Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 406
Filtrar
1.
Virology ; 595: 110090, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38718447

RESUMO

Nowadays finding the new antimicrobials is necessary due to the emerging of multidrug resistant strains. The present study aimed to isolate and characterize bacteriophages against S. aureus. Strains Huma and Simurgh were the two podovirus morphology phages which isolated and then characterized. Huma and Simurgh had a genome size of 16,853 and 17,245 bp, respectively and both were Rosenblumvirus with G + C content of 29%. No lysogeny-related genes, nor virulence genes were identified in their genomes. They were lytic only against two out of four S. aureus strains. They also were able to inhibit S. aureus for 8 h in-vitro. Both showed a rapid adsorption. Huma and Simurgh had the latent period of 80 and 60 m and the burst sizes of 45 and 40 PFU/ml and also, they showed very low cell toxicity of 1.23%-1.79% on HT-29 cells, respectively. Thus, they can be considered potential candidates for biocontrol applications.


Assuntos
Genoma Viral , Fagos de Staphylococcus , Staphylococcus aureus , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/fisiologia , Fagos de Staphylococcus/isolamento & purificação , Staphylococcus aureus/virologia , Staphylococcus aureus/genética , Humanos , Composição de Bases , Podoviridae/genética , Podoviridae/isolamento & purificação , Podoviridae/classificação , Podoviridae/fisiologia , Células HT29 , Tamanho do Genoma
2.
Int J Pharm ; 657: 124146, 2024 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-38657716

RESUMO

Host cell invasion with strong antibiotics evading is a major feature of respiratory Staphylococcus aureus infections with severe recurrence. Bacteriophage (phage) therapy and design of liposomal phage to target intracellular pathogens have been described recently. The practicality for pulmonary delivery of liposomal phage, and how formulation compositions affecting the aerosolization and intracellular bacterial killing remain unexplored. In the present study, three commonly used phospholipids (SPC, EPC, and HSPC) were selected to investigate their ability for phage K nebulization and intracellular therapy in the form of liposome-phage nanocomplexes. The three lipid nanocarriers showed protection on phage K upon mesh nebulization and the pulmonary deposition efficiency was influenced by the lipid used. Moreover, the intracellular bacterial killing was strongly depended on the lipid types, where EPC-phage exhibited the best killing performance with no relapsing. Phage K with the aid of EPC liposomes was also observed to manage the tissue infection in a 3D spheroid model more effectively than other groups. Altogether, this novel EPC liposome-phage nanocomplex can be a promising formulation approach that enables inhalable phage to manage respiratory infections caused by bacteria strongly associated with human epithelial cells.


Assuntos
Técnicas de Cocultura , Células Epiteliais , Lipossomos , Staphylococcus aureus , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/virologia , Humanos , Células Epiteliais/virologia , Fosfolipídeos/química , Bacteriófagos , Infecções Estafilocócicas , Administração por Inalação , Nanopartículas , Nebulizadores e Vaporizadores
3.
Nat Microbiol ; 9(5): 1312-1324, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38565896

RESUMO

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


Assuntos
Prófagos , Staphylococcus aureus , Prófagos/genética , Staphylococcus aureus/virologia , Staphylococcus aureus/imunologia , Autoimunidade , Lisogenia , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/imunologia , Fagos de Staphylococcus/fisiologia , Proteínas da Cauda Viral/genética , Proteínas da Cauda Viral/metabolismo , Bacteriófagos/genética , Bacteriófagos/imunologia , Bacteriófagos/fisiologia
4.
Nature ; 625(7996): 797-804, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38200316

RESUMO

Prokaryotic type III CRISPR-Cas systems provide immunity against viruses and plasmids using CRISPR-associated Rossman fold (CARF) protein effectors1-5. Recognition of transcripts of these invaders with sequences that are complementary to CRISPR RNA guides leads to the production of cyclic oligoadenylate second messengers, which bind CARF domains and trigger the activity of an effector domain6,7. Whereas most effectors degrade host and invader nucleic acids, some are predicted to contain transmembrane helices without an enzymatic function. Whether and how these CARF-transmembrane helix fusion proteins facilitate the type III CRISPR-Cas immune response remains unknown. Here we investigate the role of cyclic oligoadenylate-activated membrane protein 1 (Cam1) during type III CRISPR immunity. Structural and biochemical analyses reveal that the CARF domains of a Cam1 dimer bind cyclic tetra-adenylate second messengers. In vivo, Cam1 localizes to the membrane, is predicted to form a tetrameric transmembrane pore, and provides defence against viral infection through the induction of membrane depolarization and growth arrest. These results reveal that CRISPR immunity does not always operate through the degradation of nucleic acids, but is instead mediated via a wider range of cellular responses.


Assuntos
Bacteriófagos , Sistemas CRISPR-Cas , Potenciais da Membrana , Staphylococcus aureus , Bacteriófagos/imunologia , Bacteriófagos/metabolismo , Proteínas Associadas a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/imunologia , Nucleotídeos Cíclicos/metabolismo , RNA Guia de Sistemas CRISPR-Cas , Sistemas do Segundo Mensageiro , Staphylococcus aureus/citologia , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , Staphylococcus aureus/virologia
5.
J Mol Biol ; 436(4): 168415, 2024 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-38135177

RESUMO

Staphylococcus aureus is an important human pathogen, and the prevalence of antibiotic resistance is a major public health concern. The evolution of pathogenicity and resistance in S. aureus often involves acquisition of mobile genetic elements (MGEs). Bacteriophages play an especially important role, since transduction represents the main mechanism for horizontal gene transfer. S. aureus pathogenicity islands (SaPIs), including SaPI1, are MGEs that carry genes encoding virulence factors, and are mobilized at high frequency through interactions with specific "helper" bacteriophages, such as 80α, leading to packaging of the SaPI genomes into virions made from structural proteins supplied by the helper. Among these structural proteins is the portal protein, which forms a ring-like portal at a fivefold vertex of the capsid, through which the DNA is packaged during virion assembly and ejected upon infection of the host. We have used high-resolution cryo-electron microscopy to determine structures of the S. aureus bacteriophage 80α portal itself, produced by overexpression, and in situ in the empty and full SaPI1 virions, and show how the portal interacts with the capsid. These structures provide a basis for understanding portal and capsid assembly and the conformational changes that occur upon DNA packaging and ejection.


Assuntos
Ilhas Genômicas , Fagos de Staphylococcus , Staphylococcus aureus , Humanos , Proteínas do Capsídeo/química , Microscopia Crioeletrônica , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/virologia , Fagos de Staphylococcus/genética , Fatores de Virulência/genética , Transdução Genética , Empacotamento do DNA , Conformação de Ácido Nucleico
6.
PLoS Pathog ; 19(6): e1011433, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37289828

RESUMO

Virus host shifts, where a virus transmits to and infects a novel host species, are a major source of emerging infectious disease. Genetic similarity between eukaryotic host species has been shown to be an important determinant of the outcome of virus host shifts, but it is unclear if this is the case for prokaryotes where anti-virus defences can be transmitted by horizontal gene transfer and evolve rapidly. Here, we measure the susceptibility of 64 strains of Staphylococcaceae bacteria (48 strains of Staphylococcus aureus and 16 non-S. aureus species spanning 2 genera) to the bacteriophage ISP, which is currently under investigation for use in phage therapy. Using three methods-plaque assays, optical density (OD) assays, and quantitative (q)PCR-we find that the host phylogeny explains a large proportion of the variation in susceptibility to ISP across the host panel. These patterns were consistent in models of only S. aureus strains and models with a single representative from each Staphylococcaceae species, suggesting that these phylogenetic effects are conserved both within and among host species. We find positive correlations between susceptibility assessed using OD and qPCR and variable correlations between plaque assays and either OD or qPCR, suggesting that plaque assays alone may be inadequate to assess host range. Furthermore, we demonstrate that the phylogenetic relationships between bacterial hosts can generally be used to predict the susceptibility of bacterial strains to phage infection when the susceptibility of closely related hosts is known, although this approach produced large prediction errors in multiple strains where phylogeny was uninformative. Together, our results demonstrate the ability of bacterial host evolutionary relatedness to explain differences in susceptibility to phage infection, with implications for the development of ISP both as a phage therapy treatment and as an experimental system for the study of virus host shifts.


Assuntos
Bacteriófagos , Staphylococcaceae , Fagos de Staphylococcus , Bacteriófagos/fisiologia , Especificidade de Hospedeiro , Filogenia , Reação em Cadeia da Polimerase , Staphylococcaceae/classificação , Staphylococcaceae/virologia , Staphylococcus aureus/virologia , Fagos de Staphylococcus/fisiologia , Ensaio de Placa Viral , Replicação Viral
7.
Genes Genomics ; 45(2): 191-202, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36520268

RESUMO

BACKGROUND: Staphylococcus aureus is a major human pathogen, that can lead to various community- and hospital-acquired infections. RinA is a transcription activator of S. aureus phage φ 11 involved in phage packaging and virulence gene transfer. However, little is known about the molecular mechanism of RinA in the regulation of virulence. OBJECTIVE: We aimed to explore a novel contribution of RinA in the regulation of virulence and provide a new drug target in the treatment of S. aureus infections. METHODS: The specific functions of RinA in S. aureus were analyzed by the methods of growth curve, real-time quantitative PCR (RT-qPCR), subcellular localization, electrophoretic mobility shift assay (EMSA), infection model of Galleria mellonella larvae and the mouse subcutaneous abscess model. RESULTS: In this study, we demonstrated that RinA is a protein evenly distributed in the cytoplasm of S. aureus, and its deletion could cause the growth defects. RT-qPCR and EMSA determined that rinA could negatively regulate the expression of sarA by directly binding to its promoter, and vice versa. The Galleria mellonella larvae infection and mouse subcutaneous abscess models revealed that the rinA mutant strain exhibited obvious virulence defects. When sarA is knocked out, the virulence of S.aureus had no significantly changes whether rinA is knocked out or not. CONCLUSION: Our fndings demonstrated that phage transcription activator RinA regulates S. aureus virulence by governing sarA expression.


Assuntos
Fagos de Staphylococcus , Staphylococcus aureus , Fatores de Transcrição , Proteínas Virais , Fatores de Virulência , Animais , Camundongos , Abscesso , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/virologia , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/metabolismo , Fatores de Transcrição/genética , Proteínas Virais/genética , Virulência/genética , Fatores de Virulência/genética
8.
Microbiol Spectr ; 10(1): e0242321, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35138167

RESUMO

Bacteriophage-mediated transduction of bacterial DNA is a major route of horizontal gene transfer in the human pathogen, Staphylococcus aureus. Transduction involves the packaging of bacterial DNA by viruses and enables the transmission of virulence and resistance genes between cells. To learn more about transduction in S. aureus, we searched a transposon mutant library for genes and mutations that enhanced transfer mediated by the temperate phage, ϕ11. Using a novel screening strategy, we performed multiple rounds of transduction of transposon mutant pools selecting for an antibiotic resistance marker within the transposon element. When determining the locations of transferred mutations, we found that the screen had selected for just 1 or 2 transposon mutant(s) within each pool of 96 mutants. Subsequent analysis showed that the position of the transposon, rather than the inactivation of bacterial genes, was responsible for the phenotype. Interestingly, from multiple rounds, we identified a pattern of transduction that encompassed mobile genetic elements as well as chromosomal regions both upstream and downstream of the phage integration site. The latter was confirmed by DNA sequencing of purified phage lysates. Importantly, transduction frequencies were lower for phage lysates obtained by phage infection rather than induction. Our results confirmed previous reports of lateral transduction of bacterial DNA downstream of the integrated phage but also indicated a novel form of specialized transduction of DNA upstream of the phage. These findings illustrated the complexity of transduction processes and increased our understanding of the mechanisms by which phages transfer bacterial DNA. IMPORTANCE Horizontal transfer of DNA between bacterial cells contributes to the spread of virulence and antibiotic resistance genes in human pathogens. For Staphylococcus aureus, bacterial viruses play a major role in facilitating the horizontal transfer. These viruses, termed bacteriophages, can transfer bacterial DNA between cells by a process known as transduction, which despite its importance is only poorly characterized. Here, we employed a transposon mutant library to investigate transduction in S. aureus. We showed that the genomic location of bacterial DNA relative to where bacteriophages integrated into that bacterial genome affected how frequently that DNA was transduced. Based on serial transduction of transposon mutant pools and direct sequencing of bacterial DNA in bacteriophage particles, we demonstrated both lateral and specialized transduction. The use of mutant libraries to investigate the genomic patterns of bacterial DNA transferred between cells could help us understand how horizontal transfer influences virulence and resistance development.


Assuntos
Fagos de Staphylococcus/genética , Staphylococcus aureus/genética , Staphylococcus aureus/virologia , Transdução Genética , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Transferência Genética Horizontal , Sequências Repetitivas Dispersas , Fagos de Staphylococcus/fisiologia , Staphylococcus aureus/metabolismo
9.
Sci Rep ; 12(1): 2458, 2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35165352

RESUMO

The application of bacteriophages as antibacterial agents has many benefits in the "post-antibiotic age". To increase the number of successfully targeted bacterial strains, phage cocktails, instead of a single phage, are commonly formulated. Nevertheless, there is currently no consensus pipeline for phage cocktail development. Thus, although large cocktails increase the spectrum of activity, they could produce side effects such as the mobilization of virulence or antibiotic resistance genes. On the other hand, coinfection (simultaneous infection of one host cell by several phages) might reduce the potential for bacteria to evolve phage resistance, but some antagonistic interactions amongst phages might be detrimental for the outcome of phage cocktail application. With this in mind, we introduce here a new method, which considers the host range and each individual phage-host interaction, to design the phage mixtures that best suppress the target bacteria while minimizing the number of phages to restrict manufacturing costs. Additionally, putative phage-phage interactions in cocktails and phage-bacteria networks are compared as the understanding of the complex interactions amongst bacteriophages could be critical in the development of realistic phage therapy models in the future.


Assuntos
Bacteriófagos/metabolismo , Escherichia coli/metabolismo , Pseudomonas aeruginosa/metabolismo , Transdução de Sinais/fisiologia , Staphylococcus aureus/metabolismo , Algoritmos , Escherichia coli/virologia , Especificidade de Hospedeiro , Terapia por Fagos/métodos , Pseudomonas aeruginosa/virologia , Staphylococcus aureus/virologia
10.
Appl Environ Microbiol ; 88(3): e0148621, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-34818102

RESUMO

Staphylococcus aureus is an opportunistic pathogen that causes a wide range of infections and food poisoning in humans with antibiotic resistance, specifically to methicillin, compounding the problem. Bacteriophages (phages) provide an alternative treatment strategy, but these only infect a limited number of circulating strains and may quickly become ineffective due to bacterial resistance. To overcome these obstacles, engineered phages have been proposed, but new methods are needed for the efficient transformation of large DNA molecules into S. aureus to "boot-up" (i.e., rescue) infectious phages. We presented a new, efficient, and reproducible DNA transformation method, NEST (non-electroporation Staphylococcus transformation), for S. aureus to boot-up purified phage genomic DNA (at least 150 kb in length) and whole yeast-assembled synthetic phage genomes. This method was a powerful new tool for the transformation of DNA in S. aureus and will enable the rapid development of engineered therapeutic phages and phage cocktails against Gram-positive pathogens. IMPORTANCE The continued emergence of antibiotic-resistant bacterial pathogens has heightened the urgency for alternative antibacterial strategies. Phages provide an alternative treatment strategy but are difficult to optimize. Synthetic biology approaches have been successfully used to construct and rescue genomes of model phages but only in a limited number of highly transformable host species. In this study, we used a new, reproducible, and efficient transformation method to reconstitute a functional nonmodel Siphophage from a constructed synthetic genome. This method will facilitate the engineering of Staphylococcus and Enterococcus phages for therapeutic applications and the engineering of Staphylococcus strains by enabling transformation of higher molecular weight DNA to introduce more complex modifications.


Assuntos
Fagos de Staphylococcus , Staphylococcus aureus , DNA Viral/genética , Humanos , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/virologia , Fagos de Staphylococcus/genética , Staphylococcus aureus/virologia
11.
Sci Rep ; 11(1): 24394, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34937862

RESUMO

Staphylococcus aureus can be a harmless coloniser, but it can also cause severe infections in humans, livestock and wildlife. Regarding the latter, only few studies have been performed and knowledge on virulence factors is insufficient. The aim of the present study was to study S. aureus isolates from deceased wild beavers (Castor fiber). Seventeen isolates from eleven beavers, found in Germany and Austria, were investigated. Antimicrobial and biocide susceptibility tests were performed. Isolates were characterised using S. aureus-specific DNA microarrays, spa typing and whole-genome sequencing. From two isolates, prophages were induced by mitomycin C and studied by transmission electron microscopy. Four isolates belonged to clonal complex (CC) 8, CC12, and CC398. Twelve isolates belonged to CC1956 and one isolate was CC49. The CC49 and CC1956 isolates carried distinct lukF/S genes related to the Panton-Valentine leukocidin (PVL) from human isolates of S. aureus. These genes were located on related, but not identical, Siphovirus prophages. The beavers, from which those isolates originated, suffered from abscesses, purulent organ lesions and necrotising pneumonia, i.e., clinical manifestations resembling symptoms of severe PVL-associated disease in humans. It might thus be assumed that the "Beaver Leukocidin (BVL, lukF/S-BV)"-positive strains are beaver-specific pathogens, and further studies on their clinical role as well as on a possible transmissibility to other species, including humans, are warranted.


Assuntos
Toxinas Bacterianas/análise , Exotoxinas/análise , Leucocidinas/análise , Roedores/microbiologia , Infecções Estafilocócicas/microbiologia , Fagos de Staphylococcus/isolamento & purificação , Staphylococcus aureus/isolamento & purificação , Staphylococcus aureus/virologia , Animais , Toxinas Bacterianas/genética , Técnicas de Tipagem Bacteriana , Exotoxinas/genética , Genes Bacterianos , Genes Virais , Humanos , Leucocidinas/genética , Infecções Estafilocócicas/veterinária , Fagos de Staphylococcus/genética , Staphylococcus aureus/genética
12.
Microb Genom ; 7(12)2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34907894

RESUMO

Prophages affect bacterial fitness on multiple levels. These include bacterial infectivity, toxin secretion, virulence regulation, surface modification, immune stimulation and evasion and microbiome competition. Lysogenic conversion arms bacteria with novel accessory functions thereby increasing bacterial fitness, host adaptation and persistence, and antibiotic resistance. These properties allow the bacteria to occupy a niche long term and can contribute to chronic infections and inflammation such as chronic rhinosinusitis (CRS). In this study, we aimed to identify and characterize prophages present in Staphylococcus aureus from patients suffering from CRS in relation to CRS disease phenotype and severity. Prophage regions were identified using PHASTER. Various in silico tools like ResFinder and VF Analyzer were used to detect virulence genes and antibiotic resistance genes respectively. Progressive MAUVE and maximum likelihood were used for multiple sequence alignment and phylogenetics of prophages respectively. Disease severity of CRS patients was measured using computed tomography Lund-Mackay scores. Fifty-eight S. aureus clinical isolates (CIs) were obtained from 28 CRS patients without nasal polyp (CRSsNP) and 30 CRS patients with nasal polyp (CRSwNP). All CIs carried at least one prophage (average=3.6) and prophages contributed up to 7.7 % of the bacterial genome. Phage integrase genes were found in 55/58 (~95 %) S. aureus strains and 97/211 (~46 %) prophages. Prophages belonging to Sa3int integrase group (phiNM3, JS01, phiN315) (39/97, 40%) and Sa2int (phi2958PVL) (14/97, 14%) were the most prevalent prophages and harboured multiple virulence genes such as sak, scn, chp, lukE/D, sea. Intact prophages were more frequently identified in CRSwNP than in CRSsNP (P=0.0021). Intact prophages belonging to the Sa3int group were more frequent in CRSwNP than in CRSsNP (P=0.0008) and intact phiNM3 were exclusively found in CRSwNP patients (P=0.007). Our results expand the knowledge of prophages in S. aureus isolated from CRS patients and their possible role in disease development. These findings provide a platform for future investigations into potential tripartite associations between bacteria-prophage-human immune system, S. aureus evolution and CRS disease pathophysiology.


Assuntos
Pólipos Nasais/microbiologia , Prófagos/genética , Rinite/microbiologia , Sinusite/microbiologia , Staphylococcus aureus/virologia , Composição de Bases , Farmacorresistência Bacteriana , Tamanho do Genoma , Genoma Bacteriano , Humanos , Evasão da Resposta Imune , Pólipos Nasais/diagnóstico por imagem , Filogenia , Rinite/diagnóstico por imagem , Índice de Gravidade de Doença , Sinusite/diagnóstico por imagem , Staphylococcus aureus/classificação , Staphylococcus aureus/genética , Tomografia Computadorizada por Raios X , Fatores de Virulência/genética
13.
Microbiol Spectr ; 9(3): e0173621, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34908439

RESUMO

Although several studies have shown promising clinical outcomes of phage therapy in patients with orthopedic device-related infections, questions remain regarding the optimal application protocol, systemic effects, and the impact of the immune response. This study provides a proof-of-concept of phage therapy in a clinically relevant rabbit model of fracture-related infection (FRI) caused by Staphylococcus aureus. In a prevention setting, phage in saline (without any biomaterial-based carrier) was highly effective in the prevention of FRI, compared to systemic antibiotic prophylaxis alone. In the subsequent study involving treatment of established infection, daily administration of phage in saline through a subcutaneous access tube was compared to a single intraoperative application of a phage-loaded hydrogel and a control group receiving antibiotics only. In this setting, although a possible trend of bacterial load reduction on the implant was observed with the phage-loaded hydrogel, no superior effect of phage therapy was found compared to antibiotic treatment alone. The application of phage in saline through a subcutaneous access tube was, however, complicated by superinfection and the development of neutralizing antibodies. The latter was not found in the animals that received the phage-loaded hydrogel, which may indicate that encapsulation of phages into a carrier such as a hydrogel limits their exposure to the adaptive immune system. These studies show phage therapy can be useful in targeting orthopedic device-related infection, however, further research and improvements of these application methods are required for this complex clinical setting. IMPORTANCE Because of the growing spread of antimicrobial resistance, the use of alternative prevention and treatment strategies is gaining interest. Although the therapeutic potential of bacteriophages has been demonstrated in a number of case reports and series over the past decade, many unanswered questions remain regarding the optimal application protocol. Furthermore, a major concern during phage therapy is the induction of phage neutralizing antibodies. This study aimed at providing a proof-of-concept of phage therapy in a clinically relevant rabbit model of fracture-related infection caused by Staphylococcus aureus. Phage therapy was applied as prophylaxis in a first phase, and as treatment of an established infection in a second phase. The development of phage neutralizing antibodies was evaluated in the treatment study. This study demonstrates that phage therapy can be useful in targeting orthopedic device-related infection, especially as prophylaxis; however, further research and improvements of these application methods are required.


Assuntos
Antibacterianos/uso terapêutico , Fraturas Ósseas/microbiologia , Terapia por Fagos/métodos , Infecções Relacionadas à Prótese/terapia , Infecções Estafilocócicas/terapia , Fagos de Staphylococcus/crescimento & desenvolvimento , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Farmacorresistência Bacteriana/genética , Feminino , Fraturas Ósseas/patologia , Hidrogéis/uso terapêutico , Estudo de Prova de Conceito , Infecções Relacionadas à Prótese/microbiologia , Coelhos , Infecções Estafilocócicas/prevenção & controle , Fagos de Staphylococcus/imunologia , Staphylococcus aureus/virologia
14.
Sci Rep ; 11(1): 22951, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34824363

RESUMO

As antibiotic resistance is being a threat to public health worldwide, bacteriophages are re-highlighted as alternative antimicrobials to fight with pathogens. Various wild-type phages isolated from diverse sources have been tested, but potential mutant phages generated by genome engineering or random mutagenesis are drawing increasing attention. Here, we applied a chelating agent, sodium pyrophosphate, to the staphylococcal temperate Siphoviridae phage SA3821 to introduce random mutations. Through 30 sequential sodium pyrophosphate challenges and random selections, the suspected mutant phage SA3821M was isolated. SA3821M maintained an intact virion morphology, but exhibited better bactericidal activity against its host Staphylococcous aureus CCARM 3821 for up to 17 h and thermostability than its parent, SA3821. Sodium pyrophosphate-mediated mutations in SA3821M were absent in lysogenic development genes but concentrated (83.9%) in genes related to the phage tail, particularly in the tail tape measure protein, indicating that changes in the tail module might have been responsible for the altered traits. This intentional random mutagenesis through controlled treatments with sodium pyrophosphate could be applied to other phages as a simple but potent method to improve their traits as alternative antimicrobials.


Assuntos
Difosfatos , Mutagênese , Fagos de Staphylococcus/genética , Antibacterianos , Genoma Viral , Especificidade de Hospedeiro , Infecções Estafilocócicas/terapia , Staphylococcus aureus/virologia
15.
Genes (Basel) ; 12(11)2021 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-34828356

RESUMO

Among clinically relevant lineages of Staphylococcus aureus, the lineage or clonal complex 398 (CC398) is of particular interest. Strains from this lineage were only described as livestock colonizers until 2007. Progressively, cases of infection were reported in humans in contact with farm animals, and now, CC398 isolates are increasingly identified as the cause of severe infections even in patients without any contact with animals. These observations suggest that CC398 isolates have spread not only in the community but also in the hospital setting. In addition, several recent studies have reported that CC398 strains are evolving towards increased virulence and antibiotic resistance. Identification of the origin and emergence of this clonal complex could probably benefit future large-scale studies that aim to detect sources of contamination and infection. Current evidence indicates that the evolution of CC398 strains towards these phenotypes has been driven by the acquisition of prophages and other mobile genetic elements. In this short review, we summarize the main knowledge of this major lineage of S. aureus that has become predominant in the human clinic worldwide within a single decade.


Assuntos
DNA Bacteriano/genética , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/classificação , Animais , Animais Domésticos/microbiologia , Evolução Molecular , Humanos , Sequências Repetitivas Dispersas , Vigilância da População , Prófagos/genética , Infecções Estafilocócicas/veterinária , Staphylococcus aureus/genética , Staphylococcus aureus/virologia
16.
mBio ; 12(6): e0208321, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34781740

RESUMO

The antibacterial drone (ABD) system is based on repurposing the phage-inducible staphylococcal pathogenicity islands (SaPIs) for use as antibacterial agents that are indifferent to antibiotic resistance. The ABDs were constructed by inserting tetM for tetracycline resistance (Tcr) selection, replacing the SaPI virulence genes with bactericidal or bacteriostatic genes such as CRISPR/cas9/agrA, whose expression kills by double-strand cleavage of agrA, or CRISPR/dcas9/agrP2P3, whose expression blocks the target organism's virulence. ABD DNA is packaged in phage-like particles that attack their staphylococcal targets in vivo as well as in vitro. We determine ABD titers by transfer frequency, enumerate surviving cells as a function of multiplicity, and analyze the fate of ABD DNA with green fluorescent protein. An initial study revealed surprisingly that many more cells were killed by the ABD than were measured by transduction. Our study of this phenomenon has revealed several important features of the ABD system: (i) a significant number of entering ABD DNA molecules do not go on to establish stable transductants (i.e., are abortive); (ii) ABD cargo genes are expressed immediately following entry, even by the abortive ABDs; (iii) immediate plating on Tc-containing agar seriously underestimates particle numbers, partly owing to Tc inhibition of protein synthesis; (iv) replacement of tetM with cadA (conferring resistance to CdCl2) provides more accurate particle enumeration; (v) ABDs expressing CRISPR/cas9/agrA kill ∼99.99% of infected cells and provide the most accurate measurement of particle numbers as well as proof of principle for the system; and (vi) surprisingly, TetM interferes with stable establishment of ABD DNA independently of Tcr. IMPORTANCE For a particulate therapeutic agent, such as the ABD, accurate enumeration of particles is critical to enable evaluation of preparative procedures and calculation of therapeutic dosages. It is equally important that a selective marker used for these two purposes be biologically inert. We have long used tetM for these purposes but show here that tetM not only underestimates particle titers, by over 20-fold in some experiments, but also seriously impedes stable establishment of the therapeutic particle DNA. Given that tetM is a very convenient and widely used selective marker, publication of these findings is of considerable importance to the microbiological community as well as an interesting illustration of the unpredictable biological effects of genes taken out of their native context.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana , Fagos de Staphylococcus/fisiologia , Staphylococcus aureus/genética , Staphylococcus aureus/virologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sistemas CRISPR-Cas , Ilhas Genômicas , Infecções Estafilocócicas/microbiologia , Fagos de Staphylococcus/genética , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/metabolismo , Tetraciclina/farmacologia
17.
Microbiologyopen ; 10(5): e1245, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34713608

RESUMO

Temperate phages are bacterial viruses that after infection either reside integrated into a bacterial genome as prophages forming lysogens or multiply in a lytic lifecycle. The decision between lifestyles is determined by a switch involving a phage-encoded repressor, CI, and a promoter region from which lytic and lysogenic genes are divergently transcribed. Here, we investigate the switch of phage ɸ13 from the human pathogen Staphylococcus aureus. ɸ13 encodes several virulence factors and is prevalent in S. aureus strains colonizing humans. We show that the ɸ13 switch harbors a cI gene, a predicted mor (modulator of repression) gene, and three high-affinity operator sites binding CI. To quantify the decision between lytic and lysogenic lifestyle, we introduced reporter plasmids that carry the 1.3 kb switch region from ɸ13 with the lytic promoter fused to lacZ into S. aureus and Bacillus subtilis. Analysis of ß-galactosidase expression indicated that decision frequency is independent of host factors. The white "lysogenic" phenotype, which relies on the expression of cI, could be switched to a stable blue "lytic" phenotype by DNA damaging agents. We have characterized lifestyle decisions of phage ɸ13, and our approach may be applied to other temperate phages encoding virulence factors in S. aureus.


Assuntos
Bacteriólise , Lisogenia , Proteínas Repressoras/genética , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/fisiologia , Staphylococcus aureus/virologia , Proteínas Virais/genética , Proteínas Virais Reguladoras e Acessórias/genética , Toxinas Bacterianas/genética , Dano ao DNA , DNA Intergênico , Exotoxinas/genética , Genes Virais , Humanos , Leucocidinas/genética , Regiões Operadoras Genéticas , Fenótipo , Plasmídeos , Prófagos/fisiologia , Proteínas Repressoras/metabolismo , Staphylococcus aureus/crescimento & desenvolvimento , Proteínas Virais/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Fatores de Virulência/genética
18.
Sci Rep ; 11(1): 18994, 2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34556764

RESUMO

Staphylococcus aureus is a major human and animal pathogen although the animal-associated S. aureus can be a potential risk of human zoonoses. Acquisition of phage-related genomic islands determines the S. aureus species diversity. This study characterized and compared the genome architecture, distribution nature, and evolutionary relationship of 65 complete prophages carried by human and animal-associated S. aureus strains spreading across the European regions. The analyzed prophage genomes showed mosaic architecture with extensive variation in genome size. The phylogenetic analyses generated seven clades in which prophages of the animal-associated S. aureus scattered in all the clades. The S. aureus strains with the same SCCmec type, and clonal complex favored the harboring of similar prophage sequences and suggested that the frequency of phage-mediated horizontal gene transfer is higher between them. The presence of various virulence factors in prophages of animal-associated S. aureus suggested that these prophages could have more pathogenic potential than prophages of human-associated S. aureus. This study showed that the S. aureus phages are dispersed among the several S. aureus serotypes and around the European regions. Further, understanding the phage functional genomics is necessary for the phage-host interactions and could be used for tracing the S. aureus strains transmission.


Assuntos
Prófagos/genética , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade , Zoonoses/microbiologia , Animais , Europa (Continente) , Transferência Genética Horizontal , Genoma Bacteriano , Ilhas Genômicas , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Filogenia , Especificidade da Espécie , Infecções Estafilocócicas/transmissão , Staphylococcus aureus/virologia , Fatores de Virulência/genética , Zoonoses/transmissão
19.
Nat Microbiol ; 6(10): 1300-1308, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34518655

RESUMO

Staphylococcal pathogenicity islands (SaPIs) are a family of closely related mobile chromosomal islands that encode and disseminate the superantigen toxins, toxic shock syndrome toxin 1 and superantigen enterotoxin B (SEB). They are regulated by master repressors, which are counteracted by helper phage-encoded proteins, thereby inducing their excision, replication, packaging and intercell transfer. SaPIs are major components of the staphylococcal mobilome, occupying five chromosomal att sites, with many strains harbouring two or more. As regulatory interactions between co-resident SaPIs could have profound effects on the spread of superantigen pathobiology, we initiated the current study to search for such interactions. Using classical genetics, we found that, with one exception, their regulatory systems do not cross-react. The exception was SaPI3, which was originally considered defective because it could not be mobilized by any known helper phage. We show here that SaPI3 has an atypical regulatory module and is induced not by a phage but by many other SaPIs, including SaPI2, SaPIbov1 and SaPIn1, each encoding a conserved protein, Sis, which counteracts the SaPI3 repressor, generating an intracellular regulatory cascade: the co-resident SaPI, when conventionally induced by a helper phage, expresses its sis gene which, in turn, induces SaPI3, enabling it to spread. Using bioinformatics analysis, we have identified more than 30 closely related coancestral SEB-encoding SaPI3 relatives occupying the same att site and controlled by a conserved regulatory module, immA-immR-str'. This module is functionally analogous but unrelated to the typical SaPI regulatory module, stl-str. As SaPIs are phage satellites, SaPI3 and its relatives are SaPI satellites.


Assuntos
Ilhas Genômicas/genética , Staphylococcus aureus/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Replicação do DNA , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Transferência Genética Horizontal , Fagos de Staphylococcus/fisiologia , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/virologia , Ativação Transcricional
20.
J Microbiol ; 59(9): 840-847, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34383247

RESUMO

Endolysin, a peptidoglycan hydrolase derived from bacteriophage, has been suggested as an alternative antimicrobial agent. Many endolysins on staphylococcal phages have been identified and applied extensively against Staphylococcus spp. Among them, LysK-like endolysin, a well-studied staphylococcal endolysin, accounts for most of the identified endolysins. However, relatively little interest has been paid to LysKunlike endolysin and a few of them has been characterized. An endolysin LysSAP33 encoded on bacteriophage SAP33 shared low homology with LysK-like endolysin in sequence by 41% and domain composition (CHAP-unknown CBD). A green fluorescence assay using a fusion protein for LysSAP33_CBD indicated that the CBD domain (157-251 aa) was bound to the peptidoglycan of S. aureus. The deletion of LysSAP33_CBD at the C-terminal region resulted in a significant decrease in lytic activity and efficacy. Compared to LysK-like endolysin, LysSAP33 retained its lytic activity in a broader range of temperature, pH, and NaCl concentrations. In addition, it showed a higher activity against biofilms than LysK-like endolysin. This study could be a helpful tool to develop our understanding of staphylococcal endolysins not belonging to LysK-like endolysins and a potential biocontrol agent against biofilms.


Assuntos
Endopeptidases/metabolismo , Fagos de Staphylococcus/enzimologia , Staphylococcus aureus/virologia , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Parede Celular/metabolismo , Parede Celular/virologia , Endopeptidases/química , Endopeptidases/genética , Peptidoglicano/metabolismo , Alinhamento de Sequência , Fagos de Staphylococcus/química , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/fisiologia , Staphylococcus aureus/metabolismo , Proteínas Virais/química , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA