerviridae against multidrug-resistant S. Kentucky, S. Typhimrium and S. Enteritidis. The groups treated with the bacteriophage cocktail showed no clinical signs, no postmortem lesions, and a mortality rate of 0%, which improved the growth performance parameters. Additionally, the estimated serum levels of IgM, IgA and IL-4 were significantly greater in the bacteriophage cocktail-treated groups. Lyophilization effectively preserves the long-term storage stability of phages. Therefore, lyophilized bacteriophage cocktail therapy is a valuable approach for controlling multidrug-resistant Salmonella infections in broiler chickens.


Asunto(s)
Pollos , Farmacorresistencia Bacteriana Múltiple , Liofilización , Enfermedades de las Aves de Corral , Salmonelosis Animal , Fagos de Salmonella , Salmonella , Animales , Pollos/microbiología , Liofilización/métodos , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/terapia , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/prevención & control , Salmonelosis Animal/microbiología , Salmonelosis Animal/terapia , Salmonella/virología , Fagos de Salmonella/fisiología , Ciego/microbiología , Ciego/virología , Terapia de Fagos/métodos , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/aislamiento & purificación
10.
Biosens Bioelectron ; 266: 116727, 2024 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-39232433

RESUMEN

The isolation and identification of pathogenic bacteria from a variety of samples are critical for controlling bacterial infection-related health problems. The conventional methods, such as plate counting and polymerase chain reaction-based approaches, tend to be time-consuming and reliant on specific instruments, severely limiting the effective identification of these pathogens. In this study, we employed the specificity of the cell wall-binding (CBD) domain of the Staphylococcus aureus bacteriophage 80 alpha (80α) endolysin towards the host bacteria for isolation. Amidase 3-CBD conjugated magnetic beads successfully isolated as few as 1 × 102 CFU/mL of S. aureus cells from milk, blood, and saliva. The cell wall hydrolyzing activity of 80α endolysin promoted the genomic DNA extraction efficiency by 12.7 folds on average, compared to the commercial bacterial genomic DNA extraction kit. Then, recombinase polymerase amplification (RPA) was exploited to amplify the nuc gene of S. aureus from the extracted DNA at 37 °C for 30 min. The RPA product activated Cas12a endonuclease activity to cleave fluorescently labeled ssDNA probes. We then converted the generated signal into a fluorescent readout, detectable by either the naked eye or a portable, self-assembled instrument with ultrasensitivity. The entire procedure, from isolation to identification, can be completed within 2 h. The simplicity and sensitivity of the method developed in this study make it of great application value in S. aureus detection, especially in areas with limited resource supply.


Asunto(s)
Técnicas Biosensibles , Endopeptidasas , Staphylococcus aureus , Staphylococcus aureus/aislamiento & purificación , Staphylococcus aureus/virología , Técnicas Biosensibles/métodos , Endopeptidasas/química , Endopeptidasas/aislamiento & purificación , Endopeptidasas/genética , Bacteriófagos/química , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Humanos , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/química , Fagos de Staphylococcus/aislamiento & purificación , Animales , Técnicas de Amplificación de Ácido Nucleico/métodos , Infecciones Estafilocócicas/microbiología , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , Nucleasa Microcócica/química , Nucleasa Microcócica/metabolismo , Nucleasa Microcócica/genética , Proteínas Virales/química , Proteínas Virales/metabolismo
11.
Arch Virol ; 169(10): 196, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39256248

RESUMEN

Vibrio parahaemolyticus is a major seafood-borne zoonotic pathogen that causes gastroenteritis in humans and acute hepatopancreatic necrosis disease (AHPND) in shrimp. In this study, we isolated and characterized Vibrio phage vB_VpM-pA2SJ1, which infects clinical and AHPND-associated strains of V. parahaemolyticus. The phage genome is a linear dsDNA 51,054 bp in length with a G + C content of 43.7%, and it contains 89 open reading frames. Genome comparisons revealed basal similarity to other Vibrio phages, particularly Vibrio phage vB_VpP_1, with 84.2% identity and 46% coverage. Phylogenetic analysis based on the whole genome, the terminase large subunit, and the major capsid protein revealed that phage vB_VpM-pA2SJ1 did not cluster with other known phage families, thus indicating its uniqueness.


Asunto(s)
Bacteriófagos , Composición de Base , Genoma Viral , Sistemas de Lectura Abierta , Filogenia , Vibrio parahaemolyticus , Vibrio parahaemolyticus/virología , Vibrio parahaemolyticus/genética , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Bacteriófagos/clasificación , Animales , Penaeidae/virología , Penaeidae/microbiología , Vibriosis/microbiología , Vibriosis/virología , Vibriosis/veterinaria , Hepatopáncreas/virología , Hepatopáncreas/microbiología , Hepatopáncreas/patología , ADN Viral/genética
12.
Arch Microbiol ; 206(9): 379, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143367

RESUMEN

Multidrug-resistant Klebsiella pneumoniae (MDR-KP) poses a significant challenge in global healthcare, underscoring the urgency for innovative therapeutic approaches. Phage therapy emerges as a promising strategy amidst rising antibiotic resistance, emphasizing the crucial need to identify and characterize effective phage resources for clinical use. In this study, we introduce a novel lytic phage, RCIP0100, distinguished by its classification into the Chaoyangvirus genus and Fjlabviridae family based on International Committee on Taxonomy of Viruses (ICTV) criteria due to low genetic similarity to known phage families. Our findings demonstrate that RCIP0100 exhibits broad lytic activity against 15 out of 27 tested MDR-KP strains, including diverse profiles such as carbapenem-resistant K. pneumoniae (CR-KP). This positions phage RCIP0100 as a promising candidate for phage therapy. Strains resistant to RCIP0100 also showed increased susceptibility to various antibiotics, implying the potential for synergistic use of RCIP0100 and antibiotics as a strategic countermeasure against MDR-KP.


Asunto(s)
Antibacterianos , Bacteriófagos , Farmacorresistencia Bacteriana Múltiple , Klebsiella pneumoniae , Terapia de Fagos , Klebsiella pneumoniae/virología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Bacteriófagos/genética , Bacteriófagos/fisiología , Antibacterianos/farmacología , Infecciones por Klebsiella/microbiología , Genoma Viral , Humanos , Pruebas de Sensibilidad Microbiana
13.
Virology ; 599: 110209, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39186863

RESUMEN

While phages hold promise as an antibiotic alternative, they encounter significant challenges in combating bacterial infections, primarily due to the emergence of phage-resistant bacteria. Bacterial defence mechanisms like superinfection exclusion, CRISPR, and restriction-modification systems can hinder phage effectiveness. Innovative strategies, such as combining different phages into cocktails, have been explored to address these challenges. This review delves into these defence mechanisms and their impact at each stage of the infection cycle, their challenges, and the strategies phages have developed to counteract them. Additionally, we examine the role of phage cocktails in the evolving landscape of antibacterial treatments and discuss recent studies that highlight the effectiveness of diverse phage cocktails in targeting essential bacterial receptors and combating resistant strains.


Asunto(s)
Bacterias , Infecciones Bacterianas , Bacteriófagos , Terapia de Fagos , Bacteriófagos/fisiología , Bacteriófagos/genética , Bacterias/virología , Bacterias/genética , Infecciones Bacterianas/terapia , Infecciones Bacterianas/microbiología , Humanos , Antibacterianos/farmacología , Farmacorresistencia Bacteriana
14.
Proc Natl Acad Sci U S A ; 121(33): e2406138121, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39116131

RESUMEN

DNA recognition is critical for assembly of double-stranded DNA viruses, particularly for the initiation of packaging the viral genome into the capsid. The key component that recognizes viral DNA is the small terminase protein. Despite prior studies, the molecular mechanism for DNA recognition remained elusive. Here, we address this question by identifying the minimal site in the bacteriophage HK97 genome specifically recognized by the small terminase and determining the structure of this complex by cryoEM. The circular small terminase employs an entirely unexpected mechanism in which DNA transits through the central tunnel, and sequence-specific recognition takes place as it emerges. This recognition stems from a substructure formed by the N- and C-terminal segments of two adjacent protomers which are unstructured when DNA is absent. Such interaction ensures continuous engagement of the small terminase with DNA, enabling it to slide along the DNA while simultaneously monitoring its sequence. This mechanism allows locating and instigating packaging initiation and termination precisely at the specific cos sequence.


Asunto(s)
ADN Viral , Genoma Viral , ADN Viral/genética , ADN Viral/metabolismo , ADN Viral/química , Microscopía por Crioelectrón , Endodesoxirribonucleasas/metabolismo , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/genética , Modelos Moleculares , Empaquetamiento del ADN , Ensamble de Virus/genética , Bacteriófagos/genética , Empaquetamiento del Genoma Viral
15.
Elife ; 132024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39207920

RESUMEN

Viruses are ubiquitous in nature and play key roles in various ecosystems. Notably, some viruses (e.g. bacteriophage) exhibit alternative life cycles, such as chronic infections without cell lysis. However, the impact of chronic infections and their interactions with the host organisms remains largely unknown. Here, we found for the first time that polysaccharides induced the production of multiple temperate phages infecting two deep-sea Lentisphaerae strains (WC36 and zth2). Through physiological assays, genomic analysis, and transcriptomics assays, we found these bacteriophages were released via a chronic style without host cell lysis, which might reprogram host polysaccharide metabolism through the potential auxiliary metabolic genes. The findings presented here, together with recent discoveries made on the reprogramming of host energy-generating metabolisms by chronic bacteriophages, shed light on the poorly explored marine virus-host interaction and bring us closer to understanding the potential role of chronic viruses in marine ecosystems.


Asunto(s)
Bacteriófagos , Polisacáridos , Bacteriófagos/fisiología , Bacteriófagos/genética , Polisacáridos/metabolismo , Phaeophyceae/virología , Agua de Mar/virología , Agua de Mar/química , Interacciones Microbiota-Huesped
16.
Environ Pollut ; 359: 124713, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39134166

RESUMEN

Antibiotic resistance genes (ARGs) have been extensively observed in bacterial DNA, and more recently, in phage particles from various water sources and food items. The pivotal role played by ARG transmission in the proliferation of antibiotic resistance and emergence of new resistant strains calls for a thorough understanding of the underlying mechanisms. The aim of this study was to assess the suitability of the prototypical p-crAssphage, a proposed indicator of human fecal contamination, and the recently isolated crAssBcn phages, both belonging to the Crassvirales group, as potential indicators of ARGs. These crAss-like phages were evaluated alongside specific ARGs (blaTEM, blaCTX-M-1, blaCTX-M-9, blaVIM, blaOXA-48, qnrA, qnrS, tetW and sul1) within the total DNA and phage DNA fractions in water and food samples containing different levels of fecal pollution. In samples with high fecal load (>103 CFU/g or ml of E. coli or somatic coliphages), such as wastewater and sludge, positive correlations were found between both types of crAss-like phages and ARGs in both DNA fractions. The strongest correlation was observed between sul1 and crAssBcn phages (rho = 0.90) in sludge samples, followed by blaCTX-M-9 and p-crAssphage (rho = 0.86) in sewage samples, both in the phage DNA fraction. The use of crAssphage and crAssBcn as indicators of ARGs, considered to be emerging environmental contaminants of anthropogenic origin, is supported by their close association with the human gut. Monitoring ARGs can help to mitigate their dissemination and prevent the emergence of new resistant bacterial strains, thus safeguarding public health.


Asunto(s)
Bacteriófagos , Heces , Heces/microbiología , Heces/virología , Bacteriófagos/genética , Monitoreo del Ambiente/métodos , Humanos , Farmacorresistencia Microbiana/genética , Aguas Residuales/virología , Farmacorresistencia Bacteriana/genética , Escherichia coli/genética , Aguas del Alcantarillado , Antibacterianos/farmacología
17.
Int J Mol Sci ; 25(16)2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39201817

RESUMEN

Vibrio alginolyticus causes substantial economic losses in the aquaculture industry. With the rise of multidrug-resistant Vibrio strains, phages present a promising solution. Here, a novel lytic Vibrio phage, vB_ValC_RH2G (RH2G), that efficiently infects the pathogenic strain V. alginolyticus ATCC 17749T, was isolated from mixed wastewater from an aquatic market in Xiamen, China. Transmission electron microscopy revealed that RH2G has the morphology of Siphoviruses, featuring an icosahedral head (73 ± 2 nm diameter) and long noncontractile tail (142 ± 4 nm). A one-step growth experiment showed that RH2G had a short latent period (10 min) and a burst size of 48 phage particles per infected cell. Additionally, RH2G was highly species-specific and was relatively stable at 4-55 °C and pH 4-10. A genomic analysis showed that RH2G has a 116,749 bp double-stranded DNA genome with 43.76% GC content. The intergenomic similarity between the genome sequence of RH2G and other phages recorded in the GenBank database was below 38.8%, suggesting that RH2G represents a new genus. RH2G did not exhibit any virulence or resistance genes. Its rapid lysis capacity, lytic activity, environmental resilience, and genetic safety suggested that RH2G may be a safe candidate for phage therapy in combatting vibriosis in aquaculture settings.


Asunto(s)
Bacteriófagos , Genoma Viral , Vibrio alginolyticus , Vibrio alginolyticus/virología , Vibrio alginolyticus/genética , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Bacteriófagos/fisiología , Bacteriófagos/ultraestructura , Filogenia , Composición de Base
18.
Gut Microbes ; 16(1): 2392876, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39172643

RESUMEN

Fecal filtrate transfer (FFT) is emerging as a safer alternative to traditional fecal microbiota transplantation (FMT) - particularly in the context of necrotizing enterocolitis (NEC), a severe gastrointestinal condition affecting preterm infants. Using a preterm piglet model, FFT has demonstrated superiority over FMT in safety and NEC prevention. Since FFT is virtually devoid of bacteria, prokaryotic viruses (bacteriophages) are assumed to mediate the beneficial effects. However, this assumption remains unproven. To address this gap, we separated virus-like particles (30 kDa to 0.45 µm) of donor feces from the residual postbiotic fluid. We then compared clinical and gut microbiota responses to these fractions with the parent FFT solution after transferring them to NEC-susceptible preterm piglets. Virome transfer was equally effective as FFT in reducing the severity of NEC-like pathology. The bacterial compositional data corroborated clinical findings as virome transfer reduced the relative abundance of several NEC-associated pathogens e.g. Klebsiella pneumoniae and Clostridium perfringens. Virome transfer diversified gut viral communities with concomitant constraining effects on the bacterial composition. Unexpectedly, virome transfer, but not residual postbiotic fluid, led to earlier diarrhea. While diarrhea may be a minor concern in human infants, future work should identify ways of eliminating this side effect without losing treatment efficacy.


Asunto(s)
Enterocolitis Necrotizante , Trasplante de Microbiota Fecal , Heces , Microbioma Gastrointestinal , Enterocolitis Necrotizante/prevención & control , Enterocolitis Necrotizante/terapia , Animales , Heces/virología , Heces/microbiología , Trasplante de Microbiota Fecal/métodos , Porcinos , Humanos , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/genética , Animales Recién Nacidos , Modelos Animales de Enfermedad , Viroma , Clostridium perfringens , Bacteriófagos/genética , Bacteriófagos/fisiología , Diarrea/terapia , Diarrea/virología , Diarrea/prevención & control , Diarrea/microbiología
19.
Nat Commun ; 15(1): 7384, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39191765

RESUMEN

Toll/interleukin-1 receptor (TIR) domain-containing proteins play a critical role in immune responses in diverse organisms, but their function in bacterial systems remains to be fully elucidated. This study, focusing on Escherichia coli, addresses how TIR domain-containing proteins contribute to bacterial immunity against phage attack. Through an exhaustive survey of all E. coli genomes available in the NCBI database and testing of 32 representatives of the 90% of the identified TIR domain-containing proteins, we found that a significant proportion (37.5%) exhibit antiphage activities. These defense systems recognize a variety of phage components, thus providing a sophisticated mechanism for pathogen detection and defense. This study not only highlights the robustness of TIR systems in bacterial immunity, but also draws an intriguing parallel to the diversity seen in mammalian Toll-like receptors (TLRs), enriching our understanding of innate immune mechanisms across life forms and underscoring the evolutionary significance of these defense strategies in prokaryotes.


Asunto(s)
Bacteriófagos , Escherichia coli , Dominios Proteicos , Escherichia coli/genética , Escherichia coli/virología , Escherichia coli/inmunología , Escherichia coli/metabolismo , Bacteriófagos/genética , Bacteriófagos/inmunología , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/inmunología , Inmunidad Innata , Receptores Toll-Like/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-1/genética
20.
NPJ Biofilms Microbiomes ; 10(1): 72, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39191812

RESUMEN

Cophylogeny has been identified between gut bacteria and their animal host and is highly relevant to host health, but little research has extended to gut bacteriophages. Here we use bee model to investigate host specificity and cophylogeny in the "animal-gut bacteria-phage" tripartite system. Through metagenomic sequencing upon different bee species, the gut phageome revealed a more variable composition than the gut bacteriome. Nevertheless, the bacteriome and the phageome showed a significant association of their dissimilarity matrices, indicating a reciprocal interaction between the two kinds of communities. Most of the gut phages were host generalist at the viral cluster level but host specialist at the viral OTU level. While the dominant gut bacteria Gilliamella and Snodgrassella exhibited matched phylogeny with bee hosts, most of their phages showed a diminished level of cophylogeny. The evolutionary rates of the bee, the gut bacteria and the gut phages showed a remarkably increasing trend, including synonymous and non-synonymous substitution and gene content variation. For all of the three codiversified tripartite members, however, their genes under positive selection and genes involving gain/loss during evolution simultaneously enriched the functions into metabolism of nutrients, therefore highlighting the tripartite coevolution that results in an enhanced ecological fitness for the whole holobiont.


Asunto(s)
Bacterias , Bacteriófagos , Microbioma Gastrointestinal , Especificidad del Huésped , Filogenia , Animales , Bacteriófagos/genética , Bacteriófagos/fisiología , Abejas/virología , Abejas/microbiología , Bacterias/virología , Bacterias/genética , Bacterias/clasificación , Metagenómica/métodos , Metagenoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA