Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
Viruses ; 14(11)2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36366569

RESUMO

Staphylococcus aureus asymptomatically colonizes the nasal cavity of mammals, but it is also a leading cause of life-threatening infections. Most human nasal isolates carry Sa3 phages, which integrate into the bacterial hlb gene encoding a sphingomyelinase. The virulence factor-encoding genes carried by the Sa3-phages are highly human-specific, and most animal strains are Sa3 negative. Thus, both insertion and excision of the prophage could potentially confer a fitness advantage to S. aureus. Here, we analyzed the phage life cycle of two Sa3 phages, Φ13 and ΦN315, in different phage-cured S. aureus strains. Based on phage transfer experiments, strains could be classified into low (8325-4, SH1000, and USA300c) and high (MW2c and Newman-c) transfer strains. High-transfer strains promoted the replication of phages, whereas phage adsorption, integration, excision, or recA transcription was not significantly different between strains. RNASeq analyses of replication-deficient lysogens revealed no strain-specific differences in the CI/Mor regulatory switch. However, lytic genes were significantly upregulated in the high transfer strain MW2c Φ13 compared to strain 8325-4 Φ13. By transcriptional start site prediction, new promoter regions within the lytic modules were identified, which are likely targeted by specific host factors. Such host-phage interaction probably accounts for the strain-specific differences in phage replication and transfer frequency. Thus, the genetic makeup of the host strains may determine the rate of phage mobilization, a feature that might impact the speed at which certain strains can achieve host adaptation.


Assuntos
Toxinas Bacterianas , Infecções Estafilocócicas , Animais , Humanos , Staphylococcus aureus , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Infecções Estafilocócicas/microbiologia , Estágios do Ciclo de Vida , Mamíferos
3.
Microbiol Spectr ; 10(1): e0146621, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35196798

RESUMO

Bacterial pathogens are progressively adapting to current antimicrobial therapies with severe consequences for patients and global health care systems. This is critically underscored by the rise of methicillin resistant Staphylococcus aureus (MRSA) and other biofilm-forming staphylococci. Accordingly, alternative strategies have been explored to fight such highly multidrug resistant microorganisms, including antimicrobial photodynamic therapy (aPDT) and phage therapy. aPDT has the great advantage that it does not elicit resistance, while phage therapy allows targeting of specific pathogens. In the present study, we aimed to merge these benefits by conjugating the cell-binding domain (CBD3) of a Staphylococcus aureus phage endolysin to a photoactivatable silicon phthalocyanine (IRDye 700DX) for the development of a Staphylococcus-targeted aPDT approach. We show that, upon red-light activation, the resulting CBD3-700DX conjugate generates reactive oxygen species that effectively kill high loads of planktonic and biofilm-resident staphylococci, including MRSA. Furthermore, CBD3-700DX is readily internalized by mammalian cells, where it allows the targeted killing of intracellular MRSA upon photoactivation. Intriguingly, aPDT with CBD3-700DX also affects mammalian cells with internalized MRSA, but it has no detectable side effects on uninfected cells. Altogether, we conclude that CBD3 represents an attractive targeting agent for Staphylococcus-specific aPDT, irrespective of planktonic, biofilm-embedded, or intracellular states of the bacterium. IMPORTANCE Antimicrobial resistance is among the biggest threats to mankind today. There are two alternative antimicrobial therapies that may help to control multidrug-resistant bacteria. In phage therapy, natural antagonists of bacteria, lytic phages, are harnessed to fight pathogens. In antimicrobial photodynamic therapy (aPDT), a photosensitizer, molecular oxygen, and light are used to produce reactive oxygen species (ROS) that inflict lethal damage on pathogens. Since aPDT destroys multiple essential components in targeted pathogens, aPDT resistance is unlikely. However, the challenge in aPDT is to maximize target specificity and minimize collateral oxidative damage to host cells. We now present an antimicrobial approach that combines the best features of both alternative therapies, namely, the high target specificity of phages and the efficacy of aPDT. This is achieved by conjugating the specific cell-binding domain from a phage protein to a near-infrared photosensitizer. aPDT with the resulting conjugate shows high target specificity toward MRSA with minimal side effects.


Assuntos
Antibacterianos/farmacologia , Endopeptidases/farmacologia , Fotoquimioterapia , Infecções Estafilocócicas/microbiologia , Fagos de Staphylococcus/química , Staphylococcus/efeitos dos fármacos , Staphylococcus/fisiologia , Animais , Antibacterianos/química , Biofilmes/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla , Endopeptidases/química , Endopeptidases/metabolismo , Humanos , Indóis/química , Luz , Compostos de Organossilício/química , Fármacos Fotossensibilizantes/química , Espécies Reativas de Oxigênio/metabolismo , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus/virologia , Fagos de Staphylococcus/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33649203

RESUMO

In response to increasing frequencies of antibiotic-resistant pathogens, there has been a resurrection of interest in the use of bacteriophage to treat bacterial infections: phage therapy. Here we explore the potential of a seemingly ideal phage, PYOSa, for combination phage and antibiotic treatment of Staphylococcus aureus infections. This K-like phage has a broad host range; all 83 tested clinical isolates of S.aureus tested were susceptible to PYOSa Because of the mode of action of PYOSa, S. aureus is unlikely to generate classical receptor-site mutants resistant to PYOSa; none were observed in the 13 clinical isolates tested. PYOSa kills S. aureus at high rates. On the downside, the results of our experiments and tests of the joint action of PYOSa and antibiotics raise issues that must be addressed before PYOSa is employed clinically. Despite the maintenance of the phage, PYOSa does not clear populations of S. aureus Due to the ascent of a phenotyically diverse array of small-colony variants following an initial demise, the bacterial populations return to densities similar to that of phage-free controls. Using a combination of mathematical modeling and in vitro experiments, we postulate and present evidence for a mechanism to account for the demise-resurrection dynamics of PYOSa and S. aureus Critically for phage therapy, our experimental results suggest that treatment with PYOSa followed by bactericidal antibiotics can clear populations of S. aureus more effectively than the antibiotics alone.


Assuntos
Antibacterianos/farmacologia , Terapia por Fagos , Infecções Estafilocócicas , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/terapia , Infecções Estafilocócicas/virologia , Staphylococcus aureus/metabolismo , Staphylococcus aureus/virologia
5.
Microb Cell Fact ; 20(1): 56, 2021 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-33653327

RESUMO

BACKGROUND: Antibiotic-resistant bacteria have emerged as a serious problem; bacteriophages have, therefore, been proposed as a therapeutic alternative to antibiotics. Several authorities, such as pharmacopeia, FDA, have confirmed their safety, and some bacteriophages are commercially available worldwide. The demand for bacteriophages is expected to increase exponentially in the future; hence, there is an urgent need to mass-produce bacteriophages economically. Unlike the replication of non-lytic bacteriophages, lytic bacteriophages are replicated by lysing host bacteria, which leads to the termination of phage production; hence, strategies that can prolong the lysis of host bacteria in bacteria-bacteriophage co-cultures, are required. RESULTS: In the current study, we manipulated the inoculum concentrations of Staphylococcus aureus and phage pSa-3 (multiplicity of infection, MOI), and their energy sources to delay the bactericidal effect while optimizing phage production. We examined an increasing range of bacterial inoculum concentration (2 × 108 to 2 × 109 CFU/mL) to decrease the lag phase, in combination with a decreasing range of phage inoculum (from MOI 0.01 to 0.00000001) to delay the lysis of the host. Bacterial concentration of 2 × 108 CFU/mL and phage MOI of 0.0001 showed the maximum final phage production rate (1.68 × 1010 plaque forming unit (PFU)/mL). With this combination of phage-bacteria inoculum, we selected glycerol, glycine, and calcium as carbon, nitrogen, and divalent ion sources, respectively, for phage production. After optimization using response surface methodology, the final concentration of the lytic Staphylococcus phage was 8.63 × 1010 ± 9.71 × 109 PFU/mL (5.13-fold increase). CONCLUSIONS: Therefore, Staphylococcus phage pSa-3 production can be maximized by increasing the bacterial inoculum and reducing the seeding phage MOI, and this combinatorial strategy could decrease the phage production time. Further, we suggest that response surface methodology has the potential for optimizing the mass production of lytic bacteriophages.


Assuntos
Infecções Estafilocócicas/metabolismo , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/metabolismo , Propriedades de Superfície
6.
Viruses ; 12(5)2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32443723

RESUMO

Bacteriophage 80α is a representative of a class of temperate phages that infect Staphylococcus aureus and other Gram-positive bacteria. Many of these phages carry genes encoding toxins and other virulence factors. This phage, 80α, is also involved in high-frequency mobilization of S. aureus pathogenicity islands (SaPIs), mobile genetic elements that carry virulence factor genes. Bacteriophage 80α encodes a minor capsid protein, gp44, between the genes for the portal protein and major capsid protein. Gp44 is essential for a productive infection by 80α but not for transduction of SaPIs or plasmids. We previously demonstrated that gp44 is an ejection protein that acts to promote progression to the lytic cycle upon infection and suggested that the protein might act as an anti-repressor of CI in the lytic-lysogenic switch. However, an 80α Δ44 mutant also exhibited a reduced rate of lysogeny. Here, we show that gp44 is a non-specific DNA binding protein with affinity for the blunt ends of linear DNA. Our data suggest a model in which gp44 promotes circularization of the genome after injection into the host cell, a key initial step both for lytic growth and for the establishment of lysogeny.


Assuntos
Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/virologia , Proteínas Virais/metabolismo , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Caudovirales/genética , DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ilhas Genômicas/genética , Sequências Repetitivas Dispersas , Lisogenia , Fagos de Staphylococcus/genética , Proteínas Virais/química , Proteínas Virais/genética , Fatores de Virulência/genética
7.
Sci Rep ; 10(1): 6260, 2020 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-32277078

RESUMO

Bloodstream infections (BSIs) are considered a major cause of death worldwide. Staphylococcus spp. are one of the most BSIs prevalent bacteria, classified as high priority due to the increasing multidrug resistant strains. Thus, a fast, specific and sensitive method for detection of these pathogens is of extreme importance. In this study, we have designed a novel assay for detection of Staphylococcus in blood culture samples, which combines the advantages of a phage endolysin cell wall binding domain (CBD) as a specific probe with the accuracy and high-throughput of flow cytometry techniques. In order to select the biorecognition molecule, three different truncations of the C-terminus of Staphylococcus phage endolysin E-LM12, namely the amidase (AMI), SH3 and amidase+SH3 (AMI_SH3) were cloned fused with a green fluorescent protein. From these, a higher binding efficiency to Staphylococcus cells was observed for AMI_SH3, indicating that the amidase domain possibly contributes to a more efficient binding of the SH3 domain. The novel phage endolysin-based flow cytometry assay provided highly reliable and specific detection of 1-5 CFU of Staphylococcus in 10 mL of spiked blood, after 16 hours of enrichment culture. Overall, the method developed herein presents advantages over the standard BSIs diagnostic methods, potentially contributing to an early and effective treatment of BSIs.


Assuntos
Bacteriemia/diagnóstico , Citometria de Fluxo/métodos , Infecções Estafilocócicas/diagnóstico , Staphylococcus/isolamento & purificação , Bacteriemia/microbiologia , Proteínas de Bactérias/metabolismo , Limite de Detecção , Sondas Moleculares/metabolismo , Domínios Proteicos , Reprodutibilidade dos Testes , Infecções Estafilocócicas/microbiologia , Staphylococcus/metabolismo , Fagos de Staphylococcus/metabolismo , Proteínas Virais/metabolismo
8.
Appl Microbiol Biotechnol ; 104(11): 5145-5158, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32248441

RESUMO

Staphylococcus aureus is a notorious foodborne pathogen since it has ability to produce variety of toxins including heat-stable enterotoxin, form biofilm, and acquire resistance to antibiotics. Biocontrol of foodborne pathogens by lytic bacteriophages garners increasing interest from both researchers and food industry. In the present study, 29 phages against S. aureus were successfully isolated from chicken, pork, and fish. Characterization of the isolates revealed that phage SA46-CTH2 belonging to Podoviridae family had a number of features suitable for food industry applications such as wide host range, short latent period, large burst size, high stress tolerance, and a genome free of virulence genes. Furthermore, phage SA46-CTH2 alone or in combination with nisin exhibited great efficacy in reducing planktonic and biofilm cells of S. aureus at various conditions tested. The combination of phage SA46-CTH2 and nisin was also found to be able to inhibit the regrowth of S. aureus at both 37 and 24 °C.Key points• A total of 29 S. aureus phages were successfully isolated from fish, pork, and chicken products. • Phage SA46-CTH2 was characterized by host range, morphology, and genome sequencing. • SA46-CTH2 significantly reduced both planktonic and biofilm cells of S. aureus. • Combination of SA46-CTH2 and nisin inhibited the regrowth of S. aureus.


Assuntos
Microbiologia de Alimentos/métodos , Podoviridae/metabolismo , Fagos de Staphylococcus/isolamento & purificação , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Animais , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Galinhas/virologia , Peixes/virologia , Genoma Viral , Especificidade de Hospedeiro , Nisina/farmacologia , Podoviridae/genética , Podoviridae/isolamento & purificação , Carne de Porco/virologia , Fagos de Staphylococcus/genética , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/virologia , Virulência/efeitos dos fármacos
9.
Viruses ; 11(9)2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31443379

RESUMO

Impetigo is a contagious skin infection predominantly caused by Staphylococcus aureus. Decontamination of S. aureus from the skin is becoming more difficult because of the emergence of antibiotic-resistant strains. Bacteriophage endolysins are less likely to invoke resistance and can eliminate the target bacteria without disturbance of the normal microflora. In this study, we investigated the therapeutic potential of a recombinant endolysin derived from kayvirus S25-3 against staphylococcal impetigo in an experimental setting. First, the recombinant S25-3 endolysin required an incubation period of over 15 minutes to exhibit efficient bactericidal effects against S. aureus. Second, topical application of the recombinant S25-3 endolysin decreased the number of intraepidermal staphylococci and the size of pustules in an experimental mouse model of impetigo. Third, treatment with the recombinant S25-3 endolysin increased the diversity of the skin microbiota in the same mice. Finally, we revealed the genus-specific bacteriolytic effect of recombinant S25-3 endolysin against staphylococci, particularly S. aureus, among human skin commensal bacteria. Therefore, topical treatment with recombinant S25-3 endolysin can be a promising disease management procedure for staphylococcal impetigo by efficient bacteriolysis of S. aureus while improving the cutaneous bacterial microflora.


Assuntos
Caudovirales/metabolismo , Endopeptidases/farmacologia , Impetigo/tratamento farmacológico , Staphylococcus aureus , Administração Cutânea , Animais , Antibacterianos/administração & dosagem , Antibacterianos/farmacologia , Bacteriólise , Caudovirales/patogenicidade , Endopeptidases/administração & dosagem , Endopeptidases/genética , Genes Bacterianos , Genes Virais , Impetigo/microbiologia , Metagenômica , Camundongos , Microbiota/genética , Pseudomonas aeruginosa/virologia , RNA Ribossômico 16S , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Pele/microbiologia , Pele/patologia , Infecções Estafilocócicas/tratamento farmacológico , Fagos de Staphylococcus/metabolismo , Fagos de Staphylococcus/patogenicidade , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/virologia , Staphylococcus epidermidis/virologia , Streptococcus mitis/virologia
10.
Int J Biol Macromol ; 124: 903-914, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30517842

RESUMO

The lysogenic growth of phage ф11 in Staphylococcus aureus is controlled by a repressor (CI) that harbors an N-terminal domain (NTD), and a C-terminal domain (CTD). Previously, NTD, like CI, showed DNA binding activity and dimerized in the aqueous solution. To precisely understand the folding mechanism, function, and the stability of CI, NTD, and CTD, we have investigated their recombinant forms, rCI, rNTD, and rCTD, using various probes. The data reveal that rCTD, like rCI and rNTD, is a well-structured protein and produces dimers in the aqueous environment. However, the stability order of the dimers appears to be rCI > rCTD > rNTD. Interestingly, the stability of rNTD or rCTD looks slightly higher than that of rCI. The urea-induced equilibrium unfolding of these proteins proceeded via the production of two intermediates. The structure, surface hydrophobicity, and the dimeric status of one intermediate mostly differed from those of another intermediate or the native protein. Our MD simulation study on the representative NTD shows the substantial change in its structure and stability at the urea concentrations, which formed rNTD intermediates. Collectively, the computational data have supported the experimental data and indicated that the CI and its domains are folded by a similar multiphasic pathway.


Assuntos
Proteínas de Bactérias/química , Proteínas Repressoras/química , Fagos de Staphylococcus/genética , Proteínas Virais/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Cinética , Lisogenia , Modelos Moleculares , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/virologia , Especificidade por Substrato , Termodinâmica , Proteínas Virais/genética , Proteínas Virais/metabolismo
11.
PLoS One ; 13(10): e0205728, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30308048

RESUMO

Bacteriophages have been proven as effective antimicrobial agents in the treatment of infectious diseases and in other biocontrol applications including food preservation and disinfection. The extensive use of bacteriophages requires improved methodologies for medium- and long-term storage as well as for easy shipping. To this aim, we have determined the stability of four Staphylococcus phages (phiIPLA88, phiIPLA35, phiIPLA-RODI and phiIPLA-C1C) with antimicrobial potential at different temperatures (20°C/25°C, 4°C, -20°C, -80°C, -196°C) and during lyophilization (freeze drying) using several stabilizing additives (disaccharides, glycerol, sorbitol and skim milk). Differences between phages were observed at different temperatures (20°C/25°C, 4°C and -20°C), where phages were less stable. At lower temperatures (-80°C and -196°C), all phages showed good viability after 24 months regardless of the stabilizer. Differences between phages were also observed after lyophilization although the addition of skim milk yielded a dry powder with a stable titer after 24 months. As an alternative to facilitate storage and transportation, phage encapsulation has been also explored. Phage phiIPLA-RODI encapsulated in alginate capsules retained high viability when stored at 4°C for 6 months and at 20°C for 1 month. Moreover, the spray-dryer technique allowed obtaining dry powders containing viable encapsulated phages (phiIPLA-RODI and phiIPLA88) in both skim milk and trehalose for 12 months at 4°C. Storage of phages at 20°C was less effective; in fact, phiIPLA88 was stable for at least 12 months in trehalose but not in skim milk, while phiIPLA-RODI was stable only for 6 months in either stabilizer. These results suggest that encapsulated phages might be a suitable way for shipping phages.


Assuntos
Anti-Infecciosos/metabolismo , Fagos de Staphylococcus/metabolismo , Cápsulas , Liofilização , Humanos , Desenvolvimento Industrial , Infecções Estafilocócicas/terapia , Staphylococcus aureus/virologia , Staphylococcus epidermidis/virologia , Temperatura , Virologia/métodos
12.
Viruses ; 10(4)2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29617332

RESUMO

Staphylococcus aureus is a major causative agent of infections associated with hospital environments, where antibiotic-resistant strains have emerged as a significant threat. Phage therapy could offer a safe and effective alternative to antibiotics. Phage preparations should comply with quality and safety requirements; therefore, it is important to develop efficient production control technologies. This study was conducted to develop and evaluate a rapid and reliable method for identifying staphylococcal bacteriophages, based on detecting their specific proteins using matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) profiling that is among the suggested methods for meeting the regulations of pharmaceutical authorities. Five different phage purification techniques were tested in combination with two MALDI-TOF MS matrices. Phages, either purified by CsCl density gradient centrifugation or as resuspended phage pellets, yielded mass spectra with the highest information value if ferulic acid was used as the MALDI matrix. Phage tail and capsid proteins yielded the strongest signals whereas the culture conditions had no effect on mass spectral quality. Thirty-seven phages from Myoviridae, Siphoviridae or Podoviridae families were analysed, including 23 siphophages belonging to the International Typing Set for human strains of S. aureus, as well as phages in preparations produced by Microgen, Bohemia Pharmaceuticals and MB Pharma. The data obtained demonstrate that MALDI-TOF MS can be used to effectively distinguish between Staphylococcus-specific bacteriophages.


Assuntos
Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Fagos de Staphylococcus/classificação , Produtos Biológicos/isolamento & purificação , Fracionamento Químico/métodos , Análise por Conglomerados , Humanos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/virologia , Proteínas Virais/análise , Proteínas Virais/química , Replicação Viral
13.
Protein J ; 35(6): 407-415, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27812887

RESUMO

Phi11, a temperate bacteriophage of Staphylococcus aureus, has been found to harbor a cro repressor gene which facilitates Phi11 to adopt the lytic mode of development. The Cro protein has been found to bind very specifically to a 15-bp operator DNA, located in the Phi11 cI-cro intergenic region [1]. To investigate the effects exerted by different ions upon the interaction between Cro and its cognate operator DNA, we have employed gel shift assays as well as circular dichroism spectral analysis. In this communication, we have shown that NH4+ and acetate- ions better facilitated the binding of Cro with its cognate operator as compared to Na+, K+ and Li+. Interestingly, Mg2+, carbonate2- and Citrate3- have an inhibitory effect upon the binding. The effect of the said ions upon the structure of Cro was also investigated by circular dichroism and it was found that other than Citrate3- ions, none of the other ions destabilised the protein. On the other hand, Mg2+ and carbonate2- ions maintained the structure of the protein but severely hampered its functional activity. Citrate3- ions severely unfolded Cro and also inhibited its function. Considering all the data, NH4+ and acetate- ions appeared to be more suitable in maintaining the biological activity of Cro.


Assuntos
DNA Viral/química , Proteínas Repressoras/química , Fagos de Staphylococcus/química , Staphylococcus aureus/virologia , Proteínas Virais/química , Proteínas Virais Reguladoras e Acessórias/química , Acetatos/química , Amônia/química , Carbonatos/química , Dicroísmo Circular , Ácido Cítrico/química , DNA Viral/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Lítio/química , Magnésio/química , Potássio/química , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Proteínas Repressoras/isolamento & purificação , Proteínas Repressoras/metabolismo , Sódio/química , Fagos de Staphylococcus/metabolismo , Proteínas Virais/isolamento & purificação , Proteínas Virais/metabolismo , Proteínas Virais Reguladoras e Acessórias/isolamento & purificação , Proteínas Virais Reguladoras e Acessórias/metabolismo
14.
Appl Environ Microbiol ; 82(19): 5763-74, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27422842

RESUMO

UNLABELLED: Thanks to their wide host range and virulence, staphylococcal bacteriophages (phages) belonging to the genus Twortlikevirus (staphylococcal Twort-like phages) are regarded as ideal candidates for clinical application for Staphylococcus aureus infections due to the emergence of antibiotic-resistant bacteria of this species. To increase the usability of these phages, it is necessary to understand the mechanism underlying host recognition, especially the receptor-binding proteins (RBPs) that determine host range. In this study, we found that the staphylococcal Twort-like phage ΦSA012 possesses at least two RBPs. Genomic analysis of five mutant phages of ΦSA012 revealed point mutations in orf103, in a region unique to staphylococcal Twort-like phages. Phages harboring mutated ORF103 could not infect S. aureus strains in which wall teichoic acids (WTAs) are glycosylated with α-N-acetylglucosamine (α-GlcNAc). A polyclonal antibody against ORF103 also inhibited infection by ΦSA012 in the presence of α-GlcNAc, suggesting that ORF103 binds to α-GlcNAc. In contrast, a polyclonal antibody against ORF105, a short tail fiber component previously shown to be an RBP, inhibited phage infection irrespective of the presence of α-GlcNAc. Immunoelectron microscopy indicated that ORF103 is a tail fiber component localized at the bottom of the baseplate. From these results, we conclude that ORF103 binds α-GlcNAc in WTAs, whereas ORF105, the primary RBP, is likely to bind the WTA backbone. These findings provide insight into the infection mechanism of staphylococcal Twort-like phages. IMPORTANCE: Staphylococcus phages belonging to the genus Twortlikevirus (called staphylococcal Twort-like phages) are considered promising agents for control of Staphylococcus aureus due to their wide host range and highly lytic capabilities. Although staphylococcal Twort-like phages have been studied widely for therapeutic purposes, the host recognition process of staphylococcal Twort-like phages remains unclear. This work provides new findings about the mechanisms of host recognition of the staphylococcal Twort-like phage ΦSA012. The details of the host recognition mechanism of ΦSA012 will allow us to analyze the mechanisms of infection and expand the utility of staphylococcal Twort-like phages for the control of S. aureus.


Assuntos
Genoma Viral , Especificidade de Hospedeiro , Fagos de Staphylococcus/fisiologia , Staphylococcus aureus/virologia , Proteínas Virais/metabolismo , Ligação Proteica , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/metabolismo , Proteínas Virais/genética
15.
Antimicrob Agents Chemother ; 60(4): 2551-3, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26833148

RESUMO

HY-133 is a recombinant bacteriophage endolysin with bactericidal activity againstStaphylococcus aureus Here, HY-133 showedin vitroactivity against major African methicillin-susceptible and methicillin-resistantS. aureuslineages and ceftaroline/ceftobiprole- and borderline oxacillin-resistant isolates. HY-133 was also active againstStaphylococcus schweitzeri, a recently described species of theS. aureuscomplex. The activity of HY-133 on the tested isolates (MIC50, 0.25 µg/ml; MIC90, 0.5 µg/ml; range, 0.125 to 0.5 µg/ml) was independent of the species and strain background or antibiotic resistance.


Assuntos
Antibacterianos/farmacologia , Endopeptidases/farmacologia , Proteínas Recombinantes/farmacologia , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus/efeitos dos fármacos , África , Antibacterianos/biossíntese , Cefalosporinas/farmacologia , Endopeptidases/biossíntese , Endopeptidases/genética , Humanos , Resistência a Meticilina/genética , Testes de Sensibilidade Microbiana , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Infecções Estafilocócicas/microbiologia , Staphylococcus/crescimento & desenvolvimento , Staphylococcus/isolamento & purificação , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/isolamento & purificação , Resistência beta-Lactâmica/genética , Ceftarolina
16.
J Gen Virol ; 97(5): 1272-1281, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26873847

RESUMO

Holins are phage-encoded hydrophobic membrane proteins that spontaneously and non-specifically accumulate and form lesions in the cytoplasmic membrane. The ORF72 gene (also designated HolGH15) derived from the genome of the Staphylococcus aureus phage GH15 was predicted to encode a membrane protein. An analysis indicated that the protein encoded by HolGH15 potentially consisted of two hydrophobic transmembrane helices. This protein exhibited the structural characteristics of class II holins and belonged to the phage_holin_1 superfamily. Expression of HolGH15 in Escherichia coli BL21 cells resulted in growth retardation of the host cells, which was triggered prematurely by the addition of 2,4-dinitrophenol. The expression of HolGH15 caused morphological alterations in engineered E. coli cells, including loss of the cell wall and cytoplasmic membrane integrity and release of intracellular components, which were visualized by transmission electron microscopy. HolGH15 exerted efficient antibacterial activity at 37 °C and pH 5.2. Mutation analysis indicated that the two transmembrane domains of HolGH15 were indispensable for the activity of the full-length protein. HolGH15 showed a broad antibacterial range: it not only inhibited Staphylococcus aureus, but also demonstrated antibacterial activity against other species, including Listeria monocytogenes, Bacillus subtilis, Pseudomonas aeruginosa, Klebsiella pneumoniae and E. coli. At the minimal inhibitory concentration, HolGH15 evoked the release of cellular contents and resulted in the shrinkage and death of Staphylococcus aureus and Listeria monocytogenes cells. To the best of our knowledge, this study is the first report of a Staphylococcus aureus phage holin that exerts antibacterial activity against heterogeneous pathogens.


Assuntos
Proteínas de Membrana/metabolismo , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/virologia , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Antibacterianos , Clonagem Molecular , Citoplasma , Regulação Viral da Expressão Gênica , Genoma Viral , Listeria monocytogenes , Proteínas de Membrana/genética , Mutação , Consumo de Álcool por Menores
17.
PLoS One ; 10(11): e0142832, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26554919

RESUMO

Livestock associated methicillin-resistant Staphylococcus aureus (LA-MRSA) draws concern from the public health community because in some countries these organisms may represent the largest reservoir of MRSA outside hospital settings. Recent studies indicate LA-MRSA strains from swine are more genetically diverse than the first reported sequence type ST398. In the US, a diverse population of LA-MRSA is found including organisms of the ST398, ST9, and ST5 lineages. Occurrence of ST5 MRSA in swine is of particular concern since ST5 is among the most prevalent lineages causing clinical infections in humans. The prominence of ST5 in clinical disease is believed to result from acquisition of bacteriophages containing virulence or host-adapted genes including the immune-evasion cluster (IEC) genes carried by ß-hemolysin converting bacteriophages, whose absence in LA-MRSA ST398 is thought to contribute to reduced rates of human infection and transmission associated with this lineage. The goal of this study was to investigate the prevalence of IEC genes associated with ß-hemolysin converting bacteriophages in MRSA ST5 isolates obtained from agricultural sources, including swine, swine facilities, and humans with short- or long-term swine exposure. To gain a broader perspective, the prevalence of these genes in LA-MRSA ST5 strains was compared to the prevalence in clinical MRSA ST5 strains from humans with no known exposure to swine. IEC genes were not present in any of the tested MRSA ST5 strains from agricultural sources and the ß-hemolysin gene was intact in these strains, indicating the bacteriophage's absence. In contrast, the prevalence of the ß-hemolysin converting bacteriophage in MRSA ST5 strains from humans with no exposure to swine was 90.4%. The absence of ß-hemolysin converting bacteriophage in LA-MRSA ST5 isolates is consistent with previous reports evaluating ST398 strains and provides genetic evidence indicating LA-MRSA ST5 isolates may harbor a reduced capacity to cause severe disease in immunocompetent humans.


Assuntos
Proteínas Hemolisinas/metabolismo , Evasão da Resposta Imune/genética , Staphylococcus aureus Resistente à Meticilina/isolamento & purificação , Fagos de Staphylococcus/metabolismo , Suínos/microbiologia , Animais , Bacteriófagos , Proteínas Hemolisinas/genética , Humanos , Staphylococcus aureus Resistente à Meticilina/genética , Staphylococcus aureus Resistente à Meticilina/imunologia , Staphylococcus aureus Resistente à Meticilina/virologia
18.
Virol J ; 11: 133, 2014 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-25064136

RESUMO

BACKGROUND: Bacteriophages encode endolysins to lyse their host cell and allow escape of their progeny. Endolysins are also active against Gram-positive bacteria when applied from the outside and are thus attractive anti-bacterial agents. LysK, an endolysin from staphylococcal phage K, contains an N-terminal cysteine-histidine dependent amido-hydrolase/peptidase domain (CHAP(K)), a central amidase domain and a C-terminal SH3b cell wall-binding domain. CHAP(K) cleaves bacterial peptidoglycan between the tetra-peptide stem and the penta-glycine bridge. METHODS: The CHAP(K) domain of LysK was crystallized and high-resolution diffraction data was collected both from a native protein crystal and a methylmercury chloride derivatized crystal. The anomalous signal contained in the derivative data allowed the location of heavy atom sites and phase determination. The resulting structures were completed, refined and analyzed. The presence of calcium and zinc ions in the structure was confirmed by X-ray fluorescence emission spectroscopy. Zymogram analysis was performed on the enzyme and selected site-directed mutants. RESULTS: The structure of CHAP(K) revealed a papain-like topology with a hydrophobic cleft, where the catalytic triad is located. Ordered buffer molecules present in this groove may mimic the peptidoglycan substrate. When compared to previously solved CHAP domains, CHAP(K) contains an additional lobe in its N-terminal domain, with a structural calcium ion, coordinated by residues Asp45, Asp47, Tyr49, His51 and Asp56. The presence of a zinc ion in the active site was also apparent, coordinated by the catalytic residue Cys54 and a possible substrate analogue. Site-directed mutagenesis was used to demonstrate that residues involved in calcium binding and of the proposed active site were important for enzyme activity. CONCLUSIONS: The high-resolution structure of the CHAP(K) domain of LysK was determined, suggesting the location of the active site, the substrate-binding groove and revealing the presence of a structurally important calcium ion. A zinc ion was found more loosely bound. Based on the structure, we propose a possible reaction mechanism. Future studies will be aimed at co-crystallizing CHAP(K) with substrate analogues and elucidating its role in the complete LysK protein. This, in turn, may lead to the design of site-directed mutants with altered activity or substrate specificity.


Assuntos
Domínio Catalítico , Endopeptidases/química , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Fagos de Staphylococcus/metabolismo , Sítios de Ligação , Catálise , Endopeptidases/genética , Endopeptidases/metabolismo , Íons/metabolismo , Metais/metabolismo , Modelos Moleculares , Mutação , Ligação Proteica , Multimerização Proteica , Staphylococcus aureus/virologia
19.
Analyst ; 139(1): 179-86, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24255915

RESUMO

Rapid, specific detection of pathogenic bacteria remains a major challenge in infectious disease diagnostics. Bacteriophages can show genus- or even species-level specificity and have been developed for biosensing purposes, but the possibility of using individual phage proteins for detection has not been fully explored. This work exploits the ability of specific phage proteins, the endolysins LysK and Φ11, and the bacteriocin lysostaphin, fixed on silicon wafers to bind staphylococci. The proteins show activity against eight tested clinical isolates of S. aureus and to S. epidermidis, but no binding to Escherichia coli and limited binding to Micrococcus. Binding was quantified by clearing assays in solution and by functionalization of silicon wafers followed by light microscopy. Bacterial binding densities on functionalized surfaces were ~3 cells/100 µm(2). The small size of the proteins makes the system robust and easy to handle, and the principle is generalizable to many different biosensor platforms, including label-free systems such as optical microresonators.


Assuntos
Técnicas Biossensoriais/métodos , Fagos de Staphylococcus/química , Staphylococcus aureus/isolamento & purificação , Staphylococcus epidermidis/isolamento & purificação , Humanos , Ligação Proteica/fisiologia , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/metabolismo , Staphylococcus epidermidis/metabolismo
20.
Curr Microbiol ; 68(2): 204-10, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24077954

RESUMO

Bacteriophage genes offer a potential resource for development of new antibiotics. Here, we identify at least six genes of Staphylococcus aureus phage Sb-1 that have bactericidal activity when expressed in Escherichia coli. Since the natural host is gram-positive, and E. coli is gram-negative, it is likely that a variety of quite different bacterial pathogens would be susceptible to each of these bactericidal activities, which therefore might serve as the basis for development of new wide-spectrum antibiotics. We show that two of these gene products target E. coli protein synthesis.


Assuntos
Genes Virais , Viabilidade Microbiana/genética , Fagos de Staphylococcus/genética , Staphylococcus aureus/virologia , Clonagem Molecular , Expressão Gênica , Biossíntese de Proteínas , Fagos de Staphylococcus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...