Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
BMC Microbiol ; 24(1): 155, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704526

RESUMEN

BACKGROUND: The in-depth understanding of the role of lateral genetic transfer (LGT) in phage-prophage interactions is essential to rationalizing phage applications for human and animal therapy, as well as for food and environmental safety. This in silico study aimed to detect LGT between phages of potential industrial importance and their hosts. METHODS: A large array of genetic recombination detection algorithms, implemented in SplitsTree and RDP4, was applied to detect LGT between various Escherichia, Listeria, Salmonella, Campylobacter, Staphylococcus, Pseudomonas, and Vibrio phages and their hosts. PHASTER and RAST were employed respectively to identify prophages across the host genome and to annotate LGT-affected genes with unknown functions. PhageAI was used to gain deeper insights into the life cycle history of recombined phages. RESULTS: The split decomposition inferences (bootstrap values: 91.3-100; fit: 91.433-100), coupled with the Phi (0.0-2.836E-12) and RDP4 (P being well below 0.05) statistics, provided strong evidence for LGT between certain Escherichia, Listeria, Salmonella, and Campylobacter virulent phages and prophages of their hosts. The LGT events entailed mainly the phage genes encoding for hypothetical proteins, while some of these genetic loci appeared to have been affected even by intergeneric recombination in specific E. coli and S. enterica virulent phages when interacting with their host prophages. Moreover, it is shown that certain L. monocytogenes virulent phages could serve at least as the donors of the gene loci, involved in encoding for the basal promoter specificity factor, for L. monocytogenes. In contrast, the large genetic clusters were determined to have been simultaneously exchanged by many S. aureus prophages and some Staphylococcus temperate phages proposed earlier as potential therapeutic candidates (in their native or modified state). The above genetic clusters were found to encompass multiple genes encoding for various proteins, such as e.g., phage tail proteins, the capsid and scaffold proteins, holins, and transcriptional terminator proteins. CONCLUSIONS: It is suggested that phage-prophage interactions, mediated by LGT (including intergeneric recombination), can have a far-reaching impact on the co-evolutionary trajectories of industrial phages and their hosts especially when excessively present across microbially rich environments.


Asunto(s)
Profagos , Recombinación Genética , Profagos/genética , Campylobacter/virología , Campylobacter/genética , Staphylococcus/virología , Staphylococcus/genética , Transferencia de Gen Horizontal , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/clasificación , Listeria/virología , Listeria/genética , Salmonella/virología , Salmonella/genética , Evolución Molecular , Bacterias/virología , Bacterias/genética
2.
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
3.
Nat Commun ; 12(1): 6408, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34737316

RESUMEN

Staphylococcus aureus pathogenicity islands (SaPIs) are molecular parasites that hijack helper phages for their transfer. SaPIbov5, the prototypical member of a family of cos type SaPIs, redirects the assembly of ϕ12 helper capsids from prolate to isometric. This size and shape shift is dependent on the SaPIbov5-encoded protein Ccm, a homolog of the ϕ12 capsid protein (CP). Using cryo-electron microscopy, we have determined structures of prolate ϕ12 procapsids and isometric SaPIbov5 procapsids. ϕ12 procapsids have icosahedral end caps with Tend = 4 architecture and a Tmid = 14 cylindrical midsection, whereas SaPIbov5 procapsids have T = 4 icosahedral architecture. We built atomic models for CP and Ccm, and show that Ccm occupies the pentameric capsomers in the isometric SaPIbov5 procapsids, suggesting that preferential incorporation of Ccm pentamers prevents the cylindrical midsection from forming. Our results highlight that pirate elements have evolved diverse mechanisms to suppress phage multiplication, including the acquisition of phage capsid protein homologs.


Asunto(s)
Staphylococcus/virología , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Islas Genómicas/genética
4.
Nat Commun ; 12(1): 6965, 2021 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-34845206

RESUMEN

The host range of bacteriophages defines their impact on bacterial communities and genome diversity. Here, we characterize 94 novel staphylococcal phages from wastewater and establish their host range on a diversified panel of 117 staphylococci from 29 species. Using this high-resolution phage-bacteria interaction matrix, we unveil a multi-species host range as a dominant trait of the isolated staphylococcal phages. Phage genome sequencing shows this pattern to prevail irrespective of taxonomy. Network analysis between phage-infected bacteria reveals that hosts from multiple species, ecosystems, and drug-resistance phenotypes share numerous phages. Lastly, we show that phages throughout this network can package foreign genetic material enclosing an antibiotic resistance marker at various frequencies. Our findings indicate a weak host specialism of the tested phages, and therefore their potential to promote horizontal gene transfer in this environment.


Asunto(s)
Genoma Viral , Especificidad del Huésped , Fagos de Staphylococcus/genética , Staphylococcus/genética , Aguas Residuales , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Ecosistema , Transferencia de Gen Horizontal , Consorcios Microbianos/genética , Pruebas de Sensibilidad Microbiana , Filogenia , Staphylococcus/clasificación , Staphylococcus/efectos de los fármacos , Staphylococcus/virología , Fagos de Staphylococcus/clasificación , Fagos de Staphylococcus/aislamiento & purificación , Aguas Residuales/microbiología , Aguas Residuales/virología , Microbiología del Agua
5.
PLoS One ; 16(7): e0254382, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34292970

RESUMEN

Methicillin-resistant Staphylococcus pseudintermedius (MRSP) is an important opportunistic pathogenic bacterium of dogs that also occasionally colonize and infect humans. However, whether MRSP can adapt to human hosts is not clear and whole genome sequences of MRSP from humans are still limited. Genomic comparative analyses of 3 couples of isolates from dogs (n = 3) and humans (n = 3) belonging to ST45, ST112, and ST181, the dominant clones in Thailand were conducted to determine the degree of similarities between human and animal MRSP of a same ST. Among eight prophages, three prophages associated with the leucocidins genes (lukF/S-I), φVB88-Pro1, φVB16-Pro1 and φAP20-Pro1, were distributed in the human MRSPs, while their remnants, φAH18-Pro1, were located in the dog MRSPs. A novel composite pathogenicity island, named SpPI-181, containing two integrase genes was identified in the ST181 isolates. The distribution of the integrase genes of the eight prophages and SpPI-181 was also analysed by PCR in 77 additional MRSP isolates belonging to different STs. The PCR screen revealed diversity in prophage carriage, especially in ST45 isolates. Prophage φAK9-Pro1 was only observed in ST112 isolates from dogs and SpPI-181 was found associated with ST181 clonal lineage. Among the 3 couple of isolates, ST45 strains showed the highest number of single nucleotide polymorphisms (SNP) in their core genomes (3,612 SNPs). The genomic diversity of ST45 isolates suggested a high level of adaptation that may lead to different host colonization of successful clones. This finding provided data on the genomic differences of MRSP associated with colonization and adaption to different hosts.


Asunto(s)
Genoma Bacteriano , Islas Genómicas , Resistencia a la Meticilina/genética , Polimorfismo de Nucleótido Simple , Profagos/genética , Staphylococcus , Animales , Perros , Genes Virales , Humanos , Staphylococcus/genética , Staphylococcus/aislamiento & purificación , Staphylococcus/virología
6.
Nature ; 592(7855): 611-615, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33828299

RESUMEN

Horizontal gene transfer and mutation are the two major drivers of microbial evolution that enable bacteria to adapt to fluctuating environmental stressors1. Clustered, regularly interspaced, short palindromic repeats (CRISPR) systems use RNA-guided nucleases to direct sequence-specific destruction of the genomes of mobile genetic elements that mediate horizontal gene transfer, such as conjugative plasmids2 and bacteriophages3, thus limiting the extent to which bacteria can evolve by this mechanism. A subset of CRISPR systems also exhibit non-specific degradation of DNA4,5; however, whether and how this feature affects the host has not yet been examined. Here we show that the non-specific DNase activity of the staphylococcal type III-A CRISPR-Cas system increases mutations in the host and accelerates the generation of antibiotic resistance in Staphylococcus aureus and Staphylococcus epidermidis. These mutations require the induction of the SOS response to DNA damage and display a distinct pattern. Our results demonstrate that by differentially affecting both mechanisms that generate genetic diversity, type III-A CRISPR systems can modulate the evolution of the bacterial host.


Asunto(s)
Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/inmunología , Mutagénesis , Mutación , Staphylococcus/genética , Antibacterianos/farmacología , Bacteriófagos/clasificación , Bacteriófagos/fisiología , Proteínas Asociadas a CRISPR/metabolismo , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Desoxirribonucleasas/metabolismo , Farmacorresistencia Microbiana/efectos de los fármacos , Respuesta SOS en Genética/efectos de los fármacos , Staphylococcus/efectos de los fármacos , Staphylococcus/inmunología , Staphylococcus/virología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/virología , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/virología , Factores de Tiempo
7.
Nature ; 590(7847): 624-629, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33461211

RESUMEN

In the type III CRISPR-Cas immune response of prokaryotes, infection triggers the production of cyclic oligoadenylates that bind and activate proteins that contain a CARF domain1,2. Many type III loci are associated with proteins in which the CRISPR-associated Rossman fold (CARF) domain is fused to a restriction  endonuclease-like domain3,4. However, with the exception of the well-characterized Csm6 and Csx1 ribonucleases5,6, whether and how these inducible effectors provide defence is not known. Here we investigated a type III CRISPR accessory protein, which we name cyclic-oligoadenylate-activated single-stranded ribonuclease and single-stranded deoxyribonuclease 1 (Card1). Card1 forms a symmetrical dimer that has a large central cavity between its CRISPR-associated Rossmann fold and restriction endonuclease domains that binds cyclic tetra-adenylate. The binding of ligand results in a conformational change comprising the rotation of individual monomers relative to each other to form a more compact dimeric scaffold, in which a manganese cation coordinates the catalytic residues and activates the cleavage of single-stranded-but not double-stranded-nucleic acids (both DNA and RNA). In vivo, activation of Card1 induces dormancy of the infected hosts to provide immunity against phage infection and plasmids. Our results highlight the diversity of strategies used in CRISPR systems to provide immunity.


Asunto(s)
Nucleótidos de Adenina/metabolismo , Sistemas CRISPR-Cas/inmunología , ADN de Cadena Simple/metabolismo , Desoxirribonucleasas/metabolismo , Endorribonucleasas/metabolismo , Oligorribonucleótidos/metabolismo , ARN/metabolismo , Staphylococcus/enzimología , Staphylococcus/inmunología , Nucleótidos de Adenina/inmunología , Adenosina Trifosfato/metabolismo , Bacteriófagos/inmunología , Bacteriófagos/fisiología , Biocatálisis , Dominio Catalítico , Desoxirribonucleasas/química , Desoxirribonucleasas/genética , Endorribonucleasas/química , Endorribonucleasas/genética , Activación Enzimática , Ligandos , Manganeso/química , Manganeso/metabolismo , Modelos Moleculares , Oligorribonucleótidos/inmunología , Plásmidos/genética , Plásmidos/metabolismo , Multimerización de Proteína , Rotación , Staphylococcus/crecimiento & desarrollo , Staphylococcus/virología , Especificidad por Sustrato
8.
Sci Rep ; 10(1): 8659, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32457340

RESUMEN

A functional genetic switch from the lactococcal bacteriophage TP901-1, deciding which of two divergently transcribing promoters becomes most active and allows this bi-stable decision to be inherited in future generations requires a DNA region of less than 1 kb. The fragment encodes two repressors, CI and MOR, transcribed from the PR and PL promoters respectively. CI can repress the transcription of the mor gene at three operator sites (OR, OL, and OD), leading to the immune state. Repression of the cI gene, leading to the lytic (anti-immune) state, requires interaction between CI and MOR by an unknown mechanism, but involving a CI:MOR complex. A consensus for putative MOR binding sites (OM sites), and a common topology of three OM sites adjacent to the OR motif was here identified in diverse phage switches that encode CI and MOR homologs, in a search for DNA sequences similar to the TP901-1 switch. The OR site and all putative OM sites are important for establishment of the anti-immune repression of PR, and a putative DNA binding motif in MOR is needed for establishment of the anti-immune state. Direct evidence for binding between CI and MOR is here shown by pull-down experiments, chemical crosslinking, and size exclusion chromatography. The results are consistent with two possible models for establishment of the anti-immune repression of cI expression at the PR promoter.


Asunto(s)
Bacteriófagos/genética , Lactococcus lactis/virología , Regiones Promotoras Genéticas/genética , Elementos Reguladores de la Transcripción/genética , Proteínas Represoras/genética , Transactivadores/genética , Proteínas Reguladoras y Accesorias Virales/genética , Bacteriófagos/crecimiento & desarrollo , Sitios de Unión/genética , ADN Viral/genética , Proteínas de Unión al ADN/genética , Enterococcus/virología , Regulación Viral de la Expresión Génica/genética , Genoma Viral/genética , Lactococcus lactis/genética , Lisogenia/genética , Regiones Operadoras Genéticas/genética , Proteínas Represoras/metabolismo , Staphylococcus/virología , Streptococcus/virología , Transactivadores/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo
9.
Epidemiol Mikrobiol Imunol ; 69(1): 10-18, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32326711

RESUMEN

AIM: Staphylococcus aureus strains are the cause of frightening hospital and community infections, especially when they are resistant to antimicrobials, have important pathogenicity factors, or have biofilm production ability. Looking for novel therapeutic options which would be effective against such strains is one of the highest priorities of medicine and medical research. The study aim was to describe the occurrence of S. aureus strains and proportion of methicillin resistant strains (MRSA) detected in laboratories of the Microbiological Institute, Faculty of Medicine, Masaryk University (FM MU) and St. Anne's University Hospital, Brno in 2011-2018. Selected strains of S. aureus were tested for biofilm production ability and susceptibility to antimicrobials and Stafal®, a phage therapeutic agent. A prerequisite was to develop a simple routine method suitable for phage susceptibility testing of bacteria. MATERIAL AND METHODS: Altogether 867 clinical isolates of S. aureus and 132 strains of other species of the genus Staphylococcus (isolated in 2011-2017) were tested for susceptibility to the phage therapy preparation Stafal® using the double-layer agar method. All strains of S. aureus were tested for biofilm production ability by the modified Christensen method with the use of titration microplates and for susceptibility to antistaphylococcal antibiotics by the disk diffusion test. For 95 S. aureus strains, the outcome of the double-layer agar method (DAM) was compared with that of our newly designed method (ODM) based on optical density decrease of the bacterial suspension. RESULTS: During the study period, the laboratories of the Faculty of Medicine, Masaryk University (FM MU) and St. Anne's University Hospital, Brno detected 2900 strains of S. aureus per year on average. The proportion of MRSA among S. aureus isolates from blood culture and venous catheters ranged between 8.8-15.2 %. S. aureus strains recovered from venous catheters and blood culture were confirmed as stronger biofilm producers than those from other clinical specimens. MRSA strains showed higher biofilm production than methicillin susceptible strains (MSSA). As many as 90.4 % of S. aureus strains tested susceptible to the Stafal® preparation. Even a higher proportion, i.e. 99.0 %, of MRSA strains were Stafal® susceptible. No relationship was found between Stafal® susceptibility and biofilm production ability. Although Stafal® targets primarily S. aureus, some susceptibility (26.5 %) was also found for other staphylococcal species. A novel simple method designed for routine testing of susceptibility to phage therapy preparations based on optical density decrease was comparably sensitive and reliable as the commonly used double-layer agar method (DAM) and, in addition to being easy and rapid to perform, after prolonged suspension culture and at higher measurement frequency, it has an extra advantage of providing the possibility for monitoring also phage action dynamics. CONCLUSIONS: The proportion of MRSA strains detected in this study is comparable to that reported for the whole Czech Republic, and the biofilm production data are consistent with scientific evidence. The host range of the Stafal® preparation is relatively wide and covers most strains of S. aureus and some coagulase negative staphylococci. The highest efficiency of Stafal® (99.4 %) was observed against MRSA strains with multiple types of antibiotic resistance. In vitro testing of 867 strains of S. aureus and 132 other staphylococcal species has shown the phage therapy preparation Stafal® to be a suitable candidate therapeutic option for the treatment of staphylococcal infections, especially in case of failure of conventional antibiotic therapy. Moreover, a simple method for routine phage susceptibility testing of clinical bacterial isolates has been designed, which is an essential tool to be used in phage therapy.


Asunto(s)
Bacteriófagos , Infecciones Estafilocócicas , Staphylococcus , Antibacterianos/uso terapéutico , Bacteriófagos/fisiología , República Checa , Humanos , Técnicas In Vitro , Staphylococcus aureus Resistente a Meticilina/virología , Infecciones Estafilocócicas/terapia , Infecciones Estafilocócicas/virología , Staphylococcus/virología
10.
Viruses ; 12(3)2020 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-32156046

RESUMEN

Staphylococcal bacteriophages of the Kayvirus genus are candidates for therapeutic applications. One of their proteins, Tgl, is slightly similar to two staphylococcal virulence factors, secreted autolysins of lytic transglycosylase motifs IsaA and SceD. We show that Tgl is a lytic enzyme secreted by the bacterial transport system and localizes to cell peripheries like IsaA and SceD. It causes lysis of E. coli cells expressing the cloned tgl gene, but could be overproduced when depleted of signal peptide. S. aureus cells producing Tgl lysed in the presence of nisin, which mimics the action of phage holin. In vitro, Tgl protein was able to destroy S. aureus cell walls. The production of Tgl decreased S. aureus tolerance to vancomycin, unlike the production of SceD, which is associated with decreased sensitivity to vancomycin. In the genomes of kayviruses, the tgl gene is located a few genes away from the lysK gene, encoding the major endolysin. While lysK is a late phage gene, tgl can be transcribed by a host RNA polymerase, like phage early genes. Taken together, our data indicate that tgl belongs to the kayvirus lytic module and encodes an additional endolysin that can act in concert with LysK in cell lysis.


Asunto(s)
Biomarcadores , Fagos de Staphylococcus/fisiología , Staphylococcus/virología , Proteínas Virales/genética , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Antibacterianos/farmacología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bacteriólisis , Pared Celular , Endopeptidasas/genética , Endopeptidasas/metabolismo , Escherichia coli/genética , Genoma Viral , Viabilidad Microbiana/genética , Mutación , Plásmidos/genética , Transporte de Proteínas , Infecciones Estafilocócicas/microbiología , Staphylococcus/efectos de los fármacos , Fagos de Staphylococcus/patogenicidad , Vancomicina/farmacología , Proteínas Virales/química , Virulencia , Factores de Virulencia
11.
Nat Microbiol ; 5(4): 620-629, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32218510

RESUMEN

CRISPR-Cas adaptive immune systems protect bacteria and archaea against their invading genetic parasites, including bacteriophages/viruses and plasmids. In response to this immunity, many phages have anti-CRISPR (Acr) proteins that inhibit CRISPR-Cas targeting. To date, anti-CRISPR genes have primarily been discovered in phage or prophage genomes. Here, we uncovered acr loci on plasmids and other conjugative elements present in Firmicutes using the Listeria acrIIA1 gene as a marker. The four identified genes, found in Listeria, Enterococcus, Streptococcus and Staphylococcus genomes, can inhibit type II-A SpyCas9 or SauCas9, and are thus named acrIIA16-19. In Enterococcus faecalis, conjugation of a Cas9-targeted plasmid was enhanced by anti-CRISPRs derived from Enterococcus conjugative elements, highlighting a role for Acrs in the dissemination of plasmids. Reciprocal co-immunoprecipitation showed that each Acr protein interacts with Cas9, and Cas9-Acr complexes were unable to cleave DNA. Northern blotting suggests that these anti-CRISPRs manipulate single guide RNA length, loading or stability. Mirroring their activity in bacteria, AcrIIA16 and AcrIIA17 provide robust and highly potent broad-spectrum inhibition of distinct Cas9 proteins in human cells (for example, SpyCas9, SauCas9, SthCas9, NmeCas9 and CjeCas9). This work presents a focused analysis of non-phage Acr proteins, demonstrating a role in horizontal gene transfer bolstered by broad-spectrum CRISPR-Cas9 inhibition.


Asunto(s)
Proteína 9 Asociada a CRISPR/antagonistas & inhibidores , Sistemas CRISPR-Cas , Transferencia de Gen Horizontal , Plásmidos/metabolismo , ARN Guía de Kinetoplastida/antagonistas & inhibidores , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Conjugación Genética , ADN/antagonistas & inhibidores , ADN/genética , ADN/metabolismo , Enterococcus/genética , Enterococcus/virología , Células HEK293 , Humanos , Listeria/genética , Listeria/virología , Plásmidos/química , Unión Proteica , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Staphylococcus/genética , Staphylococcus/virología , Streptococcus/genética , Streptococcus/virología
12.
Arq. Inst. Biol ; 87: e0812019, 2020. tab
Artículo en Inglés | LILACS, VETINDEX | ID: biblio-1130055

RESUMEN

Food prepared with products derived from animals are involved in most cases of staphylococcal poisoning; therefore, the research of Staphylococcus spp. in Emmental cheese is more applicable. The objective of this study was to identify coagulase-negative Staphylococcus spp. (CNS) in cheese using biochemical and molecular techniques to detect the presence of nine genes responsible for the production of enterotoxins. From 180 samples analyzed, 204 CNS strains were obtained and identified as being 46 (22.6%) S. saprophyticus strains, 27 (13.2%) S. hominis spp. hominis strains, 22 (10.8%) S. sciuri strains, 21 (10.3%) S. xylosus strains, 19 (9.3%) S. epidermidis strains, 19 (9.3%) S. haemolyticus strains, 17 (8.3%) S. lentus strains, 17 (8.3%) S. warneri strains, 11 (5.4%) S. equorum strains and 5 (2.5%) S. cohnni . Using the PCRm protocol, 14 (6.9%) strains with the presence of the genes on the enterotoxin E (SEE)11 (78.6%), J (SEJ) 1 (7%), C (SEC) 1 (7%) and I (SEI) 1 (7%) were detected. Based on the results, the type of package is not interfered of growth and isolated that Staphylococcus spp. in cheese. It was observed that bacteria capacity to produce coagulase cannot be understood as an indicative of enterotoxigenicity; therefore, the CNS should be considered as a target of importance in the epidemiology of staphylococcal intoxications. It can be concluded that CNS need to be included in bacterial foodborne disease research, since the genes responsible for the production of toxins were detected and none of the studied samples presented Staphylococcus spp. counting above the limits allowed by legislation.(AU)


Os alimentos preparados com produtos de origem animal são os mais envolvidos em casos de intoxicação alimentar estafilocócica; portanto a pesquisa do Staphylococcus spp. em queijos tipo Emmental é relevante. O objetivo foi isolar e identificar espécies de Staphylococcus coagulase negativas (CNS)de queijo Emmental acondicionado em vários tipos de embalagem, por meio de técnicas bacteriológicas e bioquímicas e detectar, por PCR, a presença de nove genes responsáveis pela produção de enterotoxinas. Das 180 amostras, foram isoladas 204 cepas de CNS, que foram identificadas por provas bioquímicas como: 46 (22,6%) S. saprophyticus, 27 (13,2%) S. hominis spp. hominis, 22 (10,8%) S. sciuri, 21 (10,3%) S. xylosus, 19 (9,3%) S. epidermidis , 19 (9,3%) S. haemolyticus , 17 (8,3%) S. lentus , 17 (8,3%) S. warneri , 11(5,4%) S. equorum e 5 (2,5%) S. cohnii . Na PCR multiplex, em 14 (6,9%) isolados foi detectada a presença dos genes para enterotoxina E (SEE), em 11 (78,6%) J (SEJ), em 1 (7%) C (SEC) e em 1 (7%) I (SEI). Com base nos resultados, o tipo de embalagem não interferiu na multiplicação dos Staphylococcus spp. isolados dos queijos. Neste estudo, verificou-se que a capacidade para a produção de coagulase pela bactéria não pode ser concebida como indicativa de enterotoxigenicidade, portanto devem-se considerar os CNS como objeto de importância na epidemiologia das intoxicações estafilocócicas, fazendo-se necessária a atenção com relação à pesquisa dos CNS nos alimentos, uma vez que foram detectados genes responsáveis pela produção de toxinas, e nenhuma das amostras apresentou contagem para Staphylococcus spp. acima do limite permitido pela legislação.(AU)


Asunto(s)
Intoxicación Alimentaria Estafilocócica , Staphylococcus/virología , Enterotoxinas , Enfermedades Transmitidas por los Alimentos , Bacterias , Queso , Reacción en Cadena de la Polimerasa , Técnicas Bacteriológicas , Embalaje de Productos , Alimentos de Origen Animal , Inocuidad de los Alimentos , Abastecimiento de Alimentos
13.
Viruses ; 11(12)2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31835553

RESUMEN

Bacteriophages of the significant veterinary pathogen Staphylococcus pseudintermedius are rarely described morphologically and genomically in detail, and mostly include phages of the Siphoviridae family. There is currently no taxonomical classification for phages of this bacterial species. Here we describe a new phage designated vB_SpsS_QT1, which is related to phage 2638A originally described as a Staphylococcus aureus phage. Propagating strain S. aureus 2854 of the latter was reclassified by rpoB gene sequencing as S. pseudintermedius 2854 in this work. Both phages have a narrow but different host range determined on 54 strains. Morphologically, both of them belong to the family Siphoviridae, share the B1 morphotype, and differ from other staphylococcal phage genera by a single long fibre at the terminus of the tail. The complete genome of phage vB_SpsS_QT1 was sequenced with the IonTorrent platform and expertly annotated. Its linear genome with cohesive ends is 43,029 bp long and encodes 60 predicted genes with the typical modular structure of staphylococcal siphophages. A global alignment found the genomes of vB_SpsS_QT1 and 2638A to share 84% nucleotide identity, but they have no significant similarity of nucleotide sequences with other phage genomes available in public databases. Based on the morphological, phylogenetic, and genomic analyses, a novel genus Fibralongavirus in the family Siphoviridae is described with phage species vB_SpsS_QT1 and 2638A.


Asunto(s)
Siphoviridae/clasificación , Staphylococcus/virología , Genes Virales , Genoma Viral , Genómica/métodos , Especificidad del Huésped , Filogenia , Siphoviridae/ultraestructura , Virión/ultraestructura , Replicación Viral
14.
Appl Microbiol Biotechnol ; 103(16): 6809-6823, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31236618

RESUMEN

Following the emergence of antibiotic-resistant bacteria such as methicillin-resistant Staphylococcus aureus (MRSA) and methicillin-resistant Staphylococcus pseudintermedius (MRSP), phage therapy has attracted significant attention as an alternative to antibiotic treatment. Bacteriophages belonging to kayvirus (previously known as Twort-like phages) have broad host range and are strictly lytic in Staphylococcus spp. Previous work revealed that kayvirus ɸSA039 has a host-recognition mechanism distinct from those of other known kayviruses: most of kayviruses use the backbone of wall teichoic acid (WTA) as their receptor; by contrast, ɸSA039 uses the ß-N-acetylglucosamine (ß-GlcNAc) residue in WTA. In this study, we found that ɸSA039 could switch its receptor to be able to infect S. aureus lacking the ß-GlcNAc residue by acquiring a spontaneous mutation in open reading frame (ORF) 100 and ORF102. Moreover, ɸSA039 could infect S. pseudintermedius, which has a different WTA structure than S. aureus. By comparison, with newly isolated S. pseudintermedius-specific phage (SP phages), we determined that glycosylation in WTA of S. pseudintermedius is essential for adsorption of SP phages, but not ɸSA039. Finally, we describe a novel strategy of S. aureus which protects the bacteria from infection of SP phages. Notably, glycosylation of ribitol phosphate (RboP) WTA by TarM or/and TarS prevents infection of S. aureus by SP phages. These findings could help to establish a new strategy for the treatment of S. aureus and S. pseudintermedius infection, as well as provide valuable insights into the biology of phage-host interactions.


Asunto(s)
Fagos de Staphylococcus/fisiología , Staphylococcus/virología , Interferencia Viral , Acoplamiento Viral , Receptores Virales/metabolismo , Ácidos Teicoicos/metabolismo
15.
Microbiol Spectr ; 7(3)2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31172913

RESUMEN

Pathogenicity islands are members of a vast collection of genomic islands that encode important virulence, antibiotic resistance and other accessory functions and have a critical role in bacterial gene transfer. Staphylococcus aureus is host to a large family of such islands, known as SaPIs, which encode super antigen and other virulence determinants, are mobilized by helper phages and transferred at extremely high frequencies. They benefit their host cells by interfering with phage predation and enhancing horizontal gene transfer. This chapter describes their life cycle, the bases of their phage interference mechanisms, their transfer system and their conversion to antibacterial agents for treatment ofstaphylococcal infections.


Asunto(s)
Islas Genómicas/genética , Staphylococcus/genética , Staphylococcus/fisiología , Factores de Virulencia/genética , Animales , Bacteriófagos/genética , Farmacorresistencia Bacteriana/genética , Transferencia de Gen Horizontal , Genes Bacterianos/genética , Genoma Bacteriano , Humanos , Infecciones Estafilocócicas/microbiología , Staphylococcus/virología , Staphylococcus aureus/genética , Virulencia/genética
16.
Appl Environ Microbiol ; 85(11)2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30902858

RESUMEN

Bacteria in the genus Staphylococcus are important targets for phage therapy due to their prevalence as pathogens and increasing antibiotic resistance. Here we review Staphylococcus outer surface features and specific phage resistance mechanisms that define the host range, the set of strains that an individual phage can potentially infect. Phage infection goes through five distinct phases: attachment, uptake, biosynthesis, assembly, and lysis. Adsorption inhibition, encompassing outer surface teichoic acid receptor alteration, elimination, or occlusion, limits successful phage attachment and entry. Restriction-modification systems (in particular, type I and IV systems), which target phage DNA inside the cell, serve as the major barriers to biosynthesis as well as transduction and horizontal gene transfer between clonal complexes and species. Resistance to late stages of infection occurs through mechanisms such as assembly interference, in which staphylococcal pathogenicity islands siphon away superinfecting phage proteins to package their own DNA. While genes responsible for teichoic acid biosynthesis, capsule, and restriction-modification are found in most Staphylococcus strains, a variety of other host range determinants (e.g., clustered regularly interspaced short palindromic repeats, abortive infection, and superinfection immunity) are sporadic. The fitness costs of phage resistance through teichoic acid structure alteration could make staphylococcal phage therapies promising, but host range prediction is complex because of the large number of genes involved, and the roles of many of these are unknown. In addition, little is known about the genetic determinants that contribute to host range expansion in the phages themselves. Future research must identify host range determinants, characterize resistance development during infection and treatment, and examine population-wide genetic background effects on resistance selection.


Asunto(s)
Especificidad del Huésped , Fagos de Staphylococcus/fisiología , Staphylococcus/virología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Transferencia de Gen Horizontal , Islas Genómicas/genética , Interacciones Huésped-Patógeno , Proteínas de la Membrana , Terapia de Fagos , Staphylococcus/genética , Fagos de Staphylococcus/genética , Ácidos Teicoicos , Ensamble de Virus
17.
Res Vet Sci ; 122: 81-85, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30468880

RESUMEN

We aimed to isolate and characterize bacteriophages (phages) with preferential activity against methicillin-resistant Staphylococcus pseudintermedius (MRSP), a multidrug-resistant canine pathogen. Four phages were isolated from canine faeces using two MRSP strains as initial hosts. Phage host range was evaluated by the spot test on 17 MRSP, 43 methicillin-susceptible S. pseudintermedius (MSSP), and six other staphylococci isolated from dogs. Transmission electron microscopy was used for presumptive identification followed by whole genome sequencing (WGS). All phages lysed all MRSP isolates whereas only 16-28% of MSSP were lysed. Their lytic activity was limited to S. pseudintermedius and S. schleiferi. All phages had similar morphology and belonged to the Siphoviridae family. WGS indicated that the phages were 93.8-99.7% identical to each other, and exhibited the highest similarity (87%) to the temperate S. aureus phage 187. Confirmatory lytic activity tests showed that phages were able to produce clear plaques on lysogens, which was enabled by recombination of the lysogeny modules as shown by WGS of the phages after propagation and plaque formation. This study provides insight into the genetic diversity and biology of S. pseudintermedius temperate phages, which could be further developed for topical therapy of MRSP skin and wound infections.


Asunto(s)
Bacteriófagos/fisiología , Resistencia a la Meticilina , Meticilina/farmacología , Staphylococcus/efectos de los fármacos , Staphylococcus/virología , Animales , Antibacterianos/farmacología , Enfermedades de los Perros/tratamiento farmacológico , Perros , Infecciones Estafilocócicas/veterinaria , Staphylococcus/genética
18.
Appl Environ Microbiol ; 84(15)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29776929

RESUMEN

Treatment of infections caused by staphylococci has become more difficult because of the emergence of multidrug-resistant strains as well as biofilm formation. In this study, we observed the ability of the phage lysin LysGH15 to eliminate staphylococcal planktonic cells and biofilms formed by Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus haemolyticus, and Staphylococcus hominis All these strains were sensitive to LysGH15, showing reductions in bacterial counts of approximately 4 log units within 30 min after treatment with 20 µg/ml of LysGH15, and the MICs ranged from 8 µg/ml to 32 µg/ml. LysGH15 efficiently prevented biofilm formation by the four staphylococcal species at a dose of 50 µg/ml. At a higher dose (100 µg/ml), LysGH15 also showed notable disrupting activity against 24-h and 72-h biofilms formed by S. aureus and coagulase-negative species. In the in vivo experiments, a single intraperitoneal injection of LysGH15 (20 µg/mouse) administered 1 h after the injection of S. epidermidis at double the minimum lethal dose was sufficient to protect the mice. The S. epidermidis cell counts were 4 log units lower in the blood and 3 log units lower in the organs of mice 24 h after treatment with LysGH15 than in the untreated control mice. LysGH15 reduced cytokine levels in the blood and improved pathological changes in the organs. The broad antistaphylococcal activity exerted by LysGH15 on planktonic cells and biofilms makes LysGH15 a valuable treatment option for biofilm-related or non-biofilm-related staphylococcal infections.IMPORTANCE Most staphylococcal species are major causes of health care- and community-associated infections. In particular, Staphylococcus aureus is a common and dangerous pathogen, and Staphylococcus epidermidis is a ubiquitous skin commensal and opportunistic pathogen. Treatment of infections caused by staphylococci has become more difficult because of the emergence of multidrug-resistant strains as well as biofilm formation. In this study, we found that all tested S. aureus, S. epidermidis, Staphylococcus haemolyticus, and Staphylococcus hominis strains were sensitive to the phage lysin LysGH15 (MICs ranging from 8 to 32 µg/ml). More importantly, LysGH15 not only prevented biofilm formation by these staphylococci but also disrupted 24-h and 72-h biofilms. Furthermore, the in vivo efficacy of LysGH15 was demonstrated in a mouse model of S. epidermidis bacteremia. Thus, LysGH15 exhibits therapeutic potential for treating biofilm-related or non-biofilm-related infections caused by diverse staphylococci.


Asunto(s)
Biopelículas , Terapia de Fagos , Plancton/fisiología , Plancton/virología , Infecciones Estafilocócicas/terapia , Fagos de Staphylococcus/fisiología , Staphylococcus/fisiología , Staphylococcus/virología , Animales , Bacteriemia/microbiología , Bacteriemia/terapia , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Plancton/genética , Plancton/crecimiento & desarrollo , Infecciones Estafilocócicas/microbiología , Staphylococcus/genética , Staphylococcus/crecimiento & desarrollo
19.
Viruses ; 10(4)2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29584701

RESUMEN

The use of bacteriophages for killing pathogenic bacteria is a feasible alternative to antibiotics and disinfectants. To obtain the large quantities of phages required for this application, large-scale production of bacteriophages must be optimized. This study aims to define conditions that maximize the phage yield of the virulent and polyvalent staphylococcal bacteriophage vB_SauM-phiIPLA-RODI in broth culture, using the food-grade species Staphylococcus xylosus as the host strain to reduce the risk of growing massive quantities of pathogenic bacteria and therefore, to ensure the safety of the final phage stock. The effect of four variables, namely initial bacterial concentration (5.66-8.40 log10 colony-forming unit (CFU)/mL), initial phage concentration (5-8 log10 plaque-forming unit (PFU)/mL), temperature (21-40 °C) and agitation (20-250 rpm), on phage yield (response) was studied by using response surface methodology (RSM). Successive experimental designs showed that agitation did not significantly impact phage yield, while temperature did have a significant effect, with 38 °C being the optimum for phage propagation. The results allowed the design of a model to describe phage yield as a function of the initial bacterial and phage concentrations at fixed agitation (135 rpm), and optimum temperature (38 °C). The maximum experimental phage yield obtained was 9.3 log10 PFU/mL, while that predicted by the model under the optimized conditions (7.07 log10 CFU/mL initial bacterial population and 6.00 log10 PFU/mL initial phage titer) was 9.25 ± 0.30 log10 PFU/mL, with the desirability of 0.96. This yield is comparable to that obtained when the phage was propagated on the original host, Staphylococcus aureus. Bacteriophage phiIPLA-RODI showed the same host range and very similar biofilm removal ability regardless of the staphylococcal species used for its propagation. The results presented in this study show the suitability of using a food-grade strain of S. xylosus for the propagation of S. aureus infecting phages and the application of RSM to define the optimal propagation conditions.


Asunto(s)
Fermentación , Modelos Biológicos , Fagos de Staphylococcus/crecimiento & desarrollo , Staphylococcus/virología , Análisis de Varianza , Antibacterianos , Biopelículas/crecimiento & desarrollo , Recuento de Colonia Microbiana , Medios de Cultivo , Especificidad del Huésped , Reproducibilidad de los Resultados , Staphylococcus/clasificación , Staphylococcus/crecimiento & desarrollo , Fagos de Staphylococcus/fisiología , Temperatura , Ensayo de Placa Viral
20.
Virus Genes ; 54(1): 130-139, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28852930

RESUMEN

The spontaneous host-range mutants 812F1 and K1/420 are derived from polyvalent phage 812 that is almost identical to phage K, belonging to family Myoviridae and genus Kayvirus. Phage K1/420 is used for the phage therapy of staphylococcal infections. Endolysin of these mutants designated LysF1, consisting of an N-terminal cysteine-histidine-dependent aminohydrolase/peptidase (CHAP) domain and C-terminal SH3b cell wall-binding domain, has deleted middle amidase domain compared to wild-type endolysin. In this work, LysF1 and both its domains were prepared as recombinant proteins and their function was analyzed. LysF1 had an antimicrobial effect on 31 Staphylococcus species of the 43 tested. SH3b domain influenced antimicrobial activity of LysF1, since the lytic activity of the truncated variant containing the CHAP domain alone was decreased. The results of a co-sedimentation assay of SH3b domain showed that it was able to bind to three types of purified staphylococcal peptidoglycan 11.2, 11.3, and 11.8 that differ in their peptide bridge, but also to the peptidoglycan type 11.5 of Streptococcus uberis, and this capability was verified in vivo using the fusion protein with GFP and fluorescence microscopy. Using several different approaches, including NMR, we have not confirmed the previously proposed interaction of the SH3b domain with the pentaglycine bridge in the bacterial cell wall. The new naturally raised deletion mutant endolysin LysF1 is smaller than LysK, has a broad lytic spectrum, and therefore is an appropriate enzyme for practical use. The binding spectrum of SH3b domain covering all known staphylococcal peptidoglycan types is a promising feature for creating new chimeolysins by combining it with more effective catalytic domains.


Asunto(s)
Endopeptidasas/genética , Endopeptidasas/metabolismo , Especificidad del Huésped , Myoviridae/enzimología , Peptidoglicano/metabolismo , Eliminación de Secuencia , Staphylococcus/virología , Endopeptidasas/aislamiento & purificación , Proteínas Mutantes/genética , Proteínas Mutantes/aislamiento & purificación , Proteínas Mutantes/metabolismo , Myoviridae/genética , Myoviridae/fisiología , Unión Proteica , Dominios Proteicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...