Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 266
Filtrar
1.
Cell Host Microbe ; 32(5): 634-636, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38723602

RESUMEN

Bacterial genomes are littered with exogenous: competing DNA elements. Here, Sprenger et al. demonstrate that the Vibrio cholerae prophage VP882 modulates host functions via production of regulatory sRNAs to promote phage development. Alternatively, host sRNAs inhibit the VP882 lytic phase by specifically regulating phage genes.


Asunto(s)
Profagos , Vibrio cholerae , Vibrio cholerae/genética , Profagos/genética , Profagos/fisiología , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Genoma Bacteriano , Bacteriófagos/genética , Bacteriófagos/fisiología , Regulación Bacteriana de la Expresión Génica , ARN Bacteriano/genética , ARN Bacteriano/metabolismo
2.
Curr Biol ; 34(8): R331-R333, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38653204

RESUMEN

Bacterial genomes often harbor integrated viruses (prophages), which provide novel functions but also lyse cells under stressful conditions. A new paper combines mathematical models with experimental evolution to determine how prophages are maintained in bacterial populations despite their fitness costs.


Asunto(s)
Bacterias , Profagos , Profagos/genética , Profagos/fisiología , Bacterias/virología , Bacterias/genética
3.
Curr Biol ; 34(8): 1739-1749.e7, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38599209

RESUMEN

Prophages, viral sequences integrated into bacterial genomes, can be beneficial and costly. Despite the risk of prophage activation and subsequent bacterial death, active prophages are present in most bacterial genomes. However, our understanding of the selective forces that maintain prophages in bacterial populations is limited. Combining experimental evolution with stochastic modeling, we show that prophage maintenance and loss are primarily determined by environmental conditions that alter the net fitness effect of a prophage on its bacterial host. When prophages are too costly, they are rapidly lost through environment-specific sequences of selective sweeps. Conflicting selection pressures that select against the prophage but for a prophage-encoded accessory gene can maintain prophages. The dynamics of prophage maintenance additionally depend on the sociality of this accessory gene. Prophage-encoded genes that exclusively benefit the lysogen maintain prophages at higher frequencies compared with genes that benefit the entire population. That is because the latter can protect phage-free "cheaters," reducing the benefit of maintaining the prophage. Our simulations suggest that environmental variation plays a larger role than mutation rates in determining prophage maintenance. These findings highlight the complexity of selection pressures that act on mobile genetic elements and challenge our understanding of the role of environmental factors relative to random chance events in shaping the evolutionary trajectory of bacterial populations. By shedding light on the key factors that shape microbial populations in the face of environmental changes, our study significantly advances our understanding of the complex dynamics of microbial evolution and diversification.


Asunto(s)
Profagos , Profagos/genética , Profagos/fisiología , Selección Genética , Mutación , Ambiente , Lisogenia/genética , Evolución Molecular
4.
Cell Host Microbe ; 32(5): 768-778.e9, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38653241

RESUMEN

Microbiomes feature complex interactions between diverse bacteria and bacteriophages. Synthetic microbiomes offer a powerful way to study these interactions; however, a major challenge is obtaining a representative bacteriophage population during the bacterial isolation process. We demonstrate that colony isolation reliably excludes virulent viruses from sample sources with low virion-to-bacteria ratios such as feces, creating "virulent virus-free" controls. When the virulent dsDNA virome is reintroduced to a 73-strain synthetic gut microbiome in a bioreactor model of the human colon, virulent viruses target susceptible strains without significantly altering community structure or metabolism. In addition, we detected signals of prophage induction that associate with virulent predation. Overall, our findings indicate that dilution-based isolation methods generate synthetic gut microbiomes that are heavily depleted, if not devoid, of virulent viruses and that such viruses, if reintroduced, have a targeted effect on community assembly, metabolism, and prophage replication.


Asunto(s)
Bacterias , Bacteriófagos , Heces , Microbioma Gastrointestinal , Bacteriófagos/genética , Bacteriófagos/fisiología , Humanos , Heces/microbiología , Heces/virología , Bacterias/virología , Bacterias/genética , Profagos/genética , Profagos/fisiología , Viroma , Reactores Biológicos/microbiología , Reactores Biológicos/virología , Colon/microbiología , Colon/virología , Microbiota , Virulencia
5.
Appl Environ Microbiol ; 88(18): e0106822, 2022 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-36073944

RESUMEN

Pseudomonas aeruginosa is a notorious pathogen that causes various nosocomial infections. Several prophage genes located on the chromosomes of P. aeruginosa have been reported to contribute to bacterial pathogenesis via host phenotype transformations, such as serotype conversion and antibiotic resistance. However, our understanding of the molecular mechanism behind host phenotype shifts induced by prophage genes remains largely unknown. Here, we report a systematic study around a hypothetical recombinase, Pg54 (RecT), located on a 48-kb putative prophage (designated PP9W) of a clinical P. aeruginosa strain P9W. Using a ΔrecT mutant (designated P9D), we found that RecT promoted prophage PP9W excision and gene transcription via the inhibition of the gene expression level of pg40, which encodes a CI-like repressor protein. Further transcriptomic profiling and various phenotypic tests showed that RecT modulated like a suppressor to some transcription factors and vital genes of diverse cellular processes, providing multiple advantages for the host, including cell growth, biofilm formation, and virulence. The versatile functions of RecT hint at a strong impact of phage proteins on host P. aeruginosa phenotypic flexibility. IMPORTANCE Multidrug-resistant and metabolically versatile P. aeruginosa are difficult to eradicate by anti-infective therapy and frequently lead to significant morbidity and mortality. This study characterizes a putative recombinase (RecT) encoded by a prophage of a clinical P. aeruginosa strain isolated from severely burned patients, altering prophage lifestyle and host core cellular processes. It implies the potential role of RecT in the coevolution arm race between bacteria and phage. The excised free phages from the chromosome of host bacteria can be used as weapons against other sensitive competitors in diverse environments, which may increase the lysogeny frequency of different P. aeruginosa subgroups. Subsequent analyses revealed that RecT both positively and negatively affects different phenotypic traits of the host. These findings concerning RecT functions of host phenotypic flexibility improve our understanding of the association between phage recombinases and clinical P. aeruginosa, providing new insight into mitigating the pathogen infection.


Asunto(s)
Bacteriófagos , Profagos , Bacteriófagos/genética , Profagos/fisiología , Pseudomonas aeruginosa/genética , Recombinasas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo
6.
Environ Sci Technol ; 56(12): 8920-8931, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35438974

RESUMEN

Biofilms can be pervasive and problematic in water treatment and distribution systems but are difficult to eradicate due to hindered penetration of antimicrobial chemicals. Here, we demonstrate that indigenous prophages activated by low-intensity plasma have the potential for efficient bacterial inactivation and biofilm disruption. Specifically, low-intensity plasma treatment (i.e., 35.20 W) elevated the intracellular oxidative reactive species (ROS) levels by 184%, resulting in the activation of prophage lambda (λ) within antibiotic-resistant Escherichia coli K-12 (lambda+) [E. coli (λ+)]. The phage activation efficiency was 6.50-fold higher than the conventional mitomycin C induction. Following a cascading effect, the activated phages were released upon the lysis of E. coli (λ+), which propagated further and lysed phage-susceptible E. coli K-12 (lambda-) [E. coli (λ-)] within the biofilm. Bacterial intracellular ROS analysis and ROS scavenger tests revealed the importance of plasma-generated ROS (e.g., •OH, 1O2, and •O2-) and associated intracellular oxidative stress on prophage activation. In a mixed-species biofilm on a permeable membrane surface, our "inside-out" strategy could inactivate total bacteria by 49% and increase the membrane flux by 4.33-fold. Furthermore, the metagenomic analysis revealed that the decrease in bacterial abundance was closely associated with the increase in phage levels. As a proof-of-concept, this is the first demonstration of indigenous prophage activations by low-intensity plasma for antibiotic-resistant bacterial inactivation and biofilm eradication, which opens up a new avenue for managing associated microbial problems.


Asunto(s)
Bacteriófagos , Escherichia coli K12 , Gases em Plasma , Antibacterianos/farmacología , Bacterias , Biopelículas , Escherichia coli , Gases em Plasma/farmacología , Profagos/fisiología , Especies Reactivas de Oxígeno
7.
Nature ; 603(7900): 315-320, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35197633

RESUMEN

Colibactin is a chemically unstable small-molecule genotoxin that is produced by several different bacteria, including members of the human gut microbiome1,2. Although the biological activity of colibactin has been extensively investigated in mammalian systems3, little is known about its effects on other microorganisms. Here we show that colibactin targets bacteria that contain prophages, and induces lytic development through the bacterial SOS response. DNA, added exogenously, protects bacteria from colibactin, as does expressing a colibactin resistance protein (ClbS) in non-colibactin-producing cells. The prophage-inducing effects that we observe apply broadly across different phage-bacteria systems and in complex communities. Finally, we identify bacteria that have colibactin resistance genes but lack colibactin biosynthetic genes. Many of these bacteria are infected with predicted prophages, and we show that the expression of their ClbS homologues provides immunity from colibactin-triggered induction. Our study reveals a mechanism by which colibactin production could affect microbiomes and highlights a role for microbial natural products in influencing population-level events such as phage outbreaks.


Asunto(s)
Bacterias , Toxinas Bacterianas , Péptidos , Policétidos , Profagos , Activación Viral , Bacterias/efectos de los fármacos , Bacterias/virología , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacología , Bacteriólisis/efectos de los fármacos , Interacciones Microbianas/efectos de los fármacos , Péptidos/metabolismo , Péptidos/farmacología , Policétidos/metabolismo , Policétidos/farmacología , Profagos/efectos de los fármacos , Profagos/fisiología , Respuesta SOS en Genética/efectos de los fármacos , Activación Viral/efectos de los fármacos
8.
Virology ; 568: 86-100, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35149347

RESUMEN

Bacteriophage T7 is an extensively studied virulent phage, and its taxonomic family, the Autographiviridae, is broadly synonymous with a strictly virulent lifestyle. It is difficult to imagine how a T7-like phage could function in a "domesticated" temperate lifestyle, in which it is incorporated into the host's genome. Here we describe two temperate T7-like bacteriophages: ProddE, a Desulfovibrio phage, and Pasto, an Agrobacterium phage. Each contains recognizable T7-like proteins in the canonical T7-like gene order, but with the addition of lysogeny gene modules. While ProddE contains a phage-like repressor, Pasto lysogeny appears to be controlled by a novel MarR-like transcriptional regulator. In addition, we identify similar T7-like prophage elements in a wide variety of Gram-negative bacterial genomes and a small number of Gram-positive genomes. Identification of these elements in diverse bacterial species raises interesting evolutionary questions about the origins of T7-like phages and which lifestyle, temperate or virulent, is the ancestral form.


Asunto(s)
Bacteriófagos/fisiología , Caudovirales/fisiología , Evolución Biológica , Evolución Molecular , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Lisogenia , Filogenia , Profagos/fisiología , Replicación Viral
9.
mBio ; 12(6): e0225921, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34872344

RESUMEN

Bacterial pathogens commonly carry prophages that express virulence factors, and human strains of Staphylococcus aureus carry Sa3int phages, which promote immune evasion. Recently, however, these phages have been found in livestock-associated, methicillin-resistant S. aureus (LA-MRSA). This is surprising, as LA-MRSA strains contain a mutated primary bacterial integration site, which likely explains why the rare integration events that do occur mostly happen at alternative locations. Using deep sequencing, we show that after initial integration at secondary sites, Sa3int phages adapt through nucleotide changes in their attachment sequences to increase homology with alternative bacterial attachment sites. Importantly, this homology significantly enhances integrations in new rounds of infections. We propose that promiscuity of the phage-encoded tyrosine recombinase is responsible for establishment of Sa3int phages in LA-MRSA. Our results demonstrate that phages can adopt extensive population heterogeneity, leading to establishment in strains lacking bona fide integration sites. Ultimately, their presence may increase virulence and zoonotic potential of pathogens with major implications for human health. IMPORTANCE A growing number of humans are being infected by antibiotic resistant Staphylococcus aureus originating from livestock. The preference of S. aureus for humans or animals is in part determined by factors encoded by viruses (phages) that reside in the bacterial genome. Here, we reveal a process by which phages adapt to and become integrated in new strains of S. aureus lacking the preferred phage integration site. We propose that this is due to the relaxed specificity of a phage-encoded enzyme called recombinase. As this recombinase is used by many other phages, our results might have implications for a broader range of phages. Importantly, the adaptation described here enables S. aureus to jump between host organisms and increases its zoonotic threat.


Asunto(s)
Sitios de Ligazón Microbiológica , Staphylococcus aureus Resistente a Meticilina/virología , Fagos de Staphylococcus/fisiología , Adaptación Biológica , Animales , Especificidad del Huésped , Humanos , Ganado/microbiología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Staphylococcus aureus Resistente a Meticilina/fisiología , Profagos/genética , Profagos/fisiología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/veterinaria , Fagos de Staphylococcus/genética , Virulencia , Integración Viral , Zoonosis/microbiología
10.
Viruses ; 13(11)2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34835047

RESUMEN

Aquaculture is a rapidly growing food production sector. Fish farmers are experiencing increasing problems with antibiotic resistance when fighting against pathogenic bacteria such as Aeromonas salmonicida subsp. salmonicida, the causative agent of furunculosis. Phage therapy may provide an alternative, but effective use must be determined. Here, we studied the inhibition of A. salmonicida subsp. salmonicida strains by five phages (HER98 [44RR2.8t.2], HER110 [65.2], SW69-9, L9-6 and Riv-10) used individually or as combinations of two to five phages. A particular combination of four phages (HER98 [44RR2.8t.2], SW69-9, Riv-10, and HER110 [65.2]) was found to be the most effective when used at an initial multiplicity of infection (MOI) of 1 against the A. salmonicida subsp. salmonicida strain 01-B526. The same phage cocktail is effective against other strains except those bearing a prophage (named Prophage 3), which is present in 2/3 of the strains from the province of Quebec. To confirm the impact of this prophage, we tested the effectiveness of the same cocktail on strains that were either cured or lysogenized with Prophage 3. While the parental strains were sensitive to the phage cocktail, the lysogenized ones were much less sensitive. These data indicate that the prophage content of A. salmonicida subsp. salmonicida can affect the efficacy of a cocktail of virulent phages for phage therapy purposes.


Asunto(s)
Aeromonas/virología , Bacteriófagos/fisiología , Profagos/fisiología , Aeromonas/genética , Aeromonas/crecimiento & desarrollo , Animales , Acuicultura , Bacteriófagos/clasificación , Forunculosis/microbiología , Forunculosis/terapia , Islas Genómicas/genética , Especificidad del Huésped , Lisogenia , Terapia de Fagos/veterinaria
11.
Nat Microbiol ; 6(12): 1516-1525, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34819640

RESUMEN

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.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Profagos/fisiología , Fagos de Streptococcus/fisiología , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/virología , Lisogenia , Plásmidos/genética , Plásmidos/metabolismo , Profagos/genética , Fagos de Streptococcus/genética , Streptococcus pyogenes/genética , Integración Viral
12.
Microbiologyopen ; 10(5): e1245, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34713608

RESUMEN

Temperate phages are bacterial viruses that after infection either reside integrated into a bacterial genome as prophages forming lysogens or multiply in a lytic lifecycle. The decision between lifestyles is determined by a switch involving a phage-encoded repressor, CI, and a promoter region from which lytic and lysogenic genes are divergently transcribed. Here, we investigate the switch of phage ɸ13 from the human pathogen Staphylococcus aureus. ɸ13 encodes several virulence factors and is prevalent in S. aureus strains colonizing humans. We show that the ɸ13 switch harbors a cI gene, a predicted mor (modulator of repression) gene, and three high-affinity operator sites binding CI. To quantify the decision between lytic and lysogenic lifestyle, we introduced reporter plasmids that carry the 1.3 kb switch region from ɸ13 with the lytic promoter fused to lacZ into S. aureus and Bacillus subtilis. Analysis of ß-galactosidase expression indicated that decision frequency is independent of host factors. The white "lysogenic" phenotype, which relies on the expression of cI, could be switched to a stable blue "lytic" phenotype by DNA damaging agents. We have characterized lifestyle decisions of phage ɸ13, and our approach may be applied to other temperate phages encoding virulence factors in S. aureus.


Asunto(s)
Bacteriólisis , Lisogenia , Proteínas Represoras/genética , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/fisiología , Staphylococcus aureus/virología , Proteínas Virales/genética , Proteínas Reguladoras y Accesorias Virales/genética , Toxinas Bacterianas/genética , Daño del ADN , ADN Intergénico , Exotoxinas/genética , Genes Virales , Humanos , Leucocidinas/genética , Regiones Operadoras Genéticas , Fenotipo , Plásmidos , Profagos/fisiología , Proteínas Represoras/metabolismo , Staphylococcus aureus/crecimiento & desarrollo , Proteínas Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Factores de Virulencia/genética
13.
J Bacteriol ; 203(20): e0010421, 2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34370558

RESUMEN

The antifeeding prophage (Afp) produced by the bacterium Serratia entomophila is the archetypical external contractile injection system (eCIS). Afp and its orthologues are characterized by three sheath proteins, while contractile bacteriophages and pyocins encode only one. Using targeted mutagenesis, transmission electron microscopy (TEM), and pulldown studies, we interrogated the roles of the three sheath proteins (Afp2, Afp3, and Afp4) in Afp assembly, in particular the interaction between the two sequence-related helical-sheath-forming proteins Afp2 and Afp3 and their cross talk with the tail termination sheath capping protein (TrP) Afp16 in the sheath maturation process. The expressed assemblies for the afp2-deficient mutant were mostly a mixture of isolated tail fibers, detached baseplates without tail fibers, and sheathless inner tube baseplate complexes (TBCs) with a length similar to that of mature Afp, which were surrounded in many cases by fibrillar polymerized material. In the afp3-deficient mutant, variable-length TBCs with similar but shorter fibrillar polymerized material, largely bereft of tail fibers, were observed, while only detached baseplate assemblies were seen for the afp4-deficient mutant. Furthermore, we found that (i) only trans complementation of afp2 with its mutated counterpart restored mature Afp particles with full biological activity, (ii) purified Afp3 pulled down Afp2 by forming a sodium dodecyl sulfate (SDS)-resistant complex but not vice versa, (iii) Afp16 had a higher affinity for binding Afp2 or Afp3 than Afp4, and (iv) Afp4 is required for the association of the polymerized sheath on the baseplate via Afp2. A proposed model for sheath maturation and assembly in Afp is presented. IMPORTANCE Members of the contractile bacteriophage-related but evolutionarily divergent eCIS contain not one but three sheath proteins, two of which, namely, Afp2 and Afp3 in the Afp, arranged as alternate hexameric stacks constitute the helical sheath. We revealed that Afp2 and Afp3, even though they are highly similar, possess markedly distinct, crucial roles in Afp assembly. We find that Afp3, by virtue of its interaction with the tail-terminating protein Afp16, regulates tube and sheath length, while Afp2 is critical for proper sheath polymerization and the assembly of the baseplate. The resulting model for the Afp assembly will further guide the manipulation of Afp and its related eCISs as nanodelivery vehicles for pest control and phage therapy.


Asunto(s)
Profagos , Serratia/virología , Proteínas Virales/metabolismo , Regulación Viral de la Expresión Génica , Humanos , Chaperonas Moleculares , Mutagénesis , Profagos/crecimiento & desarrollo , Profagos/fisiología , Proteínas Virales/química , Proteínas Virales/genética , Replicación Viral
14.
Toxins (Basel) ; 13(7)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206386

RESUMEN

Escherichia coli O157:H7 strains often produce Shiga toxins encoded by genes on lambdoid bacteriophages that insert into multiple loci as prophages. O157 strains were classified into distinct clades that vary in virulence. Herein, we used PCR assays to examine Shiga toxin (Stx) prophage occupancy in yehV, argW, wrbA, and sbcB among 346 O157 strains representing nine clades. Overall, yehV was occupied in most strains (n = 334, 96.5%), followed by wrbA (n = 213, 61.6%), argW (n = 103, 29.8%), and sbcB (n = 93, 26.9%). Twelve occupancy profiles were identified that varied in frequency and differed across clades. Strains belonging to clade 8 were more likely to have occupied sbcB and argW sites compared to other clades (p < 0.0001), while clade 2 strains were more likely to have occupied wrbA sites (p < 0.0001). Clade 8 strains also had more than the expected number of occupied sites based on the presence of stx variants (p < 0.0001). Deletion of a 20 kb non-Stx prophage occupying yehV in a clade 8 strain resulted in an ~18-fold decrease in stx2 expression. These data highlight the complexity of Stx prophage integration and demonstrate that clade 8 strains, which were previously linked to hemolytic uremic syndrome, have unique Stx prophage occupancy profiles that can impact stx2 expression.


Asunto(s)
Escherichia coli O157/virología , Profagos/fisiología , Escherichia coli O157/genética , Lisogenia , Toxina Shiga
15.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34244443

RESUMEN

Single-stranded DNA phages of the family Microviridae have fundamentally different evolutionary origins and dynamics than the more frequently studied double-stranded DNA phages. Despite their small size (around 5 kb), which imposes extreme constraints on genomic innovation, they have adapted to become prominent members of viromes in numerous ecosystems and hold a dominant position among viruses in the human gut. We show that multiple, divergent lineages in the family Microviridae have independently become capable of lysogenizing hosts and have convergently developed hypervariable regions in their DNA pilot protein, which is responsible for injecting the phage genome into the host. By creating microviruses with combinations of genomic segments from different phages and infecting Escherichia coli as a model system, we demonstrate that this hypervariable region confers the ability of temperate Microviridae to prevent DNA injection and infection by other microviruses. The DNA pilot protein is present in most microviruses, but has been recruited repeatedly into this additional role as microviruses altered their lifestyle by evolving the ability to integrate in bacterial genomes, which linked their survival to that of their hosts. Our results emphasize that competition between viruses is a considerable and often overlooked source of selective pressure, and by producing similar evolutionary outcomes in distinct lineages, it underlies the prevalence of hypervariable regions in the genomes of microviruses and perhaps beyond.


Asunto(s)
Microvirus/fisiología , Sobreinfección/virología , Proteínas Virales/química , ADN Viral/metabolismo , Escherichia coli/virología , Inmunidad , Filogenia , Profagos/fisiología , Sobreinfección/inmunología
16.
BMC Microbiol ; 21(1): 176, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34107872

RESUMEN

BACKGROUND: The global rise in the incidence of non-tuberculosis mycobacterial infections is of increasing concern due their high levels of intrinsic antibiotic resistance. Although integrated viral genomes, called prophage, are linked to increased antibiotic resistance in some bacterial species, we know little of their role in mycobacterial drug resistance. RESULTS: We present here for the first time, evidence of increased antibiotic resistance and expression of intrinsic antibiotic resistance genes in a strain of Mycobacterium chelonae carrying prophage. Strains carrying the prophage McProf demonstrated increased resistance to amikacin. Resistance in these strains was further enhanced by exposure to sub-inhibitory concentrations of the antibiotic, acivicin, or by the presence of a second prophage, BPs. Increased expression of the virulence gene, whiB7, was observed in strains carrying both prophages, BPs and McProf, relative to strains carrying a single prophage or no prophages. CONCLUSIONS: This study provides evidence that prophage alter expression of important mycobacterial intrinsic antibiotic resistance genes and additionally offers insight into the role prophage may play in mycobacterial adaptation to stress.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana , Mycobacterium chelonae/metabolismo , Mycobacterium chelonae/virología , Profagos/fisiología , Factores de Virulencia/metabolismo , Proteínas Bacterianas/genética , Mycobacterium chelonae/efectos de los fármacos , Mycobacterium chelonae/genética , Factores de Virulencia/genética
17.
J Bacteriol ; 203(16): e0070320, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34060907

RESUMEN

Bacteriophage serine integrases catalyze highly specific recombination reactions between defined DNA segments called att sites. These reactions are reversible depending upon the presence of a second phage-encoded directionality factor. The bipartite C-terminal DNA-binding region of integrases includes a recombinase domain (RD) connected to a zinc-binding domain (ZD), which contains a long flexible coiled-coil (CC) motif that extends away from the bound DNA. We directly show that the identities of the phage A118 integrase att sites are specified by the DNA spacing between the RD and ZD DNA recognition determinants, which in turn directs the relative trajectories of the CC motifs on each subunit of the att-bound integrase dimer. Recombination between compatible dimer-bound att sites requires minimal-length CC motifs and 14 residues surrounding the tip where the pairing of CC motifs between synapsing dimers occurs. Our alanine-scanning data suggest that molecular interactions between CC motif tips may differ in integrative (attP × attB) and excisive (attL × attR) recombination reactions. We identify mutations in 5 residues within the integrase oligomerization helix that control the remodeling of dimers into tetramers during synaptic complex formation. Whereas most of these gain-of-function mutants still require the CC motifs for synapsis, one mutant efficiently, but indiscriminately, forms synaptic complexes without the CC motifs. However, the CC motifs are still required for recombination, suggesting a function for the CC motifs after the initial assembly of the integrase synaptic tetramer. IMPORTANCE The robust and exquisitely regulated site-specific recombination reactions promoted by serine integrases are integral to the life cycle of temperate bacteriophage and, in the case of the A118 prophage, are an important virulence factor of Listeria monocytogenes. The properties of these recombinases have led to their repurposing into tools for genetic engineering and synthetic biology. In this report, we identify determinants regulating synaptic complex formation between correct DNA sites, including the DNA architecture responsible for specifying the identity of recombination sites, features of the unique coiled-coil structure on the integrase that are required to initiate synapsis, and amino acid residues on the integrase oligomerization helix that control the remodeling of synapsing dimers into a tetramer active for DNA strand exchange.


Asunto(s)
Bacteriófagos/enzimología , Emparejamiento Cromosómico , Integrasas/química , Integrasas/metabolismo , Listeria monocytogenes/virología , Proteínas Virales/química , Proteínas Virales/metabolismo , Integración Viral , Secuencias de Aminoácidos , Sitios de Ligazón Microbiológica , Bacteriófagos/química , Bacteriófagos/genética , Bacteriófagos/fisiología , Integrasas/genética , Listeria monocytogenes/genética , Profagos/química , Profagos/enzimología , Profagos/genética , Profagos/fisiología , Dominios Proteicos , Recombinación Genética , Proteínas Virales/genética
18.
Infect Genet Evol ; 92: 104881, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33905883

RESUMEN

The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) is a bacterial defense mechanism against bacteriophages composed of two different parts: the CRISPR array and the Cas genes. The spacer acquisition is done by the adaptation module consisting of the hallmark Cas1 Cas2 proteins, which inserts new spacers into the CRISPR array. Here we aimed to describe the CRISPR-Cas system in Proteus mirabilis (P. mirabilis) isolates. CRISPR loci was observed in 30 genomic contents of 109 P. mirabilis isolates that each locus was consisted of two CRISPR arrays and each array had a different preserved leader sequences. Only the type I-E CRISPR-Cas system was common in these isolates. The source of the spacers was identified, including phages and prophages. CRISPR spacer origin analysis also identified a conserved PAM sequence of 5'-AAG-3' nucleotide stretch. Through collecting spacers, CRISPR arrays of P. mirabilis isolates were expanded mostly by integration of bacteriophageal source of spacers. This study shows novel findings in the area of the P-mirabilis CRISPR-Cas system. In this regard, among analyzed genome of P. mirabilis isolates, Class I CRISR-Cas systems were dominant, and all belonged to type I-E. In the flanks of the CRISPR, some other elements with regulatory role were also found. A motif of 11 nt size was found to be preserved among the analyzed genome. We believe that it might has a CRISPR-Cas system transcription facilitator by targeting the Rho element.


Asunto(s)
Sistemas CRISPR-Cas , Proteus mirabilis/genética , Bacteriófagos/fisiología , Profagos/fisiología , Proteus mirabilis/virología
19.
mBio ; 12(2)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33785627

RESUMEN

Mycobacterium abscessus is an emerging pathogen that is often refractory to antibiotic control. Treatment is further complicated by considerable variation among clinical isolates in both their genetic constitution and their clinical manifestations. Here, we show that the prophage and plasmid mobilome is a likely contributor to this variation. Prophages and plasmids are common, abundant, and highly diverse, and code for large repertoires of genes influencing virulence, antibiotic susceptibility, and defense against viral infection. At least 85% of the strains we describe carry one or more prophages, representing at least 17 distinct and diverse sequence "clusters," integrated at 18 different attB locations. The prophages code for 19 distinct configurations of polymorphic toxin and toxin-immunity systems, each with WXG-100 motifs for export through type VII secretion systems. These are located adjacent to attachment junctions, are lysogenically expressed, and are implicated in promoting growth in infected host cells. Although the plethora of prophages and plasmids confounds the understanding of M. abscessus pathogenicity, they also provide an abundance of tools for M. abscessus engineering.IMPORTANCEMycobacterium abscessus is an important emerging pathogen that is challenging to treat with current antibiotic regimens. There is substantial genomic variation in M. abscessus clinical isolates, but little is known about how this influences pathogenicity and in vivo growth. Much of the genomic variation is likely due to the large and varied mobilome, especially a large and diverse array of prophages and plasmids. The prophages are unrelated to previously characterized phages of mycobacteria and code for a diverse array of genes implicated in both viral defense and in vivo growth. Prophage-encoded polymorphic toxin proteins secreted via the type VII secretion system are common and highly varied and likely contribute to strain-specific pathogenesis.


Asunto(s)
Bacteriófagos/fisiología , Mycobacterium abscessus/genética , Mycobacterium abscessus/virología , Plásmidos/genética , Profagos/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Bacteriófagos/genética , Variación Genética , Humanos , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium abscessus/clasificación , Mycobacterium abscessus/metabolismo , Filogenia , Plásmidos/metabolismo , Profagos/genética , Sistemas de Secreción Tipo VII/genética , Sistemas de Secreción Tipo VII/metabolismo
20.
Viruses ; 13(2)2021 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-33670076

RESUMEN

Prophages are bacteriophages in the lysogenic state, where the viral genome is inserted within the bacterial chromosome. They contribute to strain genetic variability and can influence bacterial phenotypes. Prophages are highly abundant among the strains of the opportunistic pathogen Pseudomonas aeruginosa and were shown to confer specific traits that can promote strain pathogenicity. The main difficulty of studying those regions is the lack of a simple prophage-curing method for P. aeruginosa strains. In this study, we developed a novel, targeted-curing approach for prophages in P. aeruginosa. In the first step, we tagged the prophage for curing with an ampicillin resistance cassette (ampR) and further used this strain for the sacB counter-selection marker's temporal insertion into the prophage region. The sucrose counter-selection resulted in different variants when the prophage-cured mutant is the sole variant that lost the ampR cassette. Next, we validated the targeted-curing with local PCR amplification and Whole Genome Sequencing. The application of the strategy resulted in high efficiency both for curing the Pf4 prophage of the laboratory wild-type (WT) strain PAO1 and for PR2 prophage from the clinical, hard to genetically manipulate, 39016 strain. We believe this method can support the research and growing interest in prophage biology in P. aeruginosa as well as additional Gram-negative bacteria.


Asunto(s)
Profagos/genética , Fagos Pseudomonas/genética , Pseudomonas aeruginosa/virología , Virología/métodos , Genoma Viral , Lisogenia , Reacción en Cadena de la Polimerasa , Profagos/fisiología , Fagos Pseudomonas/fisiología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...