Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.571
Filter
1.
Nat Commun ; 15(1): 8315, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333115

ABSTRACT

The spread of antibiotic resistance genes (ARGs) poses a substantial threat to human health. Phage-mediated transduction could exacerbate ARG transmission. While several case studies exist, it is yet unclear to what extent phages encode and mobilize ARGs at the global scale and whether human impacts play a role in this across different habitats. Here, we combine 38,605 bacterial genomes, 1432 metagenomes, and 1186 metatranscriptomes across 12 contrasting habitats to explore the distribution of prophages and their cargo ARGs in natural and human-impacted environments. Worldwide, we observe a significant increase in the abundance, diversity, and activity of prophage-encoded ARGs in human-impacted habitats linked with relatively higher risk of past antibiotic exposure. This effect was driven by phage-encoded cargo ARGs that could be mobilized to provide increased resistance in heterologous E. coli host for a subset of analyzed strains. Our findings suggest that human activities have altered bacteria-phage interactions, enriching ARGs in prophages and making ARGs more mobile across habitats globally.


Subject(s)
Anti-Bacterial Agents , Bacteria , Drug Resistance, Bacterial , Prophages , Prophages/genetics , Humans , Drug Resistance, Bacterial/genetics , Anti-Bacterial Agents/pharmacology , Bacteria/genetics , Bacteria/virology , Bacteria/drug effects , Genome, Bacterial/genetics , Metagenome/genetics , Ecosystem , Escherichia coli/genetics , Escherichia coli/virology , Escherichia coli/drug effects , Drug Resistance, Microbial/genetics , Genes, Bacterial
2.
Emerg Microbes Infect ; 13(1): 2399950, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39259213

ABSTRACT

ABSTRACTBetween 2018 and 2024, we conducted systematic whole-genome sequencing and phylogenomic analysis on 263 V. cholerae O1 isolates from cholera patients across four provinces in the Democratic Republic of Congo (North-Kivu, South-Kivu, Tanganyika, and Kasai Oriental). These isolates were classified into the AFR10d and AFR10e sublineages of AFR10 lineage, originating from the third wave of the seventh El Tor cholera pandemic (7PET). Compared to the strains analysed between 2014 and 2017, both sublineages had few genetic changes in the core genome but recent isolates (2022-2024) had significant CTX prophage rearrangement. AFR10e spread across all four provinces, while AFR10d appeared to be extinct by the end of 2020. Since 2022, most V. cholerae O1 isolates exhibited significant CTX prophage rearrangements, including a tandem repeat of an environmental satellite phage RS1 downstream the ctxB toxin gene of the CTX-Φ-3 prophage on the large chromosome, as well as two or more arrayed copies of an environmental pre-CTX-Φ prophage precursor on the small chromosome. We used Illumina data for mapping and coverage estimation to identify isolates with unique CTX-Φ genomic features. Gene localization was then determined on MinION-derived assemblies, revealing an organization similar to that of non-O1 V. cholerae isolates found in Asia (O139 VC1374, and environmental O4 VCE232), but never described in V. cholerae O1 El Tor from the third wave. In conclusion, while the core genome of AFR10d and AFR10e showed minimal changes, significant alterations in the CTX-Φ and pre-CTX-Φ prophage content and organization were identified in AFR10e from 2022 onwards.


Subject(s)
Cholera , Disease Outbreaks , Prophages , Humans , Cholera/microbiology , Cholera/epidemiology , Cholera Toxin/genetics , Democratic Republic of the Congo/epidemiology , Evolution, Molecular , Genome, Bacterial , Phylogeny , Prophages/genetics , Vibrio cholerae/genetics , Vibrio cholerae/virology , Vibrio cholerae/isolation & purification , Vibrio cholerae/classification , Vibrio cholerae O1/genetics , Vibrio cholerae O1/virology , Vibrio cholerae O1/isolation & purification , Whole Genome Sequencing
3.
Sci Data ; 11(1): 1018, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300139

ABSTRACT

"Candidatus Liberibacter asiaticus" (CLas) is a phloem-limited alpha-proteobacteria causing Citrus Huanglongbing, the destructive disease currently threatening global citrus industry. Genomic analyses of CLas provide insights into its evolution and biology. Here, we sequenced and assembled whole genomes of 135 CLas strains originally from 20 citrus cultivars collected at ten citrus-growing provinces in China. The resulting dataset comprised 135 CLas genomes ranging from 1,221,309 bp to 1,308,521 bp, with an average coverage of 675X. Prophage typing showed that 44 strains contained Type 1 prophage, 89 strains contained Type 2 prophage, 44 strains contained Type 3 prophage, and 34 of them contained more than one type of prophage/phage. The SNP calling identified a total of 5,090 SNPs. Genome-based phylogenetic analysis revealed two major clades among CLas strains, with Clade I dominated by CLas strains containing Type 1 prophage (79/95) and Clade II dominated by CLas strains containing Type 1 or Type 3 prophage (80/95). This CLas genome dataset provides valuable resources for studying genetic diversity and evolutionary pattern of CLas strains.


Subject(s)
Citrus , Genome, Bacterial , Phylogeny , Prophages , Whole Genome Sequencing , China , Citrus/microbiology , Prophages/genetics , Rhizobiaceae/genetics , Rhizobiaceae/classification , Polymorphism, Single Nucleotide , Plant Diseases/microbiology
4.
Microb Pathog ; 195: 106907, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39218375

ABSTRACT

Streptococcus agalactiae (S. agalactiae) is an opportunistic pathogen, and to date, studies have mainly focused on S. agalactiae strains isolated from humans, dairy cows, and fish. We reported one S. agalactiae strain, named CFFB, which was isolated from a healthy Sichuan golden snub-nosed monkey. Classical bacteriological approaches, as well as, next-generation sequencing, comparative genomics, and mice challenge test were used to characterize this strain. CFFB was identified as serotype III, ST19 combination which is a common type found in human strains. Phylogenetic analysis showed that the genome of CFFB was closely related to human clinical isolates, rather far away from animal strains. In total, CFFB contained fewer virulence-associated genes and antibiotic resistance genes than human isolates that were close to CFFB in evolutionary relationships. In the mice challenge test, CFFB had a relative weak virulence that just caused death in 33 % of ICR mice at a dose of 108 CFU by intraperitoneal injection, and CFFB was reisolated from the cardiac blood of the dead mice. Meanwhile, two intact prophages (prophage 1 and 2) were identified in the CFFB genome and shared high similarities with phage Javan52 and Javan29 which from human S. agalactiae isolate Gottschalk 1002A and RBH03, respectively. Moreover, the type II-A CRISPR-Cas system was detected in the CFFB genome, and the spacers from CFFB were the same to the streptococci isolates from human. These results suggest that CFFB isolated from healthy Sichuan golden snub-nosed monkeys may have its origin in human S. agalactiae. Our results suggested some genomic similarities between the S. agalactiae colonized in Sichuan golden snub-nosed monkey and those in infected humans.


Subject(s)
Genome, Bacterial , Phylogeny , Streptococcal Infections , Streptococcus agalactiae , Animals , Streptococcus agalactiae/genetics , Streptococcus agalactiae/isolation & purification , Streptococcus agalactiae/pathogenicity , Streptococcal Infections/microbiology , Streptococcal Infections/veterinary , China , Virulence/genetics , Mice , Colobinae/microbiology , Humans , Prophages/genetics , Mice, Inbred ICR , Virulence Factors/genetics , Serogroup , High-Throughput Nucleotide Sequencing , Monkey Diseases/microbiology
5.
Viruses ; 16(9)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39339825

ABSTRACT

Prophages can alter their bacterial hosts to prevent other phages from infecting the same cell, a mechanism known as superinfection exclusion (SIE). Such alterations are facilitated by phage interactions with critical bacterial components involved in motility, adhesion, biofilm production, conjugation, antimicrobial resistance, and immune evasion. Therefore, the impact of SIE extends beyond the immediate defense against superinfection, influencing the overall fitness and virulence of the bacteria. Evaluating the interactions between phages and their bacterial targets is critical for leading phage therapy candidates like Pseudomonas aeruginosa, a Gram-negative bacterium responsible for persistent and antibiotic-resistant opportunistic infections. However, comprehensive literature on the mechanisms underlying SIE remains scarce. Here, we provide a compilation of well-characterized and potential mechanisms employed by Pseudomonas phages to establish SIE. We hypothesize that the fitness costs imposed by SIE affect bacterial virulence, highlighting the potential role of this mechanism in the management of bacterial infections.


Subject(s)
Phage Therapy , Pseudomonas Phages , Pseudomonas aeruginosa , Superinfection , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/pathogenicity , Superinfection/microbiology , Superinfection/virology , Virulence , Pseudomonas Phages/physiology , Humans , Prophages/physiology , Prophages/genetics , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Biofilms/growth & development , Bacteriophages/physiology
6.
Viruses ; 16(8)2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39205252

ABSTRACT

The last thirty years have seen a meteoric rise in the number of sequenced bacteriophage genomes, spurred on by both the rise and success of groups working to isolate and characterize phages, and the rapid and significant technological improvements and reduced costs associated with sequencing their genomes. Over the course of these decades, the tools used to glean evolutionary insights from these sequences have grown more complex and sophisticated, and we describe here the suite of computational and bioinformatic tools used extensively by the integrated research-education communities such as SEA-PHAGES and PHIRE, which are jointly responsible for 25% of all complete phage genomes in the RefSeq database. These tools are used to integrate and analyze phage genome data from different sources, for identification and precise extraction of prophages from bacterial genomes, computing "phamilies" of related genes, and displaying the complex nucleotide and amino acid level mosaicism of these genomes. While over 50,000 SEA-PHAGES students have primarily benefitted from these tools, they are freely available for the phage community at large.


Subject(s)
Bacteriophages , Computational Biology , Genome, Viral , Genomics , Bacteriophages/genetics , Bacteriophages/isolation & purification , Bacteriophages/classification , Computational Biology/methods , Genomics/methods , Software , Prophages/genetics , Databases, Genetic
7.
Appl Environ Microbiol ; 90(9): e0043424, 2024 09 18.
Article in English | MEDLINE | ID: mdl-39189727

ABSTRACT

Gene transfer agents (GTAs) are phage-like entities that package and transfer random host genome fragments between prokaryotes. RcGTA, produced by Rhodobacter capsulatus, is hypothesized to originate from a prophage ancestor. Most of the evidence supporting this hypothesis came from the finding of RcGTA-like genes in phages. More than 75% of the RcGTA genes have a phage homolog. However, only a few RcGTA homologs have been identified in a (pro)phage genome, leaving the hypothesis that GTAs evolved from prophages through gene loss with only weak evidence. We herein report the discovery of an inducible prophage (vB_MseS-P1) from a Mesorhizobium sediminum strain that contains the largest number (12) of RcGTA homologs found in a phage genome to date. We also identified three putative prophages and two prophage remnants harboring 12-14 RcGTA homologs in a Methylobacterium nodulans strain. The protein remote homology detection also revealed more RcGTA homologs from other phages than we previously thought. Moreover, the head-tail gene architecture of these newly discovered prophage-related elements closely resembles that of RcGTA. Furthermore, vB_MseS-P1 virions have structural proteins similar to RcGTA particles. Close phylogenetic relationships between certain prophage genes and RcGTA-like genes in Alphaproteobacteria further support the shared ancestry between RcGTA and prophages. Our findings provide new relatively direct evidence of the origin of RcGTA from a prophage progenitor.IMPORTANCEGTAs are important genetic elements in certain groups of bacteria and contribute to the genetic diversification, evolution, and ecological adaptation of bacteria. RcGTA, a common type of GTA, is known to package and transfer random fragments of the bacterial genome to recipient cells. However, the origin of RcGTA is still elusive. It has been hypothesized that RcGTA evolved from a prophage ancestor through gene loss. However, the few RcGTA homologs identified in a (pro)phage genome leave the hypothesis lacking direct evidence. This study uncovers the presence of a large number of RcGTA homologs in an inducible prophage and several putative prophages. The similar head-tail gene architecture and structural protein compositions of these newly discovered prophage-related elements and RcGTA further demonstrate an unprecedentedly observed close evolutionary relationship between prophages and RcGTA. Together, our findings provide more direct evidence supporting the origin of RcGTA from prophage.


Subject(s)
Prophages , Prophages/genetics , Rhodobacter capsulatus/virology , Rhodobacter capsulatus/genetics , Gene Transfer, Horizontal , Genome, Viral , Phylogeny , Evolution, Molecular , Methylobacterium/virology , Methylobacterium/genetics
8.
PLoS Biol ; 22(8): e3002746, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39110680

ABSTRACT

Understanding the dynamic evolution of Salmonella is vital for effective bacterial infection management. This study explores the role of the flexible genome, organised in regions of genomic plasticity (RGP), in shaping the pathogenicity of Salmonella lineages. Through comprehensive genomic analysis of 12,244 Salmonella spp. genomes covering 2 species, 6 subspecies, and 46 serovars, we uncover distinct integration patterns of pathogenicity-related gene clusters into RGP, challenging traditional views of gene distribution. These RGP exhibit distinct preferences for specific genomic spots, and the presence or absence of such spots across Salmonella lineages profoundly shapes strain pathogenicity. RGP preferences are guided by conserved flanking genes surrounding integration spots, implicating their involvement in regulatory networks and functional synergies with integrated gene clusters. Additionally, we emphasise the multifaceted contributions of plasmids and prophages to the pathogenicity of diverse Salmonella lineages. Overall, this study provides a comprehensive blueprint of the pathogenicity potential of Salmonella. This unique insight identifies genomic spots in nonpathogenic lineages that hold the potential for harbouring pathogenicity genes, providing a foundation for predicting future adaptations and developing targeted strategies against emerging human pathogenic strains.


Subject(s)
Genome, Bacterial , Salmonella , Salmonella/genetics , Salmonella/pathogenicity , Genome, Bacterial/genetics , Virulence/genetics , Humans , Genomics/methods , Multigene Family , Phylogeny , Plasmids/genetics , Salmonella Infections/microbiology , Prophages/genetics , Evolution, Molecular
9.
Gut Microbes ; 16(1): 2379440, 2024.
Article in English | MEDLINE | ID: mdl-39132840

ABSTRACT

Prophages can have major clinical implications through their ability to change pathogenic bacterial traits. There is limited understanding of the prophage role in ecological, evolutionary, adaptive processes and pathogenicity of Helicobacter pylori, a widespread bacterium causally associated with gastric cancer. Inferring the exact prophage genomic location and completeness requires complete genomes. The international Helicobacter pylori Genome Project (HpGP) dataset comprises 1011 H. pylori complete clinical genomes enriched with epigenetic data. We thoroughly evaluated the H. pylori prophage genomic content in the HpGP dataset. We investigated population evolutionary dynamics through phylogenetic and pangenome analyses. Additionally, we identified genome rearrangements and assessed the impact of prophage presence on bacterial gene disruption and methylome. We found that 29.5% (298) of the HpGP genomes contain prophages, of which only 32.2% (96) were complete, minimizing the burden of prophage carriage. The prevalence of H. pylori prophage sequences was variable by geography and ancestry, but not by disease status of the human host. Prophage insertion occasionally results in gene disruption that can change the global bacterial epigenome. Gene function prediction allowed the development of the first model for lysogenic-lytic cycle regulation in H. pylori. We have disclosed new prophage inactivation mechanisms that appear to occur by genome rearrangement, merger with other mobile elements, and pseudogene accumulation. Our analysis provides a comprehensive framework for H. pylori prophage biological and genomics, offering insights into lysogeny regulation and bacterial adaptation to prophages.


Subject(s)
Genome, Bacterial , Genomics , Helicobacter pylori , Phylogeny , Prophages , Helicobacter pylori/genetics , Helicobacter pylori/virology , Prophages/genetics , Prophages/physiology , Humans , Helicobacter Infections/microbiology
10.
Water Res ; 263: 122200, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39111212

ABSTRACT

Prophages are prevalent among bacterial species, including strains carrying antibiotic resistance genes (ARGs). Prophage induction can be triggered by the SOS response to stressors, leading to cell lysis. In environments polluted by chemical stressors, ARGs and prophage co-harboring strains might pose an unknown risk of spreading ARGs through chemical pollutant-mediated prophage induction and subsequent cell lysis. In this study, we investigated the effects of common non-antibiotic water pollutants, triclosan and silver nanoparticles, on triggering prophage induction in clinical isolates carrying ARGs and the subsequent uptake of released ARGs by the naturally competent bacterium Acinetobacter baylyi. Our results demonstrate that both triclosan and silver nanoparticles, at environmentally relevant concentrations and those found in commercial products, significantly enhance prophage induction among various clinical isolates. Transmission electron microscopy imaging and plaque assays confirmed the production of infectious phage particles under non-antibiotic pollutants-mediated prophage induction. In addition, the rate of ARG transformation to A. baylyi significantly increased after the release of extracellular ARGs from prophage induction-mediated cell lysis. The mechanism of non-antibiotic pollutants-mediated prophage induction is primarily associated with excessive oxidative stress, which provokes the SOS response. Our findings offer insights into the role of non-antibiotic pollutants in promoting the dissemination of ARGs by triggering prophage induction.


Subject(s)
Prophages , Prophages/genetics , Acinetobacter/drug effects , Acinetobacter/genetics , Drug Resistance, Microbial/genetics , Triclosan/pharmacology , Drug Resistance, Bacterial/genetics , Anti-Bacterial Agents/pharmacology , Metal Nanoparticles , Silver/pharmacology
11.
Microbiology (Reading) ; 170(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-39132742

ABSTRACT

Genome sequencing of Clostridium clostridioforme strain LM41 revealed the presence of an atypically high proportion of mobile genetic elements for this species, with a particularly high abundance of prophages. Bioinformatic analysis of prophage sequences sought to characterize these elements and identify prophage-linked genes contributing to enhanced fitness of the host bacteria in the dysbiotic gut. Using PHASTER, PhageScope and manual curation, this work has identified 15 prophages: 4 predicted to be intact, 2 predicted to be defective and 9 which are unclassified. Quantitative PCR (qPCR) analysis revealed spontaneous release of four of the LM41 prophages (φ1, φ2, φ4 and φ10) into the culture supernatant, with virion-like particles visualized using transmission electron microscopy. The majority (12/14) of these particles had morphology akin to podoviruses, which is consistent with morphology predictions for φ1 and φ4. We observed diversity in the lysogeny mechanisms utilized by the prophages, with examples of the classical λ-like CI/Cro system, the ICEBs1 ImmR/ImmA-like system and the Mu-like C/Ner system. Classical morons, such as toxins or immune evasion factors, were not observed. We did, however, identify a variety of genes with roles in mediating restriction modification and genetic diversity, as well as some candidate genes with potential roles in host adaptation. Despite being the most abundant entities in the intestine, there is a dearth of information about phages associated with members of the microbiome. This work begins to shed light on the contribution of these elements to the lifestyle of C. clostridioforme LM41.


Subject(s)
Clostridium , Gastrointestinal Microbiome , Prophages , Prophages/genetics , Clostridium/virology , Clostridium/genetics , Lysogeny , Genome, Bacterial , Genome, Viral , Genomics , Computational Biology
12.
Nat Commun ; 15(1): 7539, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39215040

ABSTRACT

Many bacterial immune systems recognize phage structural components to activate antiviral responses, without inhibiting the function of the phage component. These systems can be encoded in specific chromosomal loci, known as defense islands, and in mobile genetic elements such as prophages and phage-inducible chromosomal islands (PICIs). Here, we identify a family of bacterial immune systems, named Tai (for 'tail assembly inhibition'), that is prevalent in PICIs, prophages and P4-like phage satellites. Tai systems protect their bacterial host population from other phages by blocking the tail assembly step, leading to the release of tailless phages incapable of infecting new hosts. To prevent autoimmunity, some Tai-positive phages have an associated counter-defense mechanism that is expressed during the phage lytic cycle and allows for tail formation. Interestingly, the Tai defense and counter-defense genes are organized in a non-contiguous operon, enabling their coordinated expression.


Subject(s)
Bacteriophages , Prophages , Bacteriophages/genetics , Bacteriophages/physiology , Prophages/genetics , Genomic Islands/genetics , Bacteria/virology , Bacteria/genetics , Bacteria/immunology , Operon/genetics , Escherichia coli/genetics , Escherichia coli/virology , Escherichia coli/immunology , Gene Expression Regulation, Bacterial
13.
Nat Commun ; 15(1): 7244, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39174532

ABSTRACT

The filamentous 'Pf' bacteriophages of Pseudomonas aeruginosa play roles in biofilm formation and virulence, but mechanisms governing Pf prophage activation in biofilms are unclear. Here, we identify a prophage regulatory module, KKP (kinase-kinase-phosphatase), that controls virion production of co-resident Pf prophages and mediates host defense against diverse lytic phages. KKP consists of Ser/Thr kinases PfkA and PfkB, and phosphatase PfpC. The kinases have multiple host targets, one of which is MvaU, a host nucleoid-binding protein and known prophage-silencing factor. Characterization of KKP deletion and overexpression strains with transcriptional, protein-level and prophage-based approaches indicates that shifts in the balance between kinase and phosphatase activities regulate phage production by controlling MvaU phosphorylation. In addition, KKP acts as a tripartite toxin-antitoxin system that provides defense against some lytic phages. A conserved lytic phage replication protein inhibits the KKP phosphatase PfpC, stimulating toxic kinase activity and blocking lytic phage production. Thus, KKP represents a phosphorylation-based mechanism for prophage regulation and antiphage defense. The conservation of KKP gene clusters in >1000 diverse temperate prophages suggests that integrated control of temperate and lytic phage infection by KKP-like regulatory modules may play a widespread role in shaping host cell physiology.


Subject(s)
Lysogeny , Prophages , Pseudomonas aeruginosa , Lysogeny/genetics , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/genetics , Prophages/genetics , Prophages/physiology , Phosphorylation , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Pseudomonas Phages/genetics , Pseudomonas Phages/metabolism , Biofilms/growth & development , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Gene Expression Regulation, Viral
14.
Gut Microbes ; 16(1): 2390720, 2024.
Article in English | MEDLINE | ID: mdl-39167701

ABSTRACT

Phages are the most diversified and dominant members of the gut virobiota. They play a crucial role in shaping the structure and function of the gut microbial community and consequently the health of humans and animals. Phages are found mainly in the mucus, from where they can translocate to the intestinal organs and act as a modulator of gut microbiota. Understanding the vital role of phages in regulating the composition of intestinal microbiota and influencing human and animal health is an emerging area of research. The relevance of phages in the gut ecosystem is supported by substantial evidence, but the importance of phages in shaping the gut microbiota remains unclear. Although information regarding general phage ecology and development has accumulated, detailed knowledge on phage-gut microbe and phage-human interactions is lacking, and the information on the effects of phage therapy in humans remains ambiguous. In this review, we systematically assess the existing data on the structure and ecology of phages in the human and animal gut environments, their development, possible interaction, and subsequent impact on the gut ecosystem dynamics. We discuss the potential mechanisms of prophage activation and the subsequent modulation of gut bacteria. We also review the link between phages and the immune system to collect evidence on the effect of phages on shaping the gut microbial composition. Our review will improve understanding on the influence of phages in regulating the gut microbiota and the immune system and facilitate the development of phage-based therapies for maintaining a healthy and balanced gut microbiota.


Subject(s)
Bacteria , Bacteriophages , Gastrointestinal Microbiome , Humans , Bacteriophages/physiology , Gastrointestinal Microbiome/physiology , Animals , Bacteria/virology , Bacteria/classification , Phage Therapy , Prophages/physiology , Prophages/genetics
15.
mSystems ; 9(9): e0080124, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39166874

ABSTRACT

The opportunistic human pathogen Pseudomonas aeruginosa is naturally infected by a large class of temperate, transposable, Mu-like phages. We examined the genotypic and phenotypic diversity of P. aeruginosa PA14 lysogen populations as they resolve clustered regularly interspaced short palindromic repeat (CRISPR) autoimmunity, mediated by an imperfect CRISPR match to the Mu-like DMS3 prophage. After 12 days of evolution, we measured a decrease in spontaneous induction in both exponential and stationary phase growth. Co-existing variation in spontaneous induction rates in the exponential phase depended on the way the coexisting strains resolved genetic conflict. Multiple mutational modes to resolve genetic conflict between host and phage resulted in coexistence in evolved populations of single lysogens that maintained CRISPR immunity to other phages and polylysogens that lost immunity completely. This work highlights a new dimension of the role of lysogenic phages in the evolution of their hosts.IMPORTANCEThe chronic opportunistic multi-drug-resistant pathogen Pseudomonas aeruginosa is persistently infected by temperate phages. We assess the contribution of temperate phage infection to the evolution of the clinically relevant strain UCBPP-PA14. We found that a low level of clustered regularly interspaced short palindromic repeat (CRISPR)-mediated self-targeting resulted in polylysogeny evolution and large genome rearrangements in lysogens; we also found extensive diversification in CRISPR spacers and cas genes. These genomic modifications resulted in decreased spontaneous induction in both exponential and stationary phase growth, increasing lysogen fitness. This work shows the importance of considering latent phage infection in characterizing the evolution of bacterial populations.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Lysogeny , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/genetics , Lysogeny/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Pseudomonas Phages/genetics , Evolution, Molecular , Bacteriophages/genetics , Prophages/genetics
16.
Annu Rev Virol ; 11(1): 343-362, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38950439

ABSTRACT

Bacterial viruses known as phages rely on their hosts for replication and thus have developed an intimate partnership over evolutionary time. The survival of temperate phages, which can establish a chronic infection in which their genomes are maintained in a quiescent state known as a prophage, is tightly coupled with the survival of their bacterial hosts. As a result, prophages encode a diverse antiphage defense arsenal to protect themselves and the bacterial host in which they reside from further phage infection. Similarly, the survival and success of prophage-related elements such as phage-inducible chromosomal islands are directly tied to the survival and success of their bacterial host, and they also have been shown to encode numerous antiphage defenses. Here, we describe the current knowledge of antiphage defenses encoded by prophages and prophage-related mobile genetic elements.


Subject(s)
Bacteria , Interspersed Repetitive Sequences , Prophages , Prophages/genetics , Prophages/physiology , Bacteria/virology , Bacteria/genetics , Bacteriophages/genetics , Bacteriophages/physiology
17.
Sci Rep ; 14(1): 16248, 2024 07 15.
Article in English | MEDLINE | ID: mdl-39009624

ABSTRACT

Psyllid species, including the potato psyllid (PoP) Bactericera cockerelli (Sulc) (Triozidae) serve as host and vector of "Candidatus Liberibacter spp." ("Ca. Liberibacter"), which also infects diverse plant hosts, including citrus and tomato. Psyllid transmission of "Ca. Liberibacter" is circulative and propagative. The time of "Ca. Liberibacter" acquisition and therefore vector life stage most competent for bacterial transmission varies by pathosystems. Here, the potato psyllid-"Ca. Liberibacter solanacearum" (CLso) pathosystem was investigated to dissect CLso-prophage interactions in the tomato plant and PoP-psyllid host by real-time quantitative reverse transcriptase amplification of CLso genes/loci with predicted involvement in host infection and psyllid-CLso transmission. Genes/loci analyzed were associated with (1) CLso-adhesion, -invasion, -pathogenicity, and -motility, (2) prophage-adhesion and pathogenicity, and (3) CLso-lysogenic cycle. Relative gene expression was quantified by qRT-PCR amplification from total RNA isolated from CLso-infected 1st-2nd and 4th-5th nymphs and teneral adults and CLso-infected tomato plants in which CLso infection is thought to occur without SC1-SC2 replication. Gene/loci expression was host-dependent and varied with the psyllid developmental stage. Loci previously associated with repressor-anti-repressor regulation in the "Ca Liberibacter asiaticus"-prophage pathosystem, which maintains the lysogenic cycle in Asian citrus psyllid Diaphorina citri, were expressed in CLso-infected psyllids but not in CLso-infected tomato plants.


Subject(s)
Hemiptera , Plant Diseases , Prophages , Solanum lycopersicum , Animals , Hemiptera/microbiology , Prophages/genetics , Solanum lycopersicum/microbiology , Plant Diseases/microbiology , Solanum tuberosum/microbiology , Solanum tuberosum/parasitology , Insect Vectors/microbiology , Rhizobiaceae/genetics , Gene Expression Regulation, Bacterial , Life Cycle Stages/genetics
18.
Nat Commun ; 15(1): 6291, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39060226

ABSTRACT

Malawi experienced its deadliest Vibrio cholerae (Vc) outbreak following devastating cyclones, with >58,000 cases and >1700 deaths reported between March 2022 and May 2023. Here, we use population genomics to investigate the attributes and origin of the Malawi 2022-2023 Vc outbreak isolates. Our results demonstrate the predominance of ST69 clone, also known as the seventh cholera pandemic El Tor (7PET) lineage, expressing O1 Ogawa (~ 80%) serotype followed by Inaba (~ 16%) and sporadic non-O1/non-7PET serogroups (~ 4%). Phylogenetic reconstruction revealed that the Malawi outbreak strains correspond to a recent importation from Asia into Africa (sublineage AFR15). These isolates harboured known antimicrobial resistance and virulence elements, notably the ICEGEN/ICEVchHai1/ICEVchind5 SXT/R391-like integrative conjugative elements and a CTXφ prophage with the ctxB7 genotype compared to historical Malawian Vc isolates. These data suggest that the devastating cyclones coupled with the recent importation of 7PET serogroup O1 strains, may explain the magnitude of the 2022-2023 cholera outbreak in Malawi.


Subject(s)
Cholera , Disease Outbreaks , Phylogeny , Vibrio cholerae , Malawi/epidemiology , Cholera/epidemiology , Cholera/microbiology , Humans , Vibrio cholerae/genetics , Vibrio cholerae/classification , Genomics , Genome, Bacterial/genetics , Prophages/genetics , Genotype , Serogroup
19.
ISME J ; 18(1)2024 Jan 08.
Article in English | MEDLINE | ID: mdl-39023219

ABSTRACT

Arms races between mobile genetic elements and prokaryotic hosts are major drivers of ecological and evolutionary change in microbial communities. Prokaryotic defense systems such as CRISPR-Cas have the potential to regulate microbiome composition by modifying the interactions among bacteria, plasmids, and phages. Here, we used longitudinal metagenomic data from 130 healthy and diseased individuals to study how the interplay of genetic parasites and CRISPR-Cas immunity reflects on the dynamics and composition of the human gut microbiome. Based on the coordinated study of 80 000 CRISPR-Cas loci and their targets, we show that CRISPR-Cas immunity effectively modulates bacteriophage abundances in the gut. Acquisition of CRISPR-Cas immunity typically leads to a decrease in the abundance of lytic phages but does not necessarily cause their complete disappearance. Much smaller effects are observed for lysogenic phages and plasmids. Conversely, phage-CRISPR interactions shape bacterial microdiversity by producing weak selective sweeps that benefit immune host lineages. We also show that distal (and chronologically older) regions of CRISPR arrays are enriched in spacers that are potentially functional and target crass-like phages and local prophages. This suggests that exposure to reactivated prophages and other endemic viruses is a major selective pressure in the gut microbiome that drives the maintenance of long-lasting immune memory.


Subject(s)
Bacteria , Bacteriophages , CRISPR-Cas Systems , Gastrointestinal Microbiome , Humans , Bacteriophages/genetics , Bacteria/genetics , Bacteria/classification , Bacteria/virology , Metagenomics , Plasmids/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Host Microbial Interactions , Prophages/genetics , Prophages/physiology
20.
ISME J ; 18(1)2024 Jan 08.
Article in English | MEDLINE | ID: mdl-39030686

ABSTRACT

Reef-building corals depend on an intricate community of microorganisms for functioning and resilience. The infection of coral-associated bacteria by bacteriophages can modify bacterial ecological interactions, yet very little is known about phage functions in the holobiont. This gap stems from methodological limitations that have prevented the recovery of high-quality viral genomes and bacterial host assignment from coral samples. Here, we introduce a size fractionation approach that increased bacterial and viral recovery in coral metagenomes by 9-fold and 2-fold, respectively, and enabled the assembly and binning of bacterial and viral genomes at relatively low sequencing coverage. We combined these viral genomes with those derived from 677 publicly available metagenomes, viromes, and bacterial isolates from stony corals to build a global coral virus database of over 20,000 viral genomic sequences spanning four viral realms. The tailed bacteriophage families Kyanoviridae and Autographiviridae were the most abundant, replacing groups formerly referred to as Myoviridae and Podoviridae, respectively. Prophage and CRISPR spacer linkages between these viruses and 626 bacterial metagenome-assembled genomes and bacterial isolates showed that most viruses infected Alphaproteobacteria, the most abundant class, and less abundant taxa like Halanaerobiia and Bacteroidia. A host-phage-gene network identified keystone viruses with the genomic capacity to modulate bacterial metabolic pathways and direct molecular interactions with eukaryotic cells. This study reveals the genomic basis of nested symbioses between bacteriophage, bacteria, and the coral host and its endosymbiotic algae.


Subject(s)
Anthozoa , Bacteria , Bacteriophages , Genome, Viral , Bacteriophages/genetics , Bacteriophages/isolation & purification , Bacteriophages/classification , Anthozoa/virology , Anthozoa/microbiology , Animals , Bacteria/virology , Bacteria/genetics , Bacteria/classification , Metagenome , Symbiosis , Coral Reefs , Virome/genetics , Prophages/genetics
SELECTION OF CITATIONS
SEARCH DETAIL