Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33649203

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , Terapia de Fagos , Infecciones Estafilocócicas , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/terapia , Infecciones Estafilocócicas/virología , Staphylococcus aureus/metabolismo , Staphylococcus aureus/virología
2.
Microb Cell Fact ; 20(1): 56, 2021 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-33653327

RESUMEN

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.


Asunto(s)
Infecciones Estafilocócicas/metabolismo , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/metabolismo , Propiedades de Superficie
3.
Appl Microbiol Biotechnol ; 104(11): 5145-5158, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32248441

RESUMEN

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.


Asunto(s)
Microbiología de Alimentos/métodos , Podoviridae/metabolismo , Fagos de Staphylococcus/aislamiento & purificación , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/efectos de los fármacos , Animales , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Pollos/virología , Peces/virología , Genoma Viral , Especificidad del Huésped , Nisina/farmacología , Podoviridae/genética , Podoviridae/aislamiento & purificación , Carne de Cerdo/virología , Fagos de Staphylococcus/genética , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/virología , Virulencia/efectos de los fármacos
4.
Antimicrob Agents Chemother ; 60(4): 2551-3, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26833148

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , Endopeptidasas/farmacología , Proteínas Recombinantes/farmacología , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/efectos de los fármacos , Staphylococcus/efectos de los fármacos , África , Antibacterianos/biosíntesis , Cefalosporinas/farmacología , Endopeptidasas/biosíntesis , Endopeptidasas/genética , Humanos , Resistencia a la Meticilina/genética , Pruebas de Sensibilidad Microbiana , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus/crecimiento & desarrollo , Staphylococcus/aislamiento & purificación , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/aislamiento & purificación , Resistencia betalactámica/genética , Ceftarolina
5.
J Gen Virol ; 97(5): 1272-1281, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26873847

RESUMEN

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.


Asunto(s)
Proteínas de la Membrana/metabolismo , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/virología , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Antibacterianos , Clonación Molecular , Citoplasma , Regulación Viral de la Expresión Génica , Genoma Viral , Listeria monocytogenes , Proteínas de la Membrana/genética , Mutación , Consumo de Alcohol en Menores
6.
Appl Environ Microbiol ; 82(19): 5763-74, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27422842

RESUMEN

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.


Asunto(s)
Genoma Viral , Especificidad del Huésped , Fagos de Staphylococcus/fisiología , Staphylococcus aureus/virología , Proteínas Virales/metabolismo , Unión Proteica , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/metabolismo , Proteínas Virales/genética
7.
Nature ; 465(7299): 779-82, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20473284

RESUMEN

Staphylococcal superantigen-carrying pathogenicity islands (SaPIs) are discrete, chromosomally integrated units of approximately 15 kilobases that are induced by helper phages to excise and replicate. SaPI DNA is then efficiently encapsidated in phage-like infectious particles, leading to extremely high frequencies of intra- as well as intergeneric transfer. In the absence of helper phage lytic growth, the island is maintained in a quiescent prophage-like state by a global repressor, Stl, which controls expression of most of the SaPI genes. Here we show that SaPI derepression is effected by a specific, non-essential phage protein that binds to Stl, disrupting the Stl-DNA complex and thereby initiating the excision-replication-packaging cycle of the island. Because SaPIs require phage proteins to be packaged, this strategy assures that SaPIs will be transferred once induced. Several different SaPIs are induced by helper phage 80alpha and, in each case, the SaPI commandeers a different non-essential phage protein for its derepression. The highly specific interactions between different SaPI repressors and helper-phage-encoded antirepressors represent a remarkable evolutionary adaptation involved in pathogenicity island mobilization.


Asunto(s)
Islas Genómicas/genética , Virus Helper/enzimología , Proteínas Represoras/antagonistas & inhibidores , Fagos de Staphylococcus/enzimología , Staphylococcus aureus/genética , Regulación hacia Arriba/genética , Proteínas Virales/metabolismo , Alelos , Secuencia de Aminoácidos , ADN/biosíntesis , ADN/genética , Replicación del ADN , Virus Helper/genética , Virus Helper/metabolismo , Virus Helper/fisiología , Lisogenia/fisiología , Datos de Secuencia Molecular , Profagos/metabolismo , Profagos/fisiología , Pirofosfatasas/química , Pirofosfatasas/genética , Pirofosfatasas/metabolismo , Recombinación Genética/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Choque Séptico , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/metabolismo , Fagos de Staphylococcus/fisiología , Staphylococcus aureus/patogenicidad , Staphylococcus aureus/virología , Superantígenos/genética , Proteínas Virales/química , Proteínas Virales/genética
8.
Virol J ; 11: 133, 2014 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-25064136

RESUMEN

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.


Asunto(s)
Dominio Catalítico , Endopeptidasas/química , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Fagos de Staphylococcus/metabolismo , Sitios de Unión , Catálisis , Endopeptidasas/genética , Endopeptidasas/metabolismo , Iones/metabolismo , Metales/metabolismo , Modelos Moleculares , Mutación , Unión Proteica , Multimerización de Proteína , Staphylococcus aureus/virología
9.
Analyst ; 139(1): 179-86, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24255915

RESUMEN

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.


Asunto(s)
Técnicas Biosensibles/métodos , Fagos de Staphylococcus/química , Staphylococcus aureus/aislamiento & purificación , Staphylococcus epidermidis/aislamiento & purificación , Humanos , Unión Proteica/fisiología , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/metabolismo , Staphylococcus epidermidis/metabolismo
10.
Curr Microbiol ; 68(2): 204-10, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24077954

RESUMEN

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.


Asunto(s)
Genes Virales , Viabilidad Microbiana/genética , Fagos de Staphylococcus/genética , Staphylococcus aureus/virología , Clonación Molecular , Expresión Génica , Biosíntesis de Proteínas , Fagos de Staphylococcus/metabolismo
11.
mBio ; 15(6): e0067924, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38752726

RESUMEN

Phages and bacteria have a long history of co-evolution. However, these dynamics of phage-host interactions are still largely unknown; identification of phage inhibitors that remodel host metabolism will provide valuable information for target development for antimicrobials. Here, we perform a comprehensive screen for early-gene products of ΦNM1 that inhibit cell growth in Staphylococcus aureus. A small membrane protein, Gp11, with inhibitory effects on S. aureus cell division was identified. A bacterial two-hybrid library containing 345 essential S. aureus genes was constructed to screen for targets of Gp11, and Gp11 was found to interact with MurG and DivIC. Defects in cell growth and division caused by Gp11 were dependent on MurG and DivIC, which was further confirmed using CRISPRi hypersensitivity assay. Gp11 interacts with MurG, the protein essential for cell wall formation, by inhibiting the production of lipid II to regulate peptidoglycan (PG) biosynthesis on the cell membrane. Gp11 also interacts with cell division protein DivIC, an essential part of the division machinery necessary for septal cell wall assembly, to disrupt the recruitment of division protein FtsW. Mutations in Gp11 result in loss of its ability to cause growth defects, whereas infection with phage in which the gp11 gene has been deleted showed a significant increase in lipid II production in S. aureus. Together, our findings reveal that a phage early-gene product interacts with essential host proteins to disrupt PG biosynthesis and block S. aureus cell division, suggesting a potential pathway for the development of therapeutic approaches to treat pathogenic bacterial infections. IMPORTANCE: Understanding the interplay between phages and their hosts is important for the development of novel therapies against pathogenic bacteria. Although phages have been used to control methicillin-resistant Staphylococcus aureus infections, our knowledge related to the processes in the early stages of phage infection is still limited. Owing to the fact that most of the phage early proteins have been classified as hypothetical proteins with uncertain functions, we screened phage early-gene products that inhibit cell growth in S. aureus, and one protein, Gp11, selectively targets essential host genes to block the synthesis of the peptidoglycan component lipid II, ultimately leading to cell growth arrest in S. aureus. Our study provides a novel insight into the strategy by which Gp11 blocks essential host cellular metabolism to influence phage-host interaction. Importantly, dissecting the interactions between phages and host cells will contribute to the development of new and effective therapies to treat bacterial infections.


Asunto(s)
División Celular , Peptidoglicano , Fagos de Staphylococcus , Staphylococcus aureus , Proteínas Virales , Staphylococcus aureus/virología , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Peptidoglicano/metabolismo , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Pared Celular/metabolismo , Pared Celular/virología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética
12.
J Bacteriol ; 195(16): 3621-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23749973

RESUMEN

Promoter recognition in bacteria is mediated primarily by the σ subunit of RNA polymerase (RNAP), which makes sequence-specific contacts with the promoter -10 and -35 elements in the context of the RNAP holoenzyme. However, the RNAP α subunit can also contribute to promoter recognition by making sequence-specific contacts with upstream (UP) elements that are associated with a subset of promoters, including the rRNA promoters. In Escherichia coli, these interactions between the RNAP α subunit (its C-terminal domain [CTD], in particular) and UP element DNA result in significant stimulation of rRNA transcription. Among the many cellular and bacteriophage-encoded regulators of transcription initiation that have been functionally dissected, most exert their effects via a direct interaction with either the σ or the α subunit. An unusual example is provided by a phage-encoded inhibitor of RNA synthesis in Staphylococcus aureus. This protein, phage G1 gp67, which binds tightly to σ in the context of the S. aureus RNAP holoenzyme, has recently been shown to exert selective effects on transcription by inhibiting the function of the α subunit CTD (αCTD). Here we report the development of a gp67-responsive E. coli-based transcription system. We examine transcription in vitro from promoters that do or do not carry the UP element associated with a well-characterized E. coli rRNA promoter. Our findings indicate that the αCTD can increase promoter activity significantly even in the absence of an UP element. We also find that gp67 can exert αCTD-dependent or αCTD-independent effects on transcription depending on the particular promoter, indicating that the mechanism of gp67 action is context dependent.


Asunto(s)
Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Fagos de Staphylococcus/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Escherichia coli/genética , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Fagos de Staphylococcus/genética , Staphylococcus aureus
13.
Antimicrob Agents Chemother ; 57(11): 5737-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23979742

RESUMEN

Nucleotide sequencing of the fusB-flanking regions in two fusidic acid-resistant Staphylococcus epidermidis isolates with the type IV aj1-leader peptide (LP)-fusB structure (lacking aj1) revealed that their fusB gene was located on novel phage-related islands inserted downstream of smpB and are here referred to as SeRIfusB-3692 and SePIfusB-857. The novel SePIfusB-857 structure was followed by SeCI857, forming a composite pathogenicity island which contained a putative virulence gene, vapE. The linkage of fusB and vapE may contribute to bacterial adaption.


Asunto(s)
Farmacorresistencia Bacteriana/genética , Regulación Bacteriana de la Expresión Génica , Islas Genómicas , Fagos de Staphylococcus/genética , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/patogenicidad , Factores de Virulencia/genética , Adaptación Fisiológica , Antibacterianos/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Ácido Fusídico/farmacología , Pruebas de Sensibilidad Microbiana , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Análisis de Secuencia de ADN , Fagos de Staphylococcus/metabolismo , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/metabolismo , Virulencia , Factores de Virulencia/metabolismo
14.
Genes Genomics ; 45(2): 191-202, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36520268

RESUMEN

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.


Asunto(s)
Fagos de Staphylococcus , Staphylococcus aureus , Factores de Transcripción , Proteínas Virales , Factores de Virulencia , Animales , Ratones , Absceso , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Staphylococcus aureus/patogenicidad , Staphylococcus aureus/virología , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/metabolismo , Factores de Transcripción/genética , Proteínas Virales/genética , Virulencia/genética , Factores de Virulencia/genética
15.
Appl Environ Microbiol ; 78(7): 2297-305, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22286996

RESUMEN

Staphylococci cause bovine mastitis, with Staphylococcus aureus being responsible for the majority of the mastitis-based losses to the dairy industry (up to $2 billion/annum). Treatment is primarily with antibiotics, which are often ineffective and potentially contribute to resistance development. Bacteriophage endolysins (peptidoglycan hydrolases) present a promising source of alternative antimicrobials. Here we evaluated two fusion proteins consisting of the streptococcal λSA2 endolysin endopeptidase domain fused to staphylococcal cell wall binding domains from either lysostaphin (λSA2-E-Lyso-SH3b) or the staphylococcal phage K endolysin, LysK (λSA2-E-LysK-SH3b). We demonstrate killing of 16 different S. aureus mastitis isolates, including penicillin-resistant strains, by both constructs. At 100 µg/ml in processed cow milk, λSA2-E-Lyso-SH3b and λSA2-E-LysK-SH3b reduced the S. aureus bacterial load by 3 and 1 log units within 3 h, respectively, compared to a buffer control. In contrast to λSA2-E-Lyso-SH3b, however, λSA2-E-LysK-SH3b permitted regrowth of the pathogen after 1 h. In a mouse model of mastitis, infusion of 25 µg of λSA2-E-Lyso-SH3b or λSA2-E-LysK-SH3b into mammary glands reduced S. aureus CFU by 0.63 or 0.81 log units, compared to >2 log for lysostaphin. Both chimeras were synergistic with lysostaphin against S. aureus in plate lysis checkerboard assays. When tested in combination in mice, λSA2-E-LysK-SH3b and lysostaphin (12.5 µg each/gland) caused a 3.36-log decrease in CFU. Furthermore, most protein treatments reduced gland wet weights and intramammary tumor necrosis factor alpha (TNF-α) concentrations, which serve as indicators of inflammation. Overall, our animal model results demonstrate the potential of fusion peptidoglycan hydrolases as antimicrobials for the treatment of S. aureus-induced mastitis.


Asunto(s)
Endopeptidasas/farmacología , Lisostafina/farmacología , Glándulas Mamarias Animales/microbiología , Mastitis Bovina/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Staphylococcus aureus/efectos de los fármacos , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bovinos , Enfermedades de los Bovinos/tratamiento farmacológico , Enfermedades de los Bovinos/microbiología , Sinergismo Farmacológico , Endopeptidasas/genética , Endopeptidasas/metabolismo , Endopeptidasas/uso terapéutico , Femenino , Lisostafina/metabolismo , Lisostafina/uso terapéutico , Mastitis Bovina/microbiología , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Modelos Animales , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/aislamiento & purificación , Resultado del Tratamiento
16.
Viruses ; 14(11)2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36366569

RESUMEN

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.


Asunto(s)
Toxinas Bacterianas , Infecciones Estafilocócicas , Animales , Humanos , Staphylococcus aureus , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Infecciones Estafilocócicas/microbiología , Estadios del Ciclo de Vida , Mamíferos
17.
Microbiol Spectr ; 10(1): e0146621, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35196798

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , Endopeptidasas/farmacología , Fotoquimioterapia , Infecciones Estafilocócicas/microbiología , Fagos de Staphylococcus/química , Staphylococcus/efectos de los fármacos , Staphylococcus/fisiología , Animales , Antibacterianos/química , Biopelículas/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple , Endopeptidasas/química , Endopeptidasas/metabolismo , Humanos , Indoles/química , Luz , Compuestos de Organosilicio/química , Fármacos Fotosensibilizantes/química , Especies Reactivas de Oxígeno/metabolismo , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus/virología , Fagos de Staphylococcus/metabolismo
18.
Curr Microbiol ; 63(6): 538-42, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21947237

RESUMEN

LysGH15, a phage endolysin, exhibits a particularly broad lytic spectrum against Staphylococcus aureus, especially methicillin-resistant S. aureus (MRSA). Sequence analysis reveals that this endolysin contains a C-terminal cell wall binding domain (SH3b), which causes the endolysin to bind to host strains. In this study, the substrate binding affinity of the SH3b domain (LysGH15B) was evaluated. A fusion protein of LysGH15B and green fluorescent protein (LysGH15B-GFP) were cloned and expressed in Escherichia coli. Laser scanning confocal microscopy was used to detect the fluorescence of the treated cells irradiated at different excitation wavelengths and to determine the binding activity of LysGH15B-GFP and GFP. We found that LysGH15B-GFP not only generated green fluorescence, but, more importantly, also displayed specific affinity to staphylococcal isolates, especially MRSA. In contrast, the single GFP did not display any binding activity. The high affinity was attributed to the portion of LysGH15B and the binding activity of the fusion protein was specific to staphylococci. This study provides an insight into the SH3b domain of LysGH15. The specific binding activity may cause LysGH15B to serve as an anchoring device, and offer an alternative approach for cell surface attachment onto staphylococci.


Asunto(s)
Endopeptidasas/fisiología , Staphylococcus aureus Resistente a Meticilina/virología , Fagos de Staphylococcus/metabolismo , Clonación Molecular , Electroforesis en Gel de Poliacrilamida , Endopeptidasas/biosíntesis , Endopeptidasas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Microscopía Confocal , Infecciones Estafilocócicas/microbiología , Dominios Homologos src
19.
Curr Microbiol ; 63(1): 39-45, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21519800

RESUMEN

Staphylococcus aureus causes a wide range of suppurative infections in humans and animals. Due to its high virulence, ability to adopt various environmental conditions, and acquired multiple drug resistance, treatment of such infections has become difficult. Therefore, there is an immense need to develop alternate drug modalities to control this pathogen. In past few years, phage-encoded endolysin therapy has emerged as a new hope not only due to its ability to specifically kill the target bacteria irrespective of their antibiotic sensitivity but also because of minimum or no side effects, a problem associated with antibiotic therapy. In this article, we report purification of a broad spectrum anti-staphylococcal endolysin (P-27/HP endolysin) encoded by phage P-27/HP isolated from sewage water. On SDS-PAGE endolysin resolved in three polypeptides of molecular weights 33.5, 48.6, and 62.2 kDa. Endolysin exhibited maximum in vitro lytic activity at temperature between 35 and 40°C and pH 7.0. In vivo experiments revealed considerable (99.9%) elimination of S. aureus 27/HP from spleens of endolysin-treated mice and had saved them from death due to bacteremia caused by S. aureus 27/HP challenge infection. Thus, P-27/HP endolysin offers suitable substitute of antibiotics to control S. aureus infections.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Endopeptidasas/farmacología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Proteínas Virales/farmacología , Animales , Antibacterianos/química , Antibacterianos/aislamiento & purificación , Antibacterianos/metabolismo , Endopeptidasas/química , Endopeptidasas/aislamiento & purificación , Endopeptidasas/metabolismo , Estabilidad de Enzimas , Femenino , Humanos , Masculino , Ratones , Aguas del Alcantarillado/virología , Fagos de Staphylococcus/química , Fagos de Staphylococcus/aislamiento & purificación , Fagos de Staphylococcus/metabolismo , Staphylococcus aureus/fisiología , Proteínas Virales/química , Proteínas Virales/aislamiento & purificación , Proteínas Virales/metabolismo
20.
Mol Microbiol ; 72(1): 98-108, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19347993

RESUMEN

SaPI1 and SaPIbov1 are chromosomal pathogenicity islands in Staphylococcus aureus that carry tst and other superantigen genes. They are induced to excise and replicate by certain phages, are efficiently encapsidated in SaPI-specific small particles composed of phage virion proteins and are transferred at very high frequencies. In this study, we have analysed three SaPI genes that are important for the phage-SaPI interaction, int (integrase) terS (phage terminase small subunit homologue) and pif (phage interference function). SaPI1 int is required for SaPI excision, replication and packaging in a donor strain, and is required for integration in a recipient. A SaPI1 int mutant, following phage induction, produces small SaPI-specific capsids which are filled with partial phage genomes. SaPIbov1 DNA is efficiently packaged into full-sized phage heads as well as into SaPI-specific small ones, whereas SaPI1 DNA is found almost exclusively in the small capsids. TerS, however, determines DNA packaging specificity but not the choice of large versus small capsids. This choice is influenced by SaPIbov1 gene 12, which prevents phage DNA packaging into small capsids, and which is also primarily responsible for interference by SaPIbov1 with phage reproduction.


Asunto(s)
Empaquetamiento del ADN , Endodesoxirribonucleasas/metabolismo , Islas Genómicas , Integrasas/metabolismo , Fagos de Staphylococcus/metabolismo , Cápside/metabolismo , Endodesoxirribonucleasas/genética , Integrasas/genética , Mutación , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/fisiología , Staphylococcus aureus/genética , Staphylococcus aureus/virología , Especificidad por Sustrato , Proteínas Virales/genética , Proteínas Virales/metabolismo , Ensamble de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA