Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters










Publication year range
1.
Nat Microbiol ; 6(12): 1516-1525, 2021 12.
Article in English | MEDLINE | ID: mdl-34819640

ABSTRACT

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.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Prophages/physiology , Streptococcus Phages/physiology , Streptococcus pyogenes/immunology , Streptococcus pyogenes/virology , Lysogeny , Plasmids/genetics , Plasmids/metabolism , Prophages/genetics , Streptococcus Phages/genetics , Streptococcus pyogenes/genetics , Virus Integration
2.
Viruses ; 13(5)2021 05 02.
Article in English | MEDLINE | ID: mdl-34063251

ABSTRACT

Streptococcus mutans is a key bacterium in dental caries, one of the most prevalent chronic infectious diseases. Conventional treatment fails to specifically target the pathogenic bacteria, while tending to eradicate commensal bacteria. Thus, caries remains one of the most common and challenging diseases. Phage therapy, which involves the use of bacterial viruses as anti-bacterial agents, has been gaining interest worldwide. Nevertheless, to date, only a few phages have been isolated against S. mutans. In this study, we describe the isolation and characterization of a new S. mutans phage, termed SMHBZ8, from hundreds of human saliva samples that were collected, filtered, and screened. The SMHBZ8 genome was sequenced and analyzed, visualized by TEM, and its antibacterial properties were evaluated in various states. In addition, we tested the lytic efficacy of SMHBZ8 against S. mutans in a human cariogenic dentin model. The isolation and characterization of SMHBZ8 may be the first step towards developing a potential phage therapy for dental caries.


Subject(s)
Dental Caries/therapy , Phage Therapy , Streptococcus Phages/isolation & purification , Streptococcus mutans/virology , Dental Caries/microbiology , Dental Caries/virology , Genome, Viral , Humans , Saliva/virology , Streptococcus Phages/classification , Streptococcus Phages/genetics , Streptococcus Phages/physiology , Streptococcus mutans/physiology
3.
mBio ; 12(3): e0074621, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34154404

ABSTRACT

Expression of bacteriophage lysinSM1 by Streptococcus oralis strain SF100 is thought to be important for the pathogenesis of infective endocarditis, due to its ability to mediate bacterial binding to fibrinogen. To better define the lysinSM1 binding site on fibrinogen Aα, and to investigate the impact of binding on fibrinolysis, we examined the interaction of lysinSM1 with a series of recombinant fibrinogen Aα variants. These studies revealed that lysinSM1 binds the C-terminal region of fibrinogen Aα spanned by amino acid residues 534 to 610, with an affinity of equilibrium dissociation constant (KD) of 3.23 × 10-5 M. This binding site overlaps the known binding site for plasminogen, an inactive precursor of plasmin, which is a key protease responsible for degrading fibrin polymers. When tested in vitro, lysinSM1 competitively inhibited plasminogen binding to the αC region of fibrinogen Aα. It also inhibited plasminogen-mediated fibrinolysis, as measured by thromboelastography (TEG). These results indicate that lysinSM1 is a bi-functional virulence factor for streptococci, serving as both an adhesin and a plasminogen inhibitor. Thus, lysinSM1 may facilitate the attachment of bacteria to fibrinogen on the surface of damaged cardiac valves and may also inhibit plasminogen-mediated lysis of infected thrombi (vegetations) on valve surfaces. IMPORTANCE The interaction of streptococci with human fibrinogen and platelets on damaged endocardium is a central event in the pathogenesis of infective endocarditis. Streptococcus oralis can bind platelets via the interaction of bacteriophage lysinSM1 with fibrinogen on the platelet surface, and this process has been associated with increased virulence in an animal model of endocarditis. We now report that lysinSM1 binds to the αC region of the human fibrinogen Aα chain. This interaction blocks plasminogen binding to fibrinogen and inhibits fibrinolysis. In vivo, this inhibition could prevent the lysis of infected vegetations, thereby promoting bacterial persistence and virulence.


Subject(s)
Fibrinogen/metabolism , Fibrinolysis , Plasminogen/metabolism , Streptococcus Phages/physiology , Streptococcus/metabolism , Binding Sites , Endocarditis/microbiology , Fibrin/chemistry , Fibrin/metabolism , Humans , Protein Binding , Streptococcus/genetics , Streptococcus/pathogenicity , Streptococcus/virology , Streptococcus Phages/genetics , Virulence
4.
FEMS Microbiol Rev ; 44(6): 909-932, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33016324

ABSTRACT

Almost a century has elapsed since the discovery of bacteriophages (phages), and 85 years have passed since the emergence of evidence that phages can infect starter cultures, thereby impacting dairy fermentations. Soon afterward, research efforts were undertaken to investigate phage interactions regarding starter strains. Investigations into phage biology and morphology and phage-host relationships have been aimed at mitigating the negative impact phages have on the fermented dairy industry. From the viewpoint of a supplier of dairy starter cultures, this review examines the composition of an industrial phage collection, providing insight into the development of starter strains and cultures and the evolution of phages in the industry. Research advances in the diversity of phages and structural bases for phage-host recognition and an overview of the perpetual arms race between phage virulence and host defense are presented, with a perspective toward the development of improved phage-resistant starter culture systems.


Subject(s)
Host Microbial Interactions/physiology , Lactococcus/virology , Streptococcus Phages/physiology , Dairying , Streptococcus Phages/pathogenicity
5.
Sci Rep ; 9(1): 13816, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31554834

ABSTRACT

Streptococcus thermophilus is a lactic acid bacterium widely used by the dairy industry for the manufacture of yogurt and specialty cheeses. It is also a Gram-positive bacterial model to study phage-host interactions. CRISPR-Cas systems are one of the most prevalent phage resistance mechanisms in S. thermophilus. Little information is available about other host factors involved in phage replication in this food-grade streptococcal species. We used the model strain S. thermophilus SMQ-301 and its virulent phage DT1, harboring the anti-CRISPR protein AcrIIA6, to show that a host gene coding for a methionine aminopeptidase (metAP) is necessary for phage DT1 to complete its lytic cycle. A single mutation in metAP provides S. thermophilus SMQ-301 with strong resistance against phage DT1. The mutation impedes a late step of the lytic cycle since phage adsorption, DNA replication, and protein expression were not affected. When the mutated strain was complemented with the wild-type version of the gene, the phage sensitivity phenotype was restored. When this mutation was introduced into other S. thermophilus strains it provided resistance against cos-type (Sfi21dt1virus genus) phages but replication of pac-type (Sfi11virus genus) phages was not affected. The mutation in the gene coding for the MetAP induces amino acid change in a catalytic domain conserved across many bacterial species. Introducing the same mutation in Streptococcus mutans also provided a phage resistance phenotype, suggesting the wide-ranging importance of the host methionine aminopeptidase in phage replication.


Subject(s)
Aminopeptidases/genetics , Mutation , Streptococcus Phages/physiology , Streptococcus thermophilus/virology , Aminopeptidases/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain , Food Microbiology , Streptococcus Phages/genetics , Streptococcus thermophilus/enzymology , Streptococcus thermophilus/genetics , Virus Replication , Whole Genome Sequencing
6.
Viruses ; 10(10)2018 10 22.
Article in English | MEDLINE | ID: mdl-30360457

ABSTRACT

Streptococcus thermophilus strains are among the most widely employed starter cultures in dairy fermentations, second only to those of Lactococcus lactis. The extensive application of this species provides considerable opportunity for the proliferation of its infecting (bacterio)phages. Until recently, dairy streptococcal phages were classified into two groups (cos and pac groups), while more recently, two additional groups have been identified (5093 and 987 groups). This highlights the requirement for consistent monitoring of phage populations in the industry. Here, we report a survey of 35 samples of whey derived from 27 dairy fermentation facilities in ten countries against a panel of S. thermophilus strains. This culminated in the identification of 172 plaque isolates, which were characterized by multiplex PCR, restriction fragment length polymorphism analysis, and host range profiling. Based on this characterisation, 39 distinct isolates representing all four phage groups were selected for genome sequencing. Genetic diversity was observed among the cos isolates and correlations between receptor binding protein phylogeny and host range were also clear within this phage group. The 987 phages isolated within this study shared high levels of sequence similarity, yet displayed reduced levels of similarity to those identified in previous studies, indicating that they are subject to ongoing genetic diversification.


Subject(s)
Biodiversity , Streptococcus Phages/isolation & purification , Streptococcus thermophilus/virology , Dairy Products/microbiology , Fermentation , Genetic Variation , Host Specificity , Phylogeny , Streptococcus Phages/classification , Streptococcus Phages/genetics , Streptococcus Phages/physiology , Streptococcus thermophilus/metabolism
7.
Appl Environ Microbiol ; 84(23)2018 12 01.
Article in English | MEDLINE | ID: mdl-30242010

ABSTRACT

Receptors on the cell surfaces of bacterial hosts are essential during the infection cycle of bacteriophages. To date, the phage receptors of the industrial relevant dairy starter bacterium Streptococcus thermophilus remain elusive. Thus, we set out to identify cell surface structures that are involved in host recognition by dairy streptococcal phages. Five industrial S. thermophilus strains sensitive to different phages (pac type, cos type, and the new type 987), were selected to generate spontaneous bacteriophage-insensitive mutants (BIMs). Of these, approximately 50% were deselected as clustered regularly interspaced short palindromic repeat (CRISPR) mutants, while the other pool was further characterized to identify receptor mutants. On the basis of genome sequencing data, phage resistance in putative receptor mutants was attributed to nucleotide changes in genes encoding glycan biosynthetic pathways. Superresolution structured illumination microscopy was used to visualize the interactions between S. thermophilus and its phages. The phages were either regularly distributed along the cells or located at division sites of the cells. The cell wall structures mediating the latter type of phage adherence were further analyzed via phenotypic and biochemical assays. Altogether, our data suggested that phage adsorption to S. thermophilus is mediated by glycans associated with the bacterial cell surface. Specifically, the pac-type phage CHPC951 adsorbed to polysaccharides anchored to peptidoglycan, while the 987-type phage CHPC926 recognized exocellular polysaccharides associated with the cell surface.IMPORTANCEStreptococcus thermophilus is widely used in starter cultures for cheese and yoghurt production. During dairy fermentations, infections of bacteria with bacteriophages result in acidification failures and a lower quality of the final products. An understanding of the molecular factors involved in phage-host interactions, in particular, the phage receptors in dairy bacteria, is a crucial step for developing better strategies to prevent phage infections in dairy plants.


Subject(s)
Cell Wall/metabolism , Polysaccharides/metabolism , Streptococcus Phages/physiology , Streptococcus thermophilus/virology , Cell Wall/virology , Cheese/microbiology , Fermentation , Genome, Viral , Streptococcus Phages/genetics , Streptococcus thermophilus/genetics , Streptococcus thermophilus/metabolism , Yogurt/microbiology
8.
J Bacteriol ; 200(23)2018 12 01.
Article in English | MEDLINE | ID: mdl-30224437

ABSTRACT

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.


Subject(s)
Genome, Viral/genetics , Prophages/physiology , Streptococcus Phages/physiology , Streptococcus pyogenes/virology , Lysogeny , Prophages/genetics , Streptococcus Phages/genetics , Transduction, Genetic
9.
Appl Environ Microbiol ; 84(10)2018 05 15.
Article in English | MEDLINE | ID: mdl-29523549

ABSTRACT

Phages of Streptococcus thermophilus present a major threat to the production of many fermented dairy products. To date, only a few studies have assessed the biodiversity of S. thermophilus phages in dairy fermentations. In order to develop strategies to limit phage predation in this important industrial environment, it is imperative that such studies are undertaken and that phage-host interactions of this species are better defined. The present study investigated the biodiversity and evolution of phages within an Irish dairy fermentation facility over an 11-year period. This resulted in the isolation of 17 genetically distinct phages, all of which belong to the so-called cos group. The evolution of phages within the factory appears to be influenced by phages from other dairy plants introduced into the factory for whey protein powder production. Modular exchange, primarily within the regions encoding lysogeny and replication functions, was the major observation among the phages isolated between 2006 and 2016. Furthermore, the genotype of the first isolate in 2006 was observed continuously across the following decade, highlighting the ability of these phages to prevail in the factory setting for extended periods of time. The proteins responsible for host recognition were analyzed, and carbohydrate-binding domains (CBDs) were identified in the distal tail (Dit), the baseplate proteins, and the Tail-associated lysin (Tal) variable regions (VR1 and VR2) of many isolates. This supports the notion that S. thermophilus phages recognize a carbohydrate receptor on the cell surface of their host.IMPORTANCE Dairy fermentations are consistently threatened by the presence of bacterial viruses (bacteriophages or phages), which may lead to a reduction in acidification rates or even complete loss of the fermentate. These phages may persist in factories for long periods of time. The objective of the current study was to monitor the progression of phages infecting the dairy bacterium Streptococcus thermophilus over a period of 11 years in an Irish dairy plant so as to understand how these phages evolve. A focused analysis of the genomic region that encodes host recognition functions highlighted that the associated proteins harbor a variety of carbohydrate-binding domains, which corroborates the notion that phages of S. thermophilus recognize carbohydrate receptors at the initial stages of the phage cycle.


Subject(s)
Cultured Milk Products/microbiology , Streptococcus Phages/genetics , Streptococcus thermophilus/virology , Biological Evolution , Dairying , Fermentation , Genotype , Host Specificity , Ireland , Lysogeny , Phylogeny , Streptococcus Phages/classification , Streptococcus Phages/isolation & purification , Streptococcus Phages/physiology , Streptococcus thermophilus/genetics , Streptococcus thermophilus/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
10.
Clin Microbiol Infect ; 24(5): 514-521, 2018 May.
Article in English | MEDLINE | ID: mdl-28870726

ABSTRACT

OBJECTIVES: Group B Streptococcus (GBS) emerged in the 1970s as a major cause of neonatal infections, and has been increasingly associated with infections in adults since the 1990s. Prophages have been suspected to have driven these epidemiological trends. We have characterized the prophages harboured by 275 human GBS isolates belonging to the major lineages. METHODS: We applied whole genome sequencing (WGS) to 14 isolates representative of the diversity within GBS species, located and identified their prophages. Using prediction tools, we searched for prophage elements potentially involved with the ability of GBS to infect humans. Using the data obtained by WGS, we designed a PCR-based tool and studied the prophage content of 275 isolates. RESULTS: WGS of the 14 isolates revealed 22 prophages (i) distributed into six groups (A-F), (ii) similar to phages and prophages from GBS and non-GBS streptococci recovered from livestock, and (iii) carrying genes encoding factors previously associated with host adaptation and virulence. PCR-based detection of prophages revealed the presence of at least one prophage in 72.4% of the 275 isolates and a significant association between neonatal infecting isolates and prophages C, and between adult infecting isolates and prophages A. CONCLUSIONS: Our results suggest that prophages (possibly animal-associated) have conditioned bacterial adaptation and ability to cause infections in neonates and adults, and support a role of lysogeny with the emergence of GBS as a pathogen in human.


Subject(s)
Biological Evolution , Prophages/physiology , Streptococcal Infections/microbiology , Streptococcus Phages/physiology , Streptococcus agalactiae/virology , Genome, Bacterial , Genome, Viral , Genomics , Host-Pathogen Interactions , Humans , Phylogeny , Prophages/classification , Prophages/isolation & purification , Streptococcus Phages/classification , Streptococcus Phages/isolation & purification , Streptococcus agalactiae/classification , Streptococcus agalactiae/genetics , Streptococcus agalactiae/pathogenicity , Virulence/genetics , Virus Integration , Exome Sequencing
11.
Sci Rep ; 7(1): 16506, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29184097

ABSTRACT

Streptococcus suis is a Gram-positive bacterium that infects humans and various animals, causing human mortality rates ranging from 5 to 20%, as well as important losses for the swine industry. In addition, there is no effective vaccine for S. suis and isolates with increasing antibiotic multiresistance are emerging worldwide. Facing this situation, wild type or engineered bacteriophage lysins constitute a promising alternative to conventional antibiotics. In this study, we have constructed a new chimeric lysin, Csl2, by fusing the catalytic domain of Cpl-7 lysozyme to the CW_7 repeats of LySMP lysin from an S. suis phage. Csl2 efficiently kills different S. suis strains and shows noticeable activity against a few streptococci of the mitis group. Specifically, 15 µg/ml Csl2 killed 4.3 logs of S. suis serotype 2 S735 strain in 60 min, in a buffer containing 150 mM NaCl and 10 mM CaCl2, at pH 6.0. We have set up a protocol to form a good biofilm with the non-encapsulated S. suis mutant strain BD101, and the use of 30 µg/ml Csl2 was enough for dispersing such biofilms and reducing 1-2 logs the number of planktonic bacteria. In vitro results have been validated in an adult zebrafish model of infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzymes/chemistry , Enzymes/pharmacology , Recombinant Fusion Proteins , Streptococcus suis/drug effects , Amino Acid Motifs , Amino Acid Sequence , Animals , Biofilms/drug effects , Computational Biology/methods , Disease Models, Animal , Enzyme Activation , Enzymes/genetics , Enzymes/isolation & purification , Hydrolysis , Microbial Sensitivity Tests , Repetitive Sequences, Nucleic Acid , Spectrum Analysis , Streptococcal Infections/drug therapy , Streptococcal Infections/microbiology , Streptococcus Phages/physiology , Streptococcus suis/virology , Zebrafish
12.
Science ; 357(6351): 605-609, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28663439

ABSTRACT

Type III CRISPR-Cas systems in prokaryotes provide immunity against invading nucleic acids through the coordinated degradation of transcriptionally active DNA and its transcripts by the Csm effector complex. The Cas10 subunit of the complex contains an HD nuclease domain that is responsible for DNA degradation and two Palm domains with elusive functions. In addition, Csm6, a ribonuclease that is not part of the complex, is also required to provide full immunity. We show here that target RNA binding by the Csm effector complex of Streptococcus thermophilus triggers Cas10 to synthesize cyclic oligoadenylates (cA n ; n = 2 to 6) by means of the Palm domains. Acting as signaling molecules, cyclic oligoadenylates bind Csm6 to activate its nonspecific RNA degradation. This cyclic oligoadenylate-based signaling pathway coordinates different components of CRISPR-Cas to prevent phage infection and propagation.


Subject(s)
Adenine Nucleotides/metabolism , Bacterial Proteins/metabolism , CRISPR-Cas Systems , Endonucleases/metabolism , Oligoribonucleotides/metabolism , RNA Stability , Streptococcus thermophilus/metabolism , Protein Domains , Ribonucleases/metabolism , Signal Transduction , Streptococcus Phages/physiology , Streptococcus thermophilus/genetics , Streptococcus thermophilus/virology
13.
Appl Environ Microbiol ; 82(17): 5153-65, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27316953

ABSTRACT

UNLABELLED: We present the complete genome sequences of four members of a novel group of phages infecting Streptococcus thermophilus, designated here as the 987 group. Members of this phage group appear to have resulted from genetic exchange events, as evidenced by their "hybrid" genomic architecture, exhibiting DNA sequence relatedness to the morphogenesis modules of certain P335 group Lactococcus lactis phages and to the replication modules of S. thermophilus phages. All four identified members of the 987 phage group were shown to elicit adsorption affinity to both their cognate S. thermophilus hosts and a particular L. lactis starter strain. The receptor binding protein of one of these phages (as a representative of this novel group) was defined using an adsorption inhibition assay. The emergence of a novel phage group infecting S. thermophilus highlights the continuous need for phage monitoring and development of new phage control measures. IMPORTANCE: Phage predation of S. thermophilus is an important issue for the dairy industry, where viral contamination can lead to fermentation inefficiency or complete fermentation failure. Genome information and phage-host interaction studies of S. thermophilus phages, particularly those emerging in the marketplace, are an important part of limiting the detrimental impact of these viruses in the dairy environment.


Subject(s)
Bacteriophages/isolation & purification , Streptococcus Phages/isolation & purification , Streptococcus thermophilus/virology , Bacteriophages/classification , Bacteriophages/genetics , Bacteriophages/physiology , Lactic Acid/metabolism , Streptococcus Phages/classification , Streptococcus Phages/genetics , Streptococcus Phages/physiology , Streptococcus thermophilus/metabolism
14.
Sci Rep ; 6: 24597, 2016 04 22.
Article in English | MEDLINE | ID: mdl-27103053

ABSTRACT

Although an abundance of bacteriophages exists, little is known about interactions between their proteins and those of their bacterial hosts. Here, we experimentally determined the phage-host interactomes of the phages Dp-1 and Cp-1 and their underlying protein interaction network in the host Streptococcus pneumoniae. We compared our results to the interaction patterns of E. coli phages lambda and T7. Dp-1 and Cp-1 target highly connected host proteins, occupy central network positions, and reach many protein clusters through the interactions of their targets. In turn, lambda and T7 targets cluster to conserved and essential proteins in E. coli, while such patterns were largely absent in S. pneumoniae. Furthermore, targets in E. coli were mutually strongly intertwined, while targets of Dp-1 and Cp-1 were strongly connected through essential and orthologous proteins in their immediate network vicinity. In both phage-host systems, the impact of phages on their protein targets appears to extend from their network neighbors, since proteins that interact with phage targets were located in central network positions, have a strong topologically disruptive effect and touch complexes with high functional heterogeneity. Such observations suggest that the phages, biological impact is accomplished through a surprisingly limited topological reach of their targets.


Subject(s)
Host-Parasite Interactions , Protein Interaction Maps , Streptococcus Phages/physiology , Streptococcus pneumoniae/physiology , Streptococcus pneumoniae/virology , Bacterial Proteins/metabolism , Bacteriophage T7/physiology , Bacteriophage lambda/physiology , Escherichia coli/physiology , Escherichia coli/virology , Protein Interaction Mapping , Viral Proteins/metabolism
15.
PLoS One ; 10(9): e0138651, 2015.
Article in English | MEDLINE | ID: mdl-26398909

ABSTRACT

Streptococcus mutans is one of the principal agents of caries formation mainly, because of its ability to form biofilms at the tooth surface. Bacteriophages (phages) are promising antimicrobial agents that could be used to prevent or treat caries formation by S. mutans. The aim of this study was to isolate new S. mutans phages and to characterize their antimicrobial properties. A new phage, ɸAPCM01, was isolated from a human saliva sample. Its genome was closely related to the only two other available S. mutans phage genomes, M102 and M102AD. ɸAPCM01 inhibited the growth of S. mutans strain DPC6143 within hours in broth and in artificial saliva at multiplicity of infections as low as 2.5x10-5. In the presence of phage ɸAPCM01 the metabolic activity of a S. mutans biofilm was reduced after 24 h of contact and did not increased again after 48 h, and the live cells in the biofilm decreased by at least 5 log cfu/ml. Despite its narrow host range, this newly isolated S. mutans phage exhibits promising antimicrobial properties.


Subject(s)
Biofilms , Streptococcus Phages/physiology , Streptococcus mutans/virology , Humans , Microscopy, Electron, Transmission , Molecular Sequence Data , Saliva/virology , Streptococcus Phages/ultrastructure
16.
Wei Sheng Wu Xue Bao ; 55(4): 389-94, 2015 Apr 04.
Article in Chinese | MEDLINE | ID: mdl-26211312

ABSTRACT

Streptococcus suis (S. suis) is an important zoonosis and pathogen that can carry prophages. In this review, we focus on the recent advances in our understanding of lytic phage and lysogenic phage of S. suis, including the morphology of S. suis lytic phage, the functions of lysin and terminase large subunit encoded by S. suis lytic phage, comparative genomics of S. suis prophages, lysogenic. conversion between S. suis lytic phage and prophage. Furthermore, prospective evolution of interactions between phage and host was discussed.


Subject(s)
Prophages/physiology , Streptococcus Phages/physiology , Streptococcus suis/virology , Prophages/genetics , Prophages/ultrastructure , Streptococcus Phages/genetics , Streptococcus Phages/ultrastructure
17.
PLoS One ; 10(2): e0118807, 2015.
Article in English | MEDLINE | ID: mdl-25692983

ABSTRACT

Streptococcus mitis has emerged as one of the leading causes of bacterial endocarditis and is related to Streptococcus pneumoniae. Antibiotic resistance has also increased among strains of S. mitis and S. pneumoniae. Phages are being reinvestigated as alternatives to antibiotics for managing infections. In this study, the two virulent phages Cp-1 (Podoviridae) and Dp-1 (Siphoviridae), previously isolated from S. pneumoniae, were found to also infect S. mitis. Microbiological assays showed that both pneumophages could not only replicate in S. mitis but also produced more visible plaques on this host. However, the burst size and phage adsorption data were lower in S. mitis as compared to S. pneumoniae. A comparison of the genomes of each phage grown on both hosts produced identical nucleotide sequences, confirming that the same phages infect both bacterial species. We also discovered that the genomic sequence of podophage Cp-1 of the Félix d'Hérelle collection is different than the previously reported sequence and thus renamed SOCP.


Subject(s)
Streptococcus Phages/physiology , Streptococcus mitis/isolation & purification , Streptococcus pneumoniae/isolation & purification , DNA, Bacterial/genetics , Genome, Viral , Molecular Sequence Data , Sequence Analysis, DNA , Streptococcus Phages/classification , Streptococcus mitis/classification , Streptococcus mitis/virology , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/virology
18.
Nucleic Acids Res ; 43(3): 1749-58, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25589547

ABSTRACT

CRISPR-Cas systems are RNA-based immune systems that protect prokaryotes from invaders such as phages and plasmids. In adaptation, the initial phase of the immune response, short foreign DNA fragments are captured and integrated into host CRISPR loci to provide heritable defense against encountered foreign nucleic acids. Each CRISPR contains a ∼100-500 bp leader element that typically includes a transcription promoter, followed by an array of captured ∼35 bp sequences (spacers) sandwiched between copies of an identical ∼35 bp direct repeat sequence. New spacers are added immediately downstream of the leader. Here, we have analyzed adaptation to phage infection in Streptococcus thermophilus at the CRISPR1 locus to identify cis-acting elements essential for the process. We show that the leader and a single repeat of the CRISPR locus are sufficient for adaptation in this system. Moreover, we identified a leader sequence element capable of stimulating adaptation at a dormant repeat. We found that sequences within 10 bp of the site of integration, in both the leader and repeat of the CRISPR, are required for the process. Our results indicate that information at the CRISPR leader-repeat junction is critical for adaptation in this Type II-A system and likely other CRISPR-Cas systems.


Subject(s)
Adaptation, Physiological , Clustered Regularly Interspaced Short Palindromic Repeats , Streptococcus Phages/physiology , Streptococcus thermophilus/virology , Base Sequence , DNA, Viral , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Streptococcus Phages/genetics
19.
J Bacteriol ; 196(14): 2670-80, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24816604

ABSTRACT

The pathogenesis of the disease caused by Streptococcus pneumoniae begins with colonization of the upper respiratory tract. Temperate phages have been identified in the genomes of up to 70% of clinical isolates. How these phages affect the bacterial host during colonization is unknown. Here, we examined a clinical isolate that carries a novel prophage element, designated Spn1, which was detected in both integrated and episomal forms. Surprisingly, both lytic and lysogenic Spn1 genes were expressed under routine growth conditions. Using a mouse model of asymptomatic colonization, we demonstrate that the Spn1(-) strain outcompeted the Spn1(+) strain >70-fold. To determine if Spn1 causes a fitness defect through a trans-acting factor, we constructed an Spn1(+) mutant that does not become an episome or express phage genes. This mutant competed equally with the Spn1(-) strain, indicating that expression of phage genes or phage lytic activity is required to confer this fitness defect. In vitro, we demonstrate that the presence of Spn1 correlated with a defect in LytA-mediated autolysis. Furthermore, the Spn1(+) strain displayed increased chain length and resistance to lysis by penicillin compared to the Spn(-) strain, indicating that Spn1 alters the cell wall physiology of its host strain. We posit that these changes in cell wall physiology allow for tolerance of phage gene products and are responsible for the relative defect of the Spn1(+) strain during colonization. This study provides new insight into how bacteria and prophages interact and affect bacterial fitness in vivo.


Subject(s)
Pneumococcal Infections/microbiology , Prophages/physiology , Streptococcus Phages/physiology , Streptococcus pneumoniae/metabolism , Animals , Bacterial Proteins/genetics , Carrier State/microbiology , Mice , Microbial Sensitivity Tests , Molecular Sequence Data , Penicillin Resistance/genetics , Penicillins/pharmacology , Prophages/genetics , Streptococcus Phages/genetics , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/genetics
20.
Arch Virol ; 158(8): 1733-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23515875

ABSTRACT

A novel bacteriophage, JX01, specifically infecting bovine Streptococcus agalactiae was isolated from milk of mastitis-affected cattle. The phage morphology showed that JX01 belongs to the family Siphoviridae, and this phage demonstrated a broad host range. Microbiological characterization demonstrated that nearly 90 % of JX01 phage particles were adsorbed after 2.5 min of incubation, that the burst size was 20 virions released per infected host cell, and that there was a latent period of 30 min. JX01 was thermal sensitive and showed acid and alkaline resistance (pH 3-11). The genome of JX01 was found to consist of a linear, double-stranded 43,028-bp DNA molecule with a GC content of 36.81 % and 70 putative open reading frames (ORFs) plus one tRNA. Comparative genome analysis revealed high similarity between JX01 and the prophage 315.2 of Streptococcus pyogenes.


Subject(s)
DNA, Viral/chemistry , DNA, Viral/genetics , Genome, Viral , Streptococcus Phages/genetics , Streptococcus Phages/isolation & purification , Streptococcus agalactiae/virology , Animals , Base Composition , Cattle , Cluster Analysis , Molecular Sequence Data , Open Reading Frames , Phylogeny , Sequence Analysis, DNA , Sequence Homology , Siphoviridae/genetics , Siphoviridae/isolation & purification , Siphoviridae/physiology , Siphoviridae/ultrastructure , Streptococcus Phages/physiology , Streptococcus Phages/ultrastructure , Streptococcus agalactiae/isolation & purification , Virus Attachment
SELECTION OF CITATIONS
SEARCH DETAIL
...