Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Nucleic Acids Res ; 51(14): 7438-7450, 2023 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-37293964

RESUMO

The Streptococcus pyogenes type II-A CRISPR-Cas systems provides adaptive immunity through the acquisition of short DNA sequences from invading viral genomes, called spacers. Spacers are transcribed into short RNA guides that match regions of the viral genome followed by a conserved NGG DNA motif, known as the PAM. These RNA guides, in turn, are used by the Cas9 nuclease to find and destroy complementary DNA targets within the viral genome. While most of the spacers present in bacterial populations that survive phage infection target protospacers flanked by NGG sequences, there is a small fraction that target non-canonical PAMs. Whether these spacers originate through accidental acquisition of phage sequences and/or provide efficient defense is unknown. Here we found that many of them match phage target regions flanked by an NAGG PAM. Despite being scarcely present in bacterial populations, NAGG spacers provide substantial immunity in vivo and generate RNA guides that support robust DNA cleavage by Cas9 in vitro; with both activities comparable to spacers that target sequences followed by the canonical AGG PAM. In contrast, acquisition experiments showed that NAGG spacers are acquired at very low frequencies. We therefore conclude that discrimination against these sequences occurs during immunization of the host. Our results reveal unexpected differences in PAM recognition during the spacer acquisition and targeting stages of the type II-A CRISPR-Cas immune response.


Assuntos
Bacteriófagos , Sistemas CRISPR-Cas , Streptococcus pyogenes , Bacteriófagos/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Sistemas CRISPR-Cas/genética , Motivos de Nucleotídeos , Streptococcus pyogenes/fisiologia , Streptococcus pyogenes/virologia
2.
Nat Microbiol ; 6(12): 1516-1525, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34819640

RESUMO

CRISPR loci are composed of short DNA repeats separated by sequences, known as spacers, that match the genomes of invaders such as phages and plasmids. Spacers are transcribed and processed to generate RNA guides used by CRISPR-associated nucleases to recognize and destroy the complementary nucleic acids of invaders. To counteract this defence, phages can produce small proteins that inhibit these nucleases, termed anti-CRISPRs (Acrs). Here we demonstrate that the ΦAP1.1 temperate phage utilizes an alternative approach to antagonize the type II-A CRISPR response in Streptococcus pyogenes. Immediately after infection, this phage expresses a small anti-CRISPR protein, AcrIIA23, that prevents Cas9 function, allowing ΦAP1.1 to integrate into the direct repeats of the CRISPR locus, neutralizing immunity. However, acrIIA23 is not transcribed during lysogeny and phage integration/excision cycles can result in the deletion and/or transduction of spacers, enabling a complex modulation of the type II-A CRISPR immune response. A bioinformatic search identified prophages integrated not only in the CRISPR repeats, but also the cas genes, of diverse bacterial species, suggesting that prophage disruption of the CRISPR-cas locus is a recurrent mechanism to counteract immunity.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Prófagos/fisiologia , Fagos de Streptococcus/fisiologia , Streptococcus pyogenes/imunologia , Streptococcus pyogenes/virologia , Lisogenia , Plasmídeos/genética , Plasmídeos/metabolismo , Prófagos/genética , Fagos de Streptococcus/genética , Streptococcus pyogenes/genética , Integração Viral
3.
Viruses ; 13(4)2021 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-33918348

RESUMO

Bacteriophages exert strong evolutionary pressure on their microbial hosts. In their lytic lifecycle, complete bacterial subpopulations are utilized as hosts for bacteriophage replication. However, during their lysogenic lifecycle, bacteriophages can integrate into the host chromosome and alter the host's genomic make-up, possibly resulting in evolutionary important adjustments. Not surprisingly, bacteria have evolved sophisticated immune systems to protect against phage infection. Streptococcus pyogenes isolates are frequently lysogenic and their prophages have been shown to be major contributors to the virulence of this pathogen. Most S. pyogenes phage research has focused on genomic prophages in relation to virulence, but little is known about the defensive arsenal of S. pyogenes against lytic phage infection. Here, we characterized Phage A1, an S. pyogenes bacteriophage, and investigated several mechanisms that S. pyogenes utilizes to protect itself against phage predation. We show that Phage A1 belongs to the Siphoviridae family and contains a circular double-stranded DNA genome that follows a modular organization described for other streptococcal phages. After infection, the Phage A1 genome can be detected in isolated S. pyogenes survivor strains, which enables the survival of the bacterial host and Phage A1 resistance. Furthermore, we demonstrate that the type II-A CRISPR-Cas system of S. pyogenes acquires new spacers upon phage infection, which are increasingly detectable in the absence of a capsule. Lastly, we show that S. pyogenes produces membrane vesicles that bind to phages, thereby limiting the pool of phages available for infection. Altogether, this work provides novel insight into survival strategies employed by S. pyogenes to combat phage predation.


Assuntos
Viabilidade Microbiana , Fagos de Streptococcus/genética , Fagos de Streptococcus/patogenicidade , Streptococcus pyogenes/fisiologia , Streptococcus pyogenes/virologia , Sistemas CRISPR-Cas , Genoma Viral , Lisogenia , Prófagos/genética , Virulência
4.
Mol Microbiol ; 116(2): 397-415, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33756056

RESUMO

Endolysin enzymes from bacteriophage cause bacterial lysis by degrading the peptidoglycan cell wall. The streptococcal C1 phage endolysin PlyC, is the most potent endolysin described to date and can rapidly lyse group A, C, and E streptococci. PlyC is known to bind the Group A streptococcal cell wall, but the specific molecular target or the binding site within PlyC remain uncharacterized. Here we report for the first time, that the polyrhamnose backbone of the Group A streptococcal cell wall is the binding target of PlyC. We have also characterized the putative rhamnose binding groove of PlyC and found four key residues that were critical to either the folding or the cell wall binding action of PlyC. Based on our results, we suggest that the interaction between PlyC and the cell wall may not be a high-affinity interaction as previously proposed, but rather a high avidity one, allowing for PlyC's remarkable lytic activity. Resistance to our current antibiotics is reaching crisis levels and there is an urgent need to develop the antibacterial agents with new modes of action. A detailed understanding of this potent endolysin may facilitate future developments of PlyC as a tool against the rise of antibiotic resistance.


Assuntos
Bacteriófagos/metabolismo , Endopeptidases/metabolismo , Peptidoglicano/metabolismo , Ramnose/metabolismo , Streptococcus pyogenes/virologia , Bacteriófagos/genética , Sítios de Ligação/fisiologia , Membrana Celular/metabolismo , Parede Celular/metabolismo , Endopeptidases/genética , Simulação de Acoplamento Molecular , Ligação Proteica/fisiologia , Streptococcus pyogenes/metabolismo
5.
Microb Genom ; 7(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33245690

RESUMO

The major human pathogen Streptococcus pyogenes shares an intimate evolutionary history with mobile genetic elements, which in many cases carry genes encoding bacterial virulence factors. During recent whole-genome sequencing of a longitudinal sample of S. pyogenes isolates in England, we identified a lineage within emm4 that clustered with the reference genome MEW427. Like MEW427, this lineage was characterized by substantial gene loss within all three prophage regions, compared to MGAS10750 and isolates outside of the MEW427-like lineage. Gene loss primarily affected lysogeny, replicative and regulatory modules, and to a lesser and more variable extent, structural genes. Importantly, prophage-encoded superantigen and DNase genes were retained in all isolates. In isolates where the prophage elements were complete, like MGAS10750, they could be induced experimentally, but not in MEW427-like isolates with degraded prophages. We also found gene loss within the chromosomal island SpyCIM4 of MEW427-like isolates, although surprisingly, the SpyCIM4 element could not be experimentally induced in either MGAS10750-like or MEW427-like isolates. This did not, however, appear to abolish expression of the mismatch repair operon, within which this element resides. The inclusion of further emm4 genomes in our analyses ratified our observations and revealed an international emm4 lineage characterized by prophage degradation. Intriguingly, the USA population of emm4 S. pyogenes appeared to constitute predominantly MEW427-like isolates, whereas the UK population comprised both MEW427-like and MGAS10750-like isolates. The degraded and cryptic nature of these elements may have important phenotypic and fitness ramifications for emm4 S. pyogenes, and the geographical distribution of this lineage raises interesting questions on the population dynamics of the genotype.


Assuntos
Bacteriófagos/genética , Análise de Sequência de DNA/métodos , Streptococcus pyogenes/classificação , Proteínas Virais/genética , Deleção de Genes , Genoma Bacteriano , Genótipo , Filogenia , Streptococcus pyogenes/genética , Streptococcus pyogenes/virologia , Estados Unidos
6.
Nat Commun ; 11(1): 5018, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33024089

RESUMO

The re-emergence of scarlet fever poses a new global public health threat. The capacity of North-East Asian serotype M12 (emm12) Streptococcus pyogenes (group A Streptococcus, GAS) to cause scarlet fever has been linked epidemiologically to the presence of novel prophages, including prophage ΦHKU.vir encoding the secreted superantigens SSA and SpeC and the DNase Spd1. Here, we report the molecular characterization of ΦHKU.vir-encoded exotoxins. We demonstrate that streptolysin O (SLO)-induced glutathione efflux from host cellular stores is a previously unappreciated GAS virulence mechanism that promotes SSA release and activity, representing the first description of a thiol-activated bacterial superantigen. Spd1 is required for resistance to neutrophil killing. Investigating single, double and triple isogenic knockout mutants of the ΦHKU.vir-encoded exotoxins, we find that SpeC and Spd1 act synergistically to facilitate nasopharyngeal colonization in a mouse model. These results offer insight into the pathogenesis of scarlet fever-causing GAS mediated by prophage ΦHKU.vir exotoxins.


Assuntos
Exotoxinas/metabolismo , Prófagos/genética , Streptococcus pyogenes/patogenicidade , Streptococcus pyogenes/virologia , Animais , Proteínas de Bactérias/farmacologia , Linhagem Celular , Eritrócitos/efeitos dos fármacos , Exotoxinas/genética , Feminino , Glutationa/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Faringe/citologia , Escarlatina/epidemiologia , Escarlatina/microbiologia , Streptococcus pyogenes/genética , Estreptolisinas/farmacologia , Superantígenos/genética , Superantígenos/metabolismo
7.
Sci Rep ; 10(1): 11656, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32669560

RESUMO

We present the first complete, closed genome sequences of Streptococcus pyogenes strains NCTC 8198T and CCUG 4207T, the type strain of the type species of the genus Streptococcus and an important human pathogen that causes a wide range of infectious diseases. S. pyogenes NCTC 8198T and CCUG 4207T are derived from deposit of the same strain at two different culture collections. NCTC 8198T was sequenced, using a PacBio platform; the genome sequence was assembled de novo, using HGAP. CCUG 4207T was sequenced and a de novo hybrid assembly was generated, using SPAdes, combining Illumina and Oxford Nanopore sequence reads. Both strategies yielded closed genome sequences of 1,914,862 bp, identical in length and sequence identity. Combining short-read Illumina and long-read Oxford Nanopore sequence data circumvented the expected error rate of the nanopore sequencing technology, producing a genome sequence indistinguishable to the one determined with PacBio. Sequence analyses revealed five prophage regions, a CRISPR-Cas system, numerous virulence factors and no relevant antibiotic resistance genes. These two complete genome sequences of the type strain of S. pyogenes will effectively serve as valuable taxonomic and genomic references for infectious disease diagnostics, as well as references for future studies and applications within the genus Streptococcus.


Assuntos
Mapeamento Cromossômico , DNA Bacteriano/genética , Sequenciamento de Nucleotídeos em Larga Escala , Streptococcus pyogenes/genética , Fatores de Virulência/genética , Sequenciamento Completo do Genoma/métodos , Técnicas de Tipagem Bacteriana , Sequência de Bases , Sistemas CRISPR-Cas , DNA Bacteriano/metabolismo , Genoma Bacteriano , Nanoporos , Prófagos/genética , Análise de Sequência de DNA , Streptococcus pyogenes/classificação , Streptococcus pyogenes/virologia , Fatores de Virulência/metabolismo
8.
PLoS One ; 15(6): e0235002, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32574197

RESUMO

Streptococcus agalactiae or Group B Streptococcus (GBS) is a leading cause of sepsis in neonates. As a preventative measure prophylactic antibiotic administration is common in pregnant women colonised with GBS, but antibiotic-resistance and adverse effects on neonatal microbiomes may result. Use of bacteriophages (phages) is one option for targeted therapy. To this end, four phages (LF1 -LF4) were isolated from wastewater. They displayed lytic activity in vitro against S. agalactiae isolates collected from pregnant women and neonates, with 190/246 isolates (77.2%) and 10/10 (100%) isolates susceptible to at least one phage, respectively. Phage genomes ranged from 32,205-44,768 bp and all phages were members of the Siphoviridae family. High nucleotide identity (99.9%) was observed between LF1 and LF4, which were closely related to a putative prophage of S. agalactiae. The genome organisation of LF2 differed, and it showed similarity to a different S. agalactiae prophage, while LF3 was more closely related to a Streptococcus pyogenes phage. Lysogenic gene presence (integrase, repressor and regulatory modules), was suggestive of temperate phages. In a therapeutic context, temperate phages are not ideal candidates, however, the broad host range activity of these phages observed on clinical isolates in vitro is promising for future therapeutic approaches including bioengineered phage or lysin applications.


Assuntos
Sepse Neonatal/terapia , Terapia por Fagos , Siphoviridae/genética , Fagos de Streptococcus/genética , Streptococcus agalactiae/virologia , DNA Viral/isolamento & purificação , Feminino , Genômica , Especificidade de Hospedeiro/genética , Humanos , Recém-Nascido , Lisogenia , Sepse Neonatal/microbiologia , Filogenia , Gravidez , Siphoviridae/isolamento & purificação , Fagos de Streptococcus/isolamento & purificação , Streptococcus agalactiae/isolamento & purificação , Streptococcus pyogenes/virologia
10.
Acta Otolaryngol ; 140(3): 220-224, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32049553

RESUMO

Background: We previously described that adenoid tissue in children with chronic otitis media (COM) contained more mucosal biofilms than adenoid tissue removed for hypertrophy.Aims/objectives: The aim of the second part was to characterize nasopharyngeal microbiota and explore virulence of the most common middle ear pathogens.Material and methods: Bacteriological analysis was performed following a culture-based approach on the samples recovered from 30 patients of COM group (15 biofilm-positive and 15 biofilm-negative) and from 30 patients of a control group (15 biofilm-positive and 15 biofilm-negative). Virulence factors of Streptococcus pneumoniae, Streptococcus pyogenes, and Haemophilus influenzae were investigated.Results: The most frequent species were Firmicutes followed by Proteobacteria and Actinobacteria. The presence of biofilm was statistically associated with an increase of the number of bacterial species and Firmicutes phylum regardless of the condition (case/control). No virulence factors associated with invasive isolates were found for the most common middle ear pathogens.Conclusions and significance: This case-control study demonstrated that the presence of COM plus biofilm was associated with a given microbiota which contained more Firmicutes. Our study allows a better understanding of physiopathological mechanisms involved in chronic otitis media and paves the way for further investigations.


Assuntos
Tonsila Faríngea/microbiologia , Bactérias/isolamento & purificação , Otite Média/microbiologia , Análise de Variância , Biofilmes , Estudos de Casos e Controles , Criança , Doença Crônica , Farmacorresistência Bacteriana , Firmicutes/isolamento & purificação , Haemophilus influenzae/isolamento & purificação , Haemophilus influenzae/virologia , Humanos , Testes de Sensibilidade Microbiana , Nasofaringe/microbiologia , Staphylococcus aureus/isolamento & purificação , Streptococcus pneumoniae/isolamento & purificação , Streptococcus pneumoniae/virologia , Streptococcus pyogenes/isolamento & purificação , Streptococcus pyogenes/virologia
11.
Curr Issues Mol Biol ; 32: 435-472, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31166177

RESUMO

Streptococcus pyogenes (Group A Streptococcus, GAS) is a strictly human bacterial pathogen. Since the mid-1980s, GAS M1T1 clone has been the most prevalent and globally disseminated serotype and is the culprit causing invasive and severe streptococcal infections, urging a better understanding of the emergence of hypervirulent M1T1 clone from an evolutionary perspective. This review highlights the molecular and evolutionary events leading to pandemic M1T1 strains, and discusses the pressure driving the genetic acquisition of novel virulence genes and the selection of hypervirulent isolates in host. By understanding the evolutionary selection and pressures that select and shape the pandemic M1T1 clone, we could potentially develop new therapeutic strategies to tackle challenges when dealing with the globally disseminated M1T1 GAS clone.


Assuntos
Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Pandemias , Infecções Estreptocócicas/epidemiologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidade , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células Clonais , Citotoxinas/genética , Citotoxinas/metabolismo , Desoxirribonuclease I/genética , Desoxirribonuclease I/metabolismo , Evolução Molecular , Exotoxinas/genética , Exotoxinas/metabolismo , Interações Hospedeiro-Patógeno/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Recombinação Genética , Seleção Genética , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Fagos de Streptococcus/genética , Fagos de Streptococcus/metabolismo , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/virologia , Transdução Genética , Virulência
12.
Microbiol Spectr ; 7(3)2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31111820

RESUMO

The bacteriophages of Streptococcus pyogenes (group A streptococcus) play a key role in population shaping, genetic transfer, and virulence of this bacterial pathogen. Lytic phages like A25 can alter population distributions through elimination of susceptible serotypes but also serve as key mediators for genetic transfer of virulence genes and antibiotic resistance via generalized transduction. The sequencing of multiple S. pyogenes genomes has uncovered a large and diverse population of endogenous prophages that are vectors for toxins and other virulence factors and occupy multiple attachment sites in the bacterial genomes. Some of these sites for integration appear to have the potential to alter the bacterial phenotype through gene disruption. Remarkably, the phage-like chromosomal islands (SpyCI), which share many characteristics with endogenous prophages, have evolved to mediate a growth-dependent mutator phenotype while acting as global transcriptional regulators. The diverse population of prophages appears to share a large pool of genetic modules that promotes novel combinations that may help disseminate virulence factors to different subpopulations of S. pyogenes. The study of the bacteriophages of this pathogen, both lytic and lysogenic, will continue to be an important endeavor for our understanding of how S. pyogenes continues to be a significant cause of human disease.


Assuntos
Bacteriófagos/genética , Streptococcus pyogenes/genética , Streptococcus pyogenes/virologia , Toxinas Bacterianas/genética , Farmacorresistência Bacteriana/genética , Regulação Bacteriana da Expressão Gênica , Transferência Genética Horizontal , Genoma Bacteriano , Genoma Viral , Humanos , Fenótipo , Prófagos/genética , Sorogrupo , Transdução Genética , Virulência/genética , Fatores de Virulência/genética
13.
BMC Microbiol ; 19(1): 24, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30691408

RESUMO

BACKGROUND: Group A Streptococcus (GAS) is a major human pathogen, which is associated with a wide spectrum of invasive diseases, such as pharyngitis, scarlet fever, rheumatic fever, and streptococcal toxic shock syndrome (STSS). It is hypothesized that differences in GAS pathogenicity are related to the acquisition of diverse bacteriophages (phages). Nevertheless, the GAS genome also harbors clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated (cas) genes, which play an important role in eliminating foreign DNA, including those of phages. However, the structure of prophages in GAS strains is mosaic, and the phylogenetic relationship between prophages and CRISPR is not clear. In this study, we analyzed CRISPR and prophage structure using 118 complete genome sequences of GAS strains to elucidate the relationship between two genomic elements. Additionally, phylogenetic and M-type analyses were performed. RESULTS: Of the 118 GAS strains, 80 harbored type I-C and/or II-A CRISPR/cas loci. A total of 553 spacer sequences were identified from CRISPR/cas loci and sorted into 229 patterns. We identified and classified 373 prophages into 14 groups. Some prophage groups shared a common integration site, and were related to M-type. We further investigated the correlation between spacer sequences and prophages. Of the 229 spacer sequence patterns, 203 were similar to that of other GAS prophages. No spacer showed similarity with that of a specific prophage group with mutL integration site. Moreover, the average number of prophages in strains with type II-A CRISPR was significantly less than that in type I-C CRISPR and non-CRISPR strains. However, there was no statistical difference between the average number of prophages in type I-C strains and that in non-CRISPR strains. CONCLUSIONS: Our results indicated that type II-A CRISPR may play an important role in eliminating phages and that the prophage integration site may be an important criterion for the acceptance of foreign DNA by GAS. M type, spacer sequence, and prophage group data were correlated with the phylogenetic relationships of GAS. Therefore, we hypothesize that genetic characteristics and/or phylogenetic relationships of GAS may be estimated by analyzing its spacer sequences.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Filogenia , Prófagos/classificação , Streptococcus pyogenes/genética , Evolução Molecular , Genoma Bacteriano , Streptococcus pyogenes/virologia , Integração Viral
14.
PLoS One ; 13(10): e0205995, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30339682

RESUMO

Only 3% of phage genomes in NCBI nucleotide database represent phages that are active against Streptococcus sp. With the aim to increase general awareness of phage diversity, we isolated two bacteriophages, Str01 and Str03, active against health-threatening Group A Streptococcus (GAS). Both phages are members of the Siphoviridae, but their analysis revealed that Str01 and Str03 do not belong to any known genus. We identified their structural proteins based on LC-ESI29 MS/MS and list their basic thermal stability and physico-chemical features including optimum pH. Annotated genomic sequences of the phages are deposited in GenBank (NCBI accession numbers KY349816 and KY363359, respectively).


Assuntos
Bacteriófagos/genética , Genoma Viral , Streptococcus pyogenes/virologia , Bacteriófagos/isolamento & purificação , Bacteriófagos/ultraestrutura , Genes Virais , Concentração de Íons de Hidrogênio , Espectrometria de Massas , Filogenia , Temperatura , Proteínas Virais/metabolismo , Vírion/genética
15.
Buenos Aires; GCBA. Gerencia Operativa de Epidemiología; 19 oct. 2018. a) f:18 l:24 p. graf.(Boletín Epidemiológico Semanal: Ciudad Autónoma de Buenos Aires, 3, 113).
Monografia em Espanhol | UNISALUD, BINACIS, InstitutionalDB, LILACS | ID: biblio-1103130

RESUMO

Situación nacional de este tipo de infecciones, del que se observa un incremento global en las últimas décadas, en Europa y Norte América, sin que la causa haya sido determinada. En Argentina, los serotipos prevalentes, en base a datos aportados por el Laboratorio Nacional de Referencia- por él identificados en infección invasiva por Streptococcus pyogenes en población general durante el período 2000- 2016- fueron M12, M1, y M. A partir del año 2017 se observa un incremento del aislamiento de serotipo M3 principalmente en población pediátrica y no se registraron resistencias antimicrobianas específicas.Se describe la situación en la Ciudad Autónoma de Buenos Aires, la normativa a cumplir para la vigilancia de estas infecciones, y se incluyen recomendaciones para el personal de salud, la comunidad, y la comunidad educativa


Assuntos
Infecções Estreptocócicas/diagnóstico , Infecções Estreptocócicas/patologia , Infecções Estreptocócicas/prevenção & controle , Infecções Estreptocócicas/epidemiologia , Streptococcus pyogenes/patogenicidade , Streptococcus pyogenes/virologia , Controle de Infecções/métodos , Notificação de Doenças/normas , Monitoramento Epidemiológico
16.
J Bacteriol ; 200(23)2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30224437

RESUMO

Lytic bacteriophage A25, which infects Streptococcus pyogenes and several related species, has been used to better understand phage-microbe interactions due to its ability to mediate high-efficiency transduction. Most of these studies, however, are decades old and were conducted prior to the advent of next-generation sequencing and bioinformatics. The aim of our study was to gain a better understanding of the mechanism of high-efficiency transduction through analysis of the A25 genome. We show here that phage A25 is related to a family of genome prophages and became a lytic phage following escape from lysogeny. A lambdoid-like residual lysogeny module consisting of an operator site with two promoters and a cro-like antirepressor gene was identified, but the genes for the cI-like repressor and integrase are missing. Additionally, the genetic organization of the A25 genome was found to be modular in nature and similar to that of many prophages of S. pyogenes as well as from other streptococcal species. A study of A25 homology to all annotated prophages within S. pyogenes revealed near identity within the remnant lysogeny module of the A25 phage genome to the corresponding regions in resident prophages of genome strains MGAS10270 (M2), MGAS315 (M3), MGAS10570 (M4), and STAB902 (M4). Host range studies of MGAS10270, MGAS315, and MGAS10750 demonstrated that these strains were resistant to A25 infection. The resistance mechanism of superinfection immunity was confirmed experimentally through complementation of the operator region and cI-like repressor from prophage MGAS10270.2 into susceptible strains SF370, CEM1Δ4 (SF370ΔSpyCIM1), and ATCC 12204, which rendered all three strains resistant to A25 infection. In silico prediction of packaging through homology analysis of the terminase large subunit from bacteriophages within the known packaging mechanism of Gram-positive bacteria as well as the evidence of terminally redundant and/or circularly permuted sequences suggested that A25 grouped with phages employing the less stringent pac-type packaging mechanisms, which likely explains the characteristic A25 high-efficiency transduction capabilities. Only a few examples of lytic phages appearing following loss of part or all of the lysogeny module have been reported previously, and the genetic mosaicism of A25 suggests that this event may not have been a recent one. However, the discovery that this lytic bacteriophage shares some of the genetic pool of S. pyogenes prophages emphasizes the importance of genetic and biological characterization of bacteriophages when selecting phages for therapeutics or disinfectants, as phage-phage and phage-microbe interactions can be complex, requiring more than just assessment of host range and carriage of toxoid or virulence genes.IMPORTANCE Bacteriophages (bacterial viruses) play an important role in the shaping of bacterial populations as well as the dissemination of bacterial genetic material to new strains, resulting in the spread of virulence factors and antibiotic resistance genes. This study identified the genetic origins of Streptococcus pyogenes phage A25 and uncovered the molecular mechanism employed to promote horizontal transfer of DNA by transduction to new strains of this bacterium as well as identified the basis for its host range.


Assuntos
Genoma Viral/genética , Prófagos/fisiologia , Fagos de Streptococcus/fisiologia , Streptococcus pyogenes/virologia , Lisogenia , Prófagos/genética , Fagos de Streptococcus/genética , Transdução Genética
17.
Nat Microbiol ; 2(10): 1374-1380, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28785032

RESUMO

The CRISPR-Cas system owes its utility as a genome-editing tool to its origin as a prokaryotic immune system. The first demonstration of its activity against bacterial viruses (phages) is also the first record of phages evading that immunity 1 . This evasion can be due to point mutations 1 , large-scale deletions 2 , DNA modifications 3 , or phage-encoded proteins that interfere with the CRISPR-Cas system, known as anti-CRISPRs (Acrs) 4 . The latter are of biotechnological interest, as Acrs can serve as off switches for CRISPR-based genome editing 5 . Every Acr characterized to date originated from temperate phages, genomic islands, or prophages 4-8 , and shared properties with the first Acr discovered. Here, with a phage-oriented approach, we have identified an unrelated Acr in a virulent phage of Streptococcus thermophilus. In challenging a S. thermophilus strain CRISPR-immunized against a set of virulent phages, we found one that evaded the CRISPR-encoded immunity >40,000× more often than the others. Through systematic cloning of its genes, we identified an Acr solely responsible for the abolished immunity. We extended our findings by demonstrating activity in another S. thermophilus strain, against unrelated phages, and in another bacterial genus immunized using the heterologous SpCas9 system favoured for genome editing. This Acr completely abolishes SpCas9-mediated immunity in our assays.


Assuntos
Sistemas CRISPR-Cas/efeitos dos fármacos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/efeitos dos fármacos , Fagos de Streptococcus/genética , Fagos de Streptococcus/metabolismo , Streptococcus pyogenes/efeitos dos fármacos , Streptococcus pyogenes/virologia , Proteínas Virais/genética , Proteínas Virais/farmacologia , Proteínas de Bactérias/efeitos dos fármacos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Clonagem Molecular , DNA Bacteriano , Escherichia coli/genética , Escherichia coli/imunologia , Escherichia coli/virologia , Edição de Genes , Ilhas Genômicas/genética , Imunidade , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Fenótipo , Mutação Puntual , Prófagos , Streptococcus pyogenes/imunologia , Streptococcus thermophilus/genética , Streptococcus thermophilus/virologia , Transformação Bacteriana , Proteínas Virais/imunologia
18.
Virulence ; 8(7): 1390-1400, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28459299

RESUMO

BACKGROUND: A group A Streptococcus (GAS) lineage of genotype emm3, sequence type 15 (ST15) was associated with a 6 month upsurge in invasive GAS disease in the UK. The epidemic lineage (Lineage C) had lost 2 typical emm3 prophages, Φ315.1 and Φ315.2 associated with the superantigen ssa, but gained a different prophage (ΦUK-M3.1) associated with a different superantigen, speC and a DNAse spd1. METHODS AND RESULTS: The presence of speC and spd1 in Lineage C ST15 strains enhanced both in vitro mitogenic and DNase activities over non-Lineage C ST15 strains. Invasive disease models in Galleria mellonella and SPEC-sensitive transgenic mice, revealed no difference in overall invasiveness of Lineage C ST15 strains compared with non-Lineage C ST15 strains, consistent with clinical and epidemiological analysis. Lineage C strains did however markedly prolong murine nasal infection with enhanced nasal and airborne shedding compared with non-Lineage C strains. Deletion of speC or spd1 in 2 Lineage C strains identified a possible role for spd1 in airborne shedding from the murine nasopharynx. CONCLUSIONS: Nasopharyngeal infection and shedding of Lineage C strains was enhanced compared with non-Lineage C strains and this was, in part, mediated by the gain of the DNase spd1 through prophage acquisition.


Assuntos
Antígenos de Bactérias/genética , Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Transporte/genética , Doenças Nasofaríngeas/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/fisiologia , Animais , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/metabolismo , Feminino , Genótipo , Humanos , Camundongos , Mariposas , Doenças Nasofaríngeas/epidemiologia , Prófagos/genética , Prófagos/fisiologia , Infecções Estreptocócicas/epidemiologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidade , Streptococcus pyogenes/virologia , Reino Unido/epidemiologia , Virulência
19.
BMC Genomics ; 18(1): 224, 2017 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-28283023

RESUMO

BACKGROUND: During a substantial elevation in scarlet fever (SF) notifications in 2014 a national genomic study was undertaken of Streptococcus pyogenes (Group A Streptococci, GAS) isolates from patients with SF with comparison to isolates from patients with invasive disease (iGAS) to test the hypotheses that the increase in SF was due to either the introduction of one or more new/emerging strains in the population in England or the transmission of a known genetic element through the population of GAS by horizontal gene transfer (HGT) resulting in infections with an increased likelihood of causing SF. Isolates were collected to provide geographical representation, for approximately 5% SF isolates from each region from 1st April 2014 to 18th June 2014. Contemporaneous iGAS isolates for which genomic data were available were included for comparison. Data were analysed in order to determine emm gene sequence type, phylogenetic lineage and genomic clade representation, the presence of known prophage elements and the presence of genes known to confer pathogenicity and resistance to antibiotics. RESULTS: 555 isolates were analysed, 303 from patients with SF and 252 from patients with iGAS. Isolates from patients with SF were of multiple distinct emm sequence types and phylogenetic lineages. Prior to data normalisation, emm3 was the predominant type (accounting for 42.9% of SF isolates, 130/303 95%CI 37.5-48.5; 14.7% higher than the percentage of emm3 isolates found in the iGAS isolates). Post-normalisation emm types, 4 and 12, were found to be over-represented in patients with SF versus iGAS (p < 0.001). A single gene, ssa, was over-represented in isolates from patients with SF. No single phage was found to be over represented in SF vs iGAS. However, a "meta-ssa" phage defined by the presence of :315.2, SPsP6, MGAS10750.3 or HK360ssa, was found to be over represented. The HKU360.vir phage was not detected yet the HKU360.ssa phage was present in 43/63 emm12 isolates but not found to be over-represented in isolates from patients with SF. CONCLUSIONS: There is no evidence that the increased number of SF cases was a strain-specific or known mobile element specific phenomenon, as the increase in SF cases was associated with multiple lineages of GAS.


Assuntos
Genoma Bacteriano , Genômica , Escarlatina/microbiologia , Streptococcus pyogenes/genética , Antígenos de Bactérias/genética , Proteínas da Membrana Bacteriana Externa/genética , Bacteriófagos/genética , Proteínas de Transporte/genética , Análise por Conglomerados , Inglaterra/epidemiologia , Transferência Genética Horizontal , Genômica/métodos , Humanos , Tipagem de Sequências Multilocus , Filogenia , Vigilância da População , Escarlatina/epidemiologia , Streptococcus pyogenes/classificação , Streptococcus pyogenes/virologia
20.
Mol Cell ; 64(3): 616-623, 2016 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-27618488

RESUMO

CRISPR-Cas systems defend prokaryotes against viruses and plasmids. Short DNA segments of the invader, known as spacers, are stored in the CRISPR array as immunological memories. New spacers are added invariably to the 5' end of the array; therefore, the first spacer matches the latest foreign threat. Whether this highly polarized order of spacer insertion influences CRISPR-Cas immunity has not been explored. Here we show that a conserved sequence located immediately upstream of the CRISPR array specifies the site of new spacer integration. Mutation of this sequence results in erroneous incorporation of new spacers into the middle of the array. We show that spacers added through polarized acquisition give rise to more robust CRISPR-Cas immunity than spacers added to the middle of the array. This study demonstrates that the CRISPR-Cas system specifies the site of spacer integration to optimize the immune response against the most immediate threat to the host.


Assuntos
Proteínas de Bactérias/genética , Proteínas Associadas a CRISPR/genética , Sistemas CRISPR-Cas/imunologia , Endonucleases/genética , Regulação Bacteriana da Expressão Gênica , Staphylococcus aureus/genética , Streptococcus pyogenes/genética , Regiões 5' não Traduzidas , Proteínas de Bactérias/metabolismo , Bacteriófagos/imunologia , Sequência de Bases , Proteína 9 Associada à CRISPR , Proteínas Associadas a CRISPR/imunologia , Cromossomos Bacterianos/química , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Endonucleases/metabolismo , Loci Gênicos , Staphylococcus aureus/imunologia , Staphylococcus aureus/virologia , Streptococcus pyogenes/imunologia , Streptococcus pyogenes/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA