Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 261
Filtrar
1.
Food Microbiol ; 125: 104641, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39448151

RESUMO

Food contamination and biofilm formation by Shigella in food processing facilities are major causes of acute gastrointestinal infection and mortality in humans. Bacteriophages (phages) are promising alternatives to antibiotics in controlling plankton and biofilms in food matrices. This study isolated two novel phages, S2_01 and S2_02, with lytic activity against various Shigella spp. From sewage samples. Transmission electron microscopy revealed that phages S2_01 and S2_02 belonged to the Caudovirales order. On characterizing their lytic ability, phage S2_01 initially exhibited relatively weak antibacterial activity, while phage S2_02 initially displayed rapid antibacterial activity after phage application. A combination of these phages in a 1:9 ratio was selected, as it has been suggested to elicit the most rapid and sustained lysis ability for up to 24 h. It demonstrated lytic activity against various foodborne pathogens, including six Shigella spp. The phage cocktail exhibited biofilm inhibition and disruption abilities of approximately 79.29% and 42.55%, respectively, after 24 h in a 96-well microplate. In addition, inhibition (up to 23.42%) and disruption (up to 19.89%) abilities were also observed on stainless steel surfaces, and plankton growth was also significantly suppressed. Therefore, the phage cocktail formulated in this study displays great potential as a biological control agent in improving food safety against biofilms and plankton.


Assuntos
Bacteriófagos , Biofilmes , Shigella flexneri , Aço Inoxidável , Biofilmes/crescimento & desenvolvimento , Bacteriófagos/fisiologia , Shigella flexneri/virologia , Shigella flexneri/crescimento & desenvolvimento , Shigella flexneri/fisiologia , Esgotos/virologia , Esgotos/microbiologia , Contaminação de Alimentos/prevenção & controle , Contaminação de Alimentos/análise , Agentes de Controle Biológico/farmacologia , Microbiologia de Alimentos , Caudovirales/fisiologia
2.
Int J Mol Sci ; 25(18)2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39337427

RESUMO

The increasing interest in bacteriophage technology has prompted its novel applications to treat different medical conditions, most interestingly cancer. Due to their high specificity, manipulability, nontoxicity, and nanosize nature, phages are promising carriers in targeted therapy and cancer immunotherapy. This approach is particularly timely, as current challenges in cancer research include damage to healthy cells, inefficiency in targeting, obstruction by biological barriers, and drug resistance. Some cancers are being kept at the forefront of phage research, such as colorectal cancer and HCC, while others like lymphoma, cervical cancer, and myeloma have not been retouched in a decade. Common mechanisms are immunogenic antigen display on phage coats and the use of phage as transporters to carry drugs, genes, and other molecules. To date, popular phage treatments being tested are gene therapy and phage-based vaccines using M13 and λ phage, with some vaccines having advanced to human clinical trials. The results from most of these studies have been promising, but limitations in phage-based therapies such as reticuloendothelial system clearance or diffusion inefficiency must be addressed. Before phage-based therapies for cancer can be successfully used in oncology practice, more in-depth research and support from local governments are required.


Assuntos
Neoplasias , Terapia por Fagos , Humanos , Neoplasias/terapia , Neoplasias/imunologia , Terapia por Fagos/métodos , Bacteriófagos/fisiologia , Imunoterapia/métodos , Animais , Terapia Genética/métodos , Vacinas Anticâncer/uso terapêutico , Vacinas Anticâncer/imunologia
3.
Viruses ; 16(9)2024 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-39339906

RESUMO

Bacteriophages have been proposed as biological controllers to protect plants against different bacterial pathogens. In this scenario, one of the main challenges is the low viability of phages in plants and under adverse environmental conditions. This work explores the use of 12 compounds and 14 different formulations to increase the viability of a phage mixture that demonstrated biocontrol capacity against Pseudomonas syringae pv. actinidiae (Psa) in kiwi plants. The results showed that the viability of the phage mixture decreases at 44 °C, at a pH lower than 4, and under UV radiation. However, using excipients such as skim milk, casein, and glutamic acid can prevent the viability loss of the phages under these conditions. Likewise, it was demonstrated that the use of these compounds prolongs the presence of phages in kiwi plants from 48 h to at least 96 h. In addition, it was observed that phages remained stable for seven weeks when stored in powder with skim milk, casein, or sucrose after lyophilization and at 4 °C. Finally, the phages with glutamic acid, sucrose, or skim milk maintained their antimicrobial activity against Psa on kiwi leaves and persisted within kiwi plants when added through roots. This study contributes to overcoming the challenges associated with the use of phages as biological controllers in agriculture.


Assuntos
Doenças das Plantas , Pseudomonas syringae , Pseudomonas syringae/virologia , Pseudomonas syringae/efeitos dos fármacos , Doenças das Plantas/virologia , Doenças das Plantas/prevenção & controle , Doenças das Plantas/microbiologia , Agricultura/métodos , Actinidia/química , Bacteriófagos/fisiologia , Viabilidade Microbiana/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Agentes de Controle Biológico/farmacologia , Excipientes/química , Excipientes/farmacologia , Folhas de Planta/virologia , Folhas de Planta/química
4.
Int Orthop ; 48(11): 2809-2825, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39254722

RESUMO

BACKGROUND: Periprosthetic Joint Infection (PJI) following hip and knee arthroplasty is a catastrophic complication in orthopaedic surgery. It has long been a key focus for orthopaedic surgeons in terms of prevention and management. With the increasing incidence of antibiotic resistance in recent years, finding more targeted treatment methods has become an increasingly urgent issue. Bacteriophage Therapy (BT) has emerged as a promising adjunctive treatment for bone and joint infections in recent years. It not only effectively kills bacteria but also demonstrates significant anti-biofilm activity, garnering substantial clinical interest due to its demonstrated efficacy and relatively low incidence of adverse effects. PURPOSE: This review aims to systematically evaluate the efficacy and safety of bacteriophage therapy in treating PJI following hip and knee arthroplasty, providing additional reference for its future clinical application. METHODS: Following predefined inclusion and exclusion criteria, our team conducted a systematic literature search across seven databases (PubMed, Embase, Web of Science, Cochrane Library, ClinicalTrials.gov, CNKI, and WanFang Database). The search was conducted up to May 2024 and included multiple clinical studies on the use of bacteriophage therapy for treating PJI after hip and knee arthroplasty to assess its efficacy and safety. RESULTS: This systematic review included 16 clinical studies after screening, consisting of 15 case reports and one prospective controlled clinical trial, involving a total of 42 patients with PJI treated with bacteriophage therapy. The average patient age was 62.86 years, and 43 joints were treated, with patients undergoing an average of 5.25 surgeries. The most common pathogen in these infections was Staphylococcus aureus, accounting for 18 cases. 33 patients received cocktail therapy, while nine were treated with a single bacteriophage preparation. Additionally, all patients underwent suppressive antibiotic therapy (SAT) postoperatively. All patients were followed up for an average of 13.55 months. There were two cases of recurrence, one of which resulted in amputation one year postoperatively. The remaining patients showed good recovery outcomes. Overall, the results from the included studies indicate that bacteriophage therapy effectively eradicates infectious strains in various cases of PJI, with minimal side effects, demonstrating promising clinical efficacy. CONCLUSION: In the treatment of PJI following hip and knee arthroplasty, bacteriophages, whether used alone or in combination as cocktail therapy, have shown therapeutic potential. However, thorough preoperative evaluation is essential, and appropriate bacteriophage types and treatment regimens must be selected based on bacteriological evidence. Future large-scale, randomized controlled, and prospective trials are necessary to validate the efficacy and safety of this therapy.


Assuntos
Artroplastia do Joelho , Terapia por Fagos , Infecções Relacionadas à Prótese , Humanos , Infecções Relacionadas à Prótese/terapia , Infecções Relacionadas à Prótese/etiologia , Artroplastia do Joelho/efeitos adversos , Terapia por Fagos/métodos , Artroplastia de Quadril/efeitos adversos , Bacteriófagos/fisiologia
5.
Microbiol Spectr ; 12(10): e0420023, 2024 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-39264185

RESUMO

Jumbo phages are characterized by their remarkably large-sized genome and unique life cycles. Jumbo phages belonging to Chimalliviridae family protect the replicating phage DNA from host immune systems like CRISPR-Cas and restriction-modification system through a phage nucleus structure. Several recent studies have provided new insights into jumbo phage infection biology, but the pan-genome diversity of jumbo phages and their relationship with CRISPR-Cas targeting beyond Chimalliviridae are not well understood. In this study, we used pan-genome analysis to identify orthologous gene families shared among 331 jumbo phages with complete genomes. We show that jumbo phages lack a universally conserved set of core genes but identified seven "soft-core genes" conserved in over 50% of these phages. These genes primarily govern DNA-related activities, such as replication, repair, or nucleotide synthesis. Jumbo phages exhibit a wide array of accessory and unique genes, underscoring their genetic diversity. Phylogenetic analyses of the soft-core genes revealed frequent horizontal gene transfer events between jumbo phages, non-jumbo phages, and occasionally even giant eukaryotic viruses, indicating a polyphyletic evolutionary nature. We categorized jumbo phages into 11 major viral clusters (VCs) spanning 130 sub-clusters, with the majority being multi-genus jumbo phage clusters. Moreover, through the analysis of hallmark genes related to CRISPR-Cas targeting, we predict that many jumbo phages can evade host immune systems using both known and yet-to-be-identified mechanisms. In summary, our study enhances our understanding of jumbo phages, shedding light on their pan-genome diversity and remarkable genome protection capabilities. IMPORTANCE: Jumbo phages are large bacterial viruses known for more than 50 years. However, only in recent years, a significant number of complete genome sequences of jumbo phages have become available. In this study, we employed comparative genomic approaches to investigate the genomic diversity and genome protection capabilities of the 331 jumbo phages. Our findings revealed that jumbo phages exhibit high genetic diversity, with only a few genes being relatively conserved across jumbo phages. Interestingly, our data suggest that jumbo phages employ yet-to-be-identified strategies to protect their DNA from the host immune system, such as CRISPR-Cas.


Assuntos
Bacteriófagos , Sistemas CRISPR-Cas , Variação Genética , Genoma Viral , Genômica , Filogenia , Sistemas CRISPR-Cas/genética , Genoma Viral/genética , Bacteriófagos/genética , Bacteriófagos/fisiologia , Transferência Genética Horizontal/genética
6.
Int J Mol Sci ; 25(16)2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39201374

RESUMO

As knowledge of the gut microbiome has expanded our understanding of the symbiotic and dysbiotic relationships between the human host and its microbial constituents, the influence of gastrointestinal (GI) microbes both locally and beyond the intestine has become evident. Shifts in bacterial populations have now been associated with several conditions including Crohn's disease (CD), Ulcerative Colitis (UC), irritable bowel syndrome (IBS), Alzheimer's disease, Parkinson's Disease, liver diseases, obesity, metabolic syndrome, anxiety, depression, and cancers. As the bacteria in our gut thrive on the food we eat, diet plays a critical role in the functional aspects of our gut microbiome, influencing not only health but also the development of disease. While the bacterial microbiome in the context of disease is well studied, the associated gut phageome-bacteriophages living amongst and within our bacterial microbiome-is less well understood. With growing evidence that fluctuations in the phageome also correlate with dysbiosis, how diet influences this population needs to be better understood. This review surveys the current understanding of the effects of diet on the gut phageome.


Assuntos
Bacteriófagos , Dieta , Disbiose , Microbioma Gastrointestinal , Humanos , Microbioma Gastrointestinal/fisiologia , Bacteriófagos/fisiologia , Disbiose/microbiologia , Animais , Viroma
7.
Nat Commun ; 15(1): 7473, 2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39209841

RESUMO

The antagonistic coevolution of microbes and viruses influences fundamentally the diversity of microbial communities. Information on how environmental variables interact with emergent defense-counterdefense strategies and community composition is, however, still scarce. Following biological intuition, diversity should increase with improved growth conditions, which offset evolutionary costs; however, laboratory and regional data suggest that microbial diversity decreases in nutrient-rich conditions. Moreover, global oceanic data show that microbial and viral diversity decline for high latitudes, although the underlying mechanisms are unknown. This article addresses these gaps by introducing an eco-evolutionary model for bacteria-virus antagonistic coevolution. The theory presented here harmonizes the observations above and identifies negative density dependence and viral plasticity (dependence of virus performance on host physiological state) as key drivers: environmental conditions selecting for slow host growth also limit viral performance, facilitating the survival of a diverse host community; host diversity, in turn, enables viral portfolio effects and bet-hedging strategies that sustain viral diversity. From marine microbes to phage therapy against antibiotic-resistant bacteria or cancer cells, the ubiquity of antagonistic coevolution highlights the need to consider eco-evolutionary interactions across a gradient of growth conditions.


Assuntos
Bactérias , Biodiversidade , Bactérias/virologia , Bactérias/genética , Bacteriófagos/fisiologia , Evolução Biológica , Microbiota , Interações entre Hospedeiro e Microrganismos , Vírus/genética , Interações Hospedeiro-Patógeno
8.
Int J Mol Sci ; 25(15)2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39125890

RESUMO

Patients with cystic fibrosis (CF) are prone to developing life-threatening lung infections with a variety of pathogens that are difficult to eradicate, such as Burkholderia cepacia complex (Bcc), Hemophilus influenzae, Mycobacterium abscessus (Mab), Pseudomonas aeruginosa, and Staphylococcus aureus. These infections still remain an important issue, despite the therapy for CF having considerably improved in recent years. Moreover, prolonged exposure to antibiotics in combination favors the development and spread of multi-resistant bacteria; thus, the development of alternative strategies is crucial to counter antimicrobial resistance. In this context, phage therapy, i.e., the use of phages, viruses that specifically infect bacteria, has become a promising strategy. In this review, we aim to address the current status of phage therapy in the management of multidrug-resistant infections, from compassionate use cases to ongoing clinical trials, as well as the challenges this approach presents in the particular context of CF patients.


Assuntos
Infecções Bacterianas , Fibrose Cística , Farmacorresistência Bacteriana Múltipla , Terapia por Fagos , Fibrose Cística/terapia , Fibrose Cística/microbiologia , Humanos , Terapia por Fagos/métodos , Infecções Bacterianas/terapia , Antibacterianos/uso terapêutico , Bacteriófagos/fisiologia
9.
Microbiol Spectr ; 12(9): e0081724, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39101807

RESUMO

The discovery of Replication Competent Circular DNA molecules in mammalian cells and tissues is being linked to debilitating diseases, such as multiple sclerosis (MS), bovine spongiform encephalopathy (BSE), and colorectal cancer (CRC). These circular DNA molecules, otherwise known as bovine meat and milk factors (BMMFs) and Slow Progressive Hidden INfections of variable (X) latency (SPHINX), bear significant (80%) sequence similarity with the plasmids of Acinetobacter baumannii strains. Nanostructures, such as bacterial outer membrane vesicles (OMVs) serve as vehicles for transporting biomolecular cargo and have the potential to facilitate interkingdom lateral mobility of DNA. Strengthening the proposed hypothesis, this study demonstrates that OMVs derived from A. baumannii DS002 carrying four plasmids and genome (pTS236) of phage, AbDs1, successfully reached different parts of the body, including the central nervous system, following the injection of fluorescein isothiocyanate (FITC)-labeled OMVs into experimental mice. Out of the four OMV-associated plasmids, three (pTS4586, pTS9900, and pTS134338) were identified within the lumen, and the fourth one (pTS11291) was found on the surface of OMVs. In addition to the indigenous plasmids, the phage-encoded protein, Orf96, anchored on the surface of the OMVs by establishing a strong interaction with the OMV-associated porin, OmpA. Intriguingly, a subset of labeled OMVs, when incubated with Neuro2A cells, translocated across the membrane and reached to the cytoplasmic space of the cells. Collectively, the experimental evidence presented herein underscores the promising potential of OMVs as vehicles for delivering molecular cargo containing plasmids and phage genomes to diverse mammalian tissues and cells. IMPORTANCE: Several independent studies have demonstrated the existence of replication competent circular DNA molecules of bacterial and viral origin in mammalian cells and tissues. However, studies about their origin and lateral mobility to mammalian cells are scarce. Our work describes the existence of circular DNA, similar to that of DNA molecules identified in mammalian cells, OMVs derived from soil isolate of A. baumannii DS002. Furthermore, the work also provides visual evidence that demonstrates the passage of labeled OMVs to different organs of experimental mice within hours after intravenously administering OMVs into experimental mice. Some of the labeled OMVs have even crossed the membrane of Neuro2A, suggesting the existence of interkingdom horizontal mobility between bacteria and mammals.


Assuntos
Acinetobacter baumannii , DNA Circular , Transferência Genética Horizontal , Plasmídeos , Animais , Camundongos , Plasmídeos/genética , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Bovinos , DNA Circular/genética , DNA Circular/metabolismo , Membrana Externa Bacteriana/metabolismo , Feminino , Bacteriófagos/genética , Bacteriófagos/fisiologia , Carne/microbiologia , Leite/microbiologia , Infecções por Acinetobacter/microbiologia , Vesículas Extracelulares/metabolismo , Camundongos Endogâmicos BALB C , DNA Bacteriano/genética
10.
Viruses ; 16(7)2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-39066214

RESUMO

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.


Assuntos
Fibrose Cística , Terapia por Fagos , Infecções por Pseudomonas , Fagos de Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virologia , Humanos , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/terapia , Fibrose Cística/microbiologia , Fagos de Pseudomonas/genética , Fagos de Pseudomonas/fisiologia , Animais , Bacteriófagos/fisiologia , Bacteriófagos/genética , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico
11.
Nat Commun ; 15(1): 5626, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38992046

RESUMO

As bacteriophages continue to gain regulatory approval for personalized human therapy against antibiotic-resistant infections, there is a need for transformative technologies for rapid target identification through multiple, large, decentralized therapeutic phages biobanks. Here, we design a high throughput phage screening platform comprised of a portable library of individual shelf-stable, ready-to-use phages, in all-inclusive solid tablets. Each tablet encapsulates one phage along with luciferin and luciferase enzyme stabilized in a sugar matrix comprised of pullulan and trehalose capable of directly detecting phage-mediated adenosine triphosphate (ATP) release through ATP bioluminescence reaction upon bacterial cell burst. The tablet composition also enhances desiccation tolerance of all components, which should allow easier and cheaper international transportation of phages and as a result, increased accessibility to therapeutic phages. We demonstrate high throughput screening by identifying target phages for select multidrug-resistant clinical isolates of Pseudomonas aeruginosa, Salmonella enterica, Escherichia coli, and Staphylococcus aureus with targets identified within 30-120 min.


Assuntos
Bacteriófagos , Escherichia coli , Ensaios de Triagem em Larga Escala , Terapia por Fagos , Medicina de Precisão , Staphylococcus aureus , Humanos , Terapia por Fagos/métodos , Ensaios de Triagem em Larga Escala/métodos , Escherichia coli/virologia , Escherichia coli/metabolismo , Escherichia coli/genética , Bacteriófagos/genética , Bacteriófagos/fisiologia , Staphylococcus aureus/virologia , Medicina de Precisão/métodos , Pseudomonas aeruginosa/virologia , Trifosfato de Adenosina/metabolismo , Salmonella enterica/virologia , Farmacorresistência Bacteriana Múltipla/genética
12.
Int J Antimicrob Agents ; 64(3): 107276, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39009289

RESUMO

OBJECTIVES: Staphylococcus aureus and Pseudomonas aeruginosa co-infections in patients with cystic fibrosis (CF) are associated with disease severity. Their treatment is complicated by biofilm formation in the sticky mucus obstructing the airways. We investigated the activity of phages-antibiotics combinations using a dual species biofilm (P. aeruginosa/S. aureus) formed in artificial sputum medium. METHODS: Biofilmswere incubated with broad-spectrum antibiotics (meropenem, ceftazidime, ciprofloxacin, tobramycin) combined with a cocktail of two (bacterio)phages (PSP3 and ISP) proven active via spot tests and double agar on P. aeruginosa PAO1 and S. aureus ATCC 25923. RESULTS: At the highest tested concentrations (100 x MIC), antibiotics alone caused a 20-50% reduction in biomass and reduced S. aureus and P. aeruginosa CFU of 2.3 to 2.8 and 2.1 to 3.6 log10, respectively. Phages alone reduced biofilm biomass by 23% and reduced P. aeruginosa CFU of 2.1 log10, but did not affect S. aureus viability. Phages enhanced antibiotic effects on biomass and exhibited additive effects with antibiotics against P. aeruginosa, but not against S. aureus. Following inhibition of bacterial respiration by phages in planktonic cultures rationalised these observations by demonstrating that PSP3 was effective at multiplicities of infection (MOI) as low as 10-4 plaque forming units (PFU)/CFU on P. aeruginosa, but ISP, at higher MOI (> 0.1) against S. aureus. CONCLUSION: Pre-screening inhibition of bacterial respiration by phages may assist in selecting those showing activity at sufficiently low titers to showcase anti-biofilm activity in this complex but clinically-relevant in vitro model of biofilm.


Assuntos
Antibacterianos , Biofilmes , Fibrose Cística , Pseudomonas aeruginosa , Staphylococcus aureus , Biofilmes/efeitos dos fármacos , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/virologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/virologia , Staphylococcus aureus/fisiologia , Antibacterianos/farmacologia , Fibrose Cística/microbiologia , Humanos , Testes de Sensibilidade Microbiana , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/tratamento farmacológico , Terapia por Fagos/métodos , Bacteriófagos/fisiologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/tratamento farmacológico
13.
BMC Microbiol ; 24(1): 211, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38877452

RESUMO

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


Assuntos
Bacteriófagos , Biofilmes , Genoma Viral , Klebsiella pneumoniae , Klebsiella pneumoniae/virologia , Klebsiella pneumoniae/genética , Biofilmes/crescimento & desenvolvimento , Bacteriófagos/genética , Bacteriófagos/fisiologia , Bacteriófagos/classificação , Bacteriófagos/isolamento & purificação , Myoviridae/genética , Myoviridae/fisiologia , Myoviridae/classificação , Farmacorresistência Bacteriana Múltipla/genética , Filogenia , DNA Viral/genética , Composição de Bases , Terapia por Fagos
14.
mSystems ; 9(6): e0103623, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38727217

RESUMO

Temperate bacteriophages (phages) are common features of bacterial genomes and can act as self-amplifying biological weapons, killing susceptible competitors and thus increasing the fitness of their bacterial hosts (lysogens). Despite their prevalence, however, the key characteristics of an effective temperate phage weapon remain unclear. Here, we use systematic mathematical analyses coupled with experimental tests to understand what makes an effective temperate phage weapon. We find that effectiveness is controlled by phage life history traits-in particular, the probability of lysis and induction rate-but that the optimal combination of traits varies with the initial frequency of a lysogen within a population. As a consequence, certain phage weapons can be detrimental when their hosts are rare yet beneficial when their hosts are common, while subtle changes in individual life history traits can completely reverse the impact of an individual phage weapon on lysogen fitness. We confirm key predictions of our model experimentally, using temperate phages isolated from the clinically relevant Liverpool epidemic strain of Pseudomonas aeruginosa. Through these experiments, we further demonstrate that nutrient availability can also play a critical role in driving frequency-dependent patterns in phage-mediated competition. Together, these findings highlight the complex and context-dependent nature of temperate phage weapons and the importance of both ecological and evolutionary processes in shaping microbial community dynamics more broadly. IMPORTANCE: Temperate bacteriophages-viruses that integrate within bacterial DNA-are incredibly common within bacterial genomes and can act as powerful self-amplifying weapons. Bacterial hosts that carry temperate bacteriophages can thus gain a fitness advantage within a given niche by killing competitors. But what makes an effective phage weapon? Here, we first use a simple mathematical model to explore the factors determining bacteriophage weapon utility. Our models suggest that bacteriophage weapons are nuanced and context-dependent; an individual bacteriophage may be beneficial or costly depending upon tiny changes to how it behaves or the bacterial community it inhabits. We then confirm these mathematical predictions experimentally, using phages isolated from cystic fibrosis patients. But, in doing so, we also find that another factor-nutrient availability-plays a key role in shaping bacteriophage-mediated competition. Together, our results provide new insights into how temperate bacteriophages modulate bacterial communities.


Assuntos
Lisogenia , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virologia , Bacteriófagos/genética , Bacteriófagos/fisiologia
15.
Viruses ; 16(5)2024 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-38793624

RESUMO

Bacteriophages exert strong selection on their bacterial hosts to evolve resistance. At the same time, the fitness costs on bacteria following phage resistance may change their virulence, which may affect the therapeutic outcomes of phage therapy. In this study, we set out to assess the costs of phage resistance on the in vitro virulence of priority 1 nosocomial pathogenic bacterium, Acinetobacter baumannii. By subjecting phage-resistant variant Ev5-WHG of A. baumannii WHG40004 to several in vitro virulence profiles, we found that its resistance to phage is associated with reduced fitness in host microenvironments. Also, the mutant exhibited impaired adhesion and invasion to mammalian cells, as well as increased susceptibility to macrophage phagocytosis. Furthermore, the whole-genome sequencing of the mutant revealed that there exist multiple mutations which may play a role in phage resistance and altered virulence. Altogether, this study demonstrates that resistance to phage can significantly alter phenotypes associated with virulence in Acinetobacter baumannii.


Assuntos
Infecções por Acinetobacter , Acinetobacter baumannii , Bacteriófagos , Fenótipo , Acinetobacter baumannii/virologia , Acinetobacter baumannii/patogenicidade , Acinetobacter baumannii/genética , Virulência/genética , Bacteriófagos/genética , Bacteriófagos/fisiologia , Bacteriófagos/patogenicidade , Infecções por Acinetobacter/microbiologia , Animais , Humanos , Macrófagos/microbiologia , Macrófagos/virologia , Mutação , Fagocitose , Sequenciamento Completo do Genoma , Camundongos
16.
Virology ; 596: 110101, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38754335

RESUMO

This study characterizes a newly isolated Demerecviridae phage, named vB_SalS_PSa2, belonging to the phage T5 group. The main variations between vB_SalS_PSa2 and T5 concern structural proteins related to morphology and host recognition. vB_SalS_PSa2 is infective to 19 out of the 25 tested Salmonella enterica (including the rare "Sendai" and "Equine" serotypes) and Escherichia coli isolates, most of them being multidrug resistant. vB_SalS_PSa2 displayed good thermal stability (4-60 °C) and broad pH stability (4.0-12.0). It also exhibited antibacterial activity against S. enterica sv. Paratyphi A Enb50 at 4 °C in milk during the whole tested period (5 d), and for 3-6 h at both 25 and 37 °C. Furthermore, vB_SalS_PSa2 was able to inhibit biofilm formation and to show degradation activity on mature biofilms of E. coli K12 and S. enterica sv. Paratyphi Enb50 in both LB and milk. Altogether, these results indicate that phage vB_SalS_PSa2 is a valuable candidate for controlling foodborne S. enterica and E. coli pathogens.


Assuntos
Escherichia coli , Salmonella enterica , Salmonella enterica/virologia , Escherichia coli/virologia , Leite/virologia , Animais , Microbiologia de Alimentos , Genoma Viral , Biofilmes/crescimento & desenvolvimento , Fagos de Salmonella/fisiologia , Fagos de Salmonella/isolamento & purificação , Fagos de Salmonella/classificação , Fagos de Salmonella/genética , Bacteriófagos/fisiologia , Bacteriófagos/genética , Bacteriófagos/classificação , Bacteriófagos/isolamento & purificação , Concentração de Íons de Hidrogênio , Filogenia , Especificidade de Hospedeiro
17.
Nucleic Acids Res ; 52(10): 5895-5911, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38716875

RESUMO

Argonautes are an evolutionary conserved family of programmable nucleases that identify target nucleic acids using small guide oligonucleotides. In contrast to eukaryotic Argonautes (eAgos) that act on RNA, most studied prokaryotic Argonautes (pAgos) recognize DNA targets. Similarly to eAgos, pAgos can protect prokaryotic cells from invaders, but the biogenesis of guide oligonucleotides that confer them specificity to their targets remains poorly understood. Here, we have identified a new group of RNA-guided pAgo nucleases and demonstrated that a representative pAgo from this group, AmAgo from the mesophilic bacterium Alteromonas macleodii, binds guide RNAs of varying lengths for specific DNA targeting. Unlike most pAgos and eAgos, AmAgo is strictly specific to hydroxylated RNA guides containing a 5'-adenosine. AmAgo and related pAgos are co-encoded with a conserved RNA endonuclease from the HEPN superfamily (Ago-associated protein, Agap-HEPN). In vitro, Agap cleaves RNA between guanine and adenine nucleotides producing hydroxylated 5'-A guide oligonucleotides bound by AmAgo. In vivo, Agap cooperates with AmAgo in acquiring guide RNAs and counteracting bacteriophage infection. The AmAgo-Agap pair represents the first example of a pAgo system that autonomously produces RNA guides for DNA targeting and antiviral defense, which holds promise for programmable DNA targeting in biotechnology.


Assuntos
Alteromonas , Proteínas Argonautas , DNA Viral , RNA Guia de Sistemas CRISPR-Cas , Ribonucleases , Proteínas Argonautas/metabolismo , Proteínas Argonautas/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Ribonucleases/metabolismo , RNA Guia de Sistemas CRISPR-Cas/genética , RNA Guia de Sistemas CRISPR-Cas/metabolismo , Alteromonas/enzimologia , Alteromonas/virologia , DNA Viral/metabolismo , Bacteriófagos/fisiologia
18.
PLoS Biol ; 22(4): e3002566, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38652717

RESUMO

Phage therapy is a therapeutic approach to treat multidrug-resistant (MDR) infections that employs lytic bacteriophages (phages) to eliminate bacteria. Despite the abundant evidence for its success as an antimicrobial in Eastern Europe, there is scarce data regarding its effects on the human host. Here, we aimed to understand how lytic phages interact with cells of the airway epithelium, the tissue site that is colonized by bacterial biofilms in numerous chronic respiratory disorders. Using a panel of Pseudomonas aeruginosa phages and human airway epithelial cells (AECs) derived from a person with cystic fibrosis (CF), we determined that interactions between phages and epithelial cells depend on specific phage properties as well as physiochemical features of the microenvironment. Although poor at internalizing phages, the airway epithelium responds to phage exposure by changing its transcriptional profile and secreting antiviral and proinflammatory cytokines that correlate with specific phage families. Overall, our findings indicate that mammalian responses to phages are heterogenous and could potentially alter the way that respiratory local defenses aid in bacterial clearance during phage therapy. Thus, besides phage receptor specificity in a particular bacterial isolate, the criteria to select lytic phages for therapy should be expanded to include mammalian cell responses.


Assuntos
Fibrose Cística , Citocinas , Células Epiteliais , Pseudomonas aeruginosa , Humanos , Pseudomonas aeruginosa/virologia , Células Epiteliais/virologia , Células Epiteliais/metabolismo , Células Epiteliais/imunologia , Citocinas/metabolismo , Fibrose Cística/terapia , Fibrose Cística/imunologia , Fibrose Cística/metabolismo , Terapia por Fagos , Bacteriófagos/fisiologia , Bacteriófagos/genética , Mucosa Respiratória/virologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/imunologia , Infecções por Pseudomonas/terapia , Infecções por Pseudomonas/imunologia , Fagos de Pseudomonas/metabolismo , Biofilmes
19.
J Virol ; 98(3): e0173123, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38329345

RESUMO

In our 2012 genome announcement (J Virol 86:11403-11404, 2012, https://doi.org/10.1128/JVI.01954-12), we initially identified the host bacterium of bacteriophage Enc34 as Enterobacter cancerogenus using biochemical tests. However, later in-house DNA sequencing revealed that the true host is a strain of Hafnia alvei. Capitalizing on our new DNA-sequencing capabilities, we also refined the genomic termini of Enc34, confirming a 60,496-bp genome with 12-nucleotide 5' cohesive ends. IMPORTANCE: Our correction reflects the evolving landscape of bacterial identification, where molecular methods have supplanted traditional biochemical tests. This case underscores the significance of revisiting past identifications, as seemingly known bacterial strains may yield unexpected discoveries, necessitating essential updates to the scientific record. Despite the host identity correction, our genome announcement retains importance as the first complete genome sequence of a Hafnia alvei bacteriophage.


Assuntos
Bacteriófagos , Hafnia alvei , Tropismo ao Hospedeiro , Bacteriófagos/classificação , Bacteriófagos/genética , Bacteriófagos/isolamento & purificação , Bacteriófagos/fisiologia , Enterobacter/química , Enterobacter/virologia , Genoma Viral/genética , Hafnia alvei/classificação , Hafnia alvei/genética , Hafnia alvei/virologia , Erro Científico Experimental , Análise de Sequência de DNA
20.
Nat Struct Mol Biol ; 31(5): 767-776, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38321146

RESUMO

The bacterial cyclic oligonucleotide-based antiphage signaling system (CBASS) is similar to the cGAS-STING system in humans, containing an enzyme that synthesizes a cyclic nucleotide on viral infection and an effector that senses the second messenger for the antiviral response. Cap5, containing a SAVED domain coupled to an HNH DNA endonuclease domain, is the most abundant CBASS effector, yet the mechanism by which it becomes activated for cell killing remains unknown. We present here high-resolution structures of full-length Cap5 from Pseudomonas syringae (Ps) with second messengers. The key to PsCap5 activation is a dimer-to-tetramer transition, whereby the binding of second messenger to dimer triggers an open-to-closed transformation of the SAVED domains, furnishing a surface for assembly of the tetramer. This movement propagates to the HNH domains, juxtaposing and converting two HNH domains into states for DNA destruction. These results show how Cap5 effects bacterial cell suicide and we provide proof-in-principle data that the CBASS can be extrinsically activated to limit bacterial infections.


Assuntos
Proteínas de Bactérias , Endonucleases , Pseudomonas syringae , Pseudomonas syringae/química , Pseudomonas syringae/enzimologia , Pseudomonas syringae/virologia , Proteínas de Bactérias/química , Endonucleases/química , Ligantes , Modelos Químicos , Ativação Enzimática , DNA/química , DNA/metabolismo , Nucleotídeos Cíclicos/química , Fosfatos de Dinucleosídeos/química , Apoproteínas/química , Bacteriófagos/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA