Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.476
Filter
1.
BMC Vet Res ; 20(1): 413, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39272082

ABSTRACT

BACKGROUND: Due to the diversity of Shiga toxin-producing Escherichia coli (STEC) isolates, detecting highly pathogenic strains in foodstuffs is challenging. Currently, reference protocols for STEC rely on the molecular detection of eae and the stx1 and/or stx2 genes, followed by the detection of serogroup-specific wzx or wzy genes related to the top 7 serogroups. However, these screening methods do not distinguish between samples in which a STEC possessing both determinants are present and those containing two or more organisms, each containing one of these genes. This study aimed to evaluate ecf1, Z2098, Z2099, and nleA genes as single markers and their combinations (ecf1/Z2098, ecf1/Z2099, ecf1/nleA, Z2098/Z2099, Z2098/nleA, and Z2099/nleA) as genetic markers to detect potentially pathogenic STEC by the polymerase chain reaction (PCR) in 96 animal samples, as well as in 52 whole genome sequences of human samples via in silico PCR analyses. RESULTS: In animal isolates, Z2098 and Z2098/Z2099 showed a strong association with the detected top 7 isolates, with 100% and 69.2% of them testing positive, respectively. In human isolates, Z2099 was detected in 95% of the top 7 HUS isolates, while Z2098/Z2099 and ecf1/Z2099 were detected in 87.5% of the top 7 HUS isolates. CONCLUSIONS: Overall, using a single gene marker, Z2098, Z2099, and ecf1 are sensitive targets for screening the top 7 STEC isolates, and the combination of Z2098/Z2099 offers a more targeted initial screening method to detect the top 7 STEC isolates. Detecting non-top 7 STEC in both animal and human samples proved challenging due to inconsistent characteristics associated with the genetic markers studied.


Subject(s)
Enterohemorrhagic Escherichia coli , Escherichia coli Infections , Shiga-Toxigenic Escherichia coli , Shiga-Toxigenic Escherichia coli/genetics , Shiga-Toxigenic Escherichia coli/isolation & purification , Animals , Genetic Markers , Escherichia coli Infections/veterinary , Escherichia coli Infections/microbiology , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/isolation & purification , Humans , Plasmids/genetics , Computer Simulation , Cattle , Polymerase Chain Reaction/veterinary , Sheep , Genomic Islands/genetics
2.
J Bacteriol ; 206(9): e0014324, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39230524

ABSTRACT

A major challenge faced by Vibrio cholerae is constant predation by bacteriophage (phage) in aquatic reservoirs and during infection of human hosts. To overcome phage predation, V. cholerae has acquired and/or evolved a myriad of phage defense systems. Although several novel defense systems have been discovered, we hypothesized that more were encoded in V. cholerae given the low diversity of phages that have been isolated, which infect this species. Using a V. cholerae genomic library, we identified a Type IV restriction system consisting of two genes within a 16-kB region of the Vibrio pathogenicity island-2, which we name TgvA and TgvB (Type I-embedded gmrSD-like system of VPI-2). We show that both TgvA and TgvB are required for defense against T2, T4, and T6 by targeting glucosylated 5-hydroxymethylcytosine (5hmC). T2 or T4 phages that lose the glucose modifications are resistant to TgvAB defense but exhibit a significant evolutionary tradeoff, becoming susceptible to other Type IV restriction systems that target unglucosylated 5hmC. We also show that the Type I restriction-modification system that embeds the tgvAB genes protects against phage T3, secΦ18, secΦ27, and λ, suggesting that this region is a phage defense island. Our study uncovers a novel Type IV restriction system in V. cholerae, increasing our understanding of the evolution and ecology of V. cholerae, while highlighting the evolutionary interplay between restriction systems and phage genome modification.IMPORTANCEBacteria are constantly being predated by bacteriophage (phage). To counteract this predation, bacteria have evolved a myriad of defense systems. Some of these systems specifically digest infecting phage by recognizing unique base modifications present on the phage DNA. In this study, we discover a Type IV restriction system encoded in V. cholerae, which we name TgvAB, and demonstrate it recognizes and restricts phage that have 5-hydroxymethylcytosine glucosylated DNA. Moreover, the evolution of resistance to TgvAB render phage susceptible to other Type IV restriction systems, demonstrating a significant evolutionary tradeoff. These results enhance our understanding of the evolution of V. cholerae and more broadly how bacteria evade phage predation.


Subject(s)
5-Methylcytosine , Bacteriophages , Vibrio cholerae , Vibrio cholerae/virology , Vibrio cholerae/genetics , 5-Methylcytosine/metabolism , 5-Methylcytosine/analogs & derivatives , Bacteriophages/genetics , Bacteriophages/physiology , Genomic Islands , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
3.
Bull Exp Biol Med ; 177(4): 465-469, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39264563

ABSTRACT

Two bacteriophages specifically active against to pathogenic strains of the Salmonella genus were isolated. The morphology of phage colonies (size, transparency, and shape of the plaque edge, and halo) and the spectrum of their lytic activity and interaction with microbial cells (adsorption rate, duration of the latency, and reproductive efficiency) were examined. Using genome-wide sequencing, we determined the taxonomic position of bacteriophages and verified the absence of unwanted genes encoding toxins, adhesins, and invasins, as well as pathogenicity islands responsible for antibiotic resistance. In addition, phage stability under different physical conditions and their productivity were studied.


Subject(s)
Phage Therapy , Salmonella Phages , Salmonella Phages/genetics , Salmonella Phages/isolation & purification , Humans , Salmonella Infections/microbiology , Salmonella Infections/therapy , Salmonella Infections/drug therapy , Salmonella/virology , Salmonella/drug effects , Salmonella/genetics , Genome, Viral/genetics , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Genomic Islands/genetics
4.
Microb Genom ; 10(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-39235832

ABSTRACT

Several authors have attributed the explosive outbreak of gastroenteritis that occurred in Czechoslovakia in 1965 to a toxigenic strain of Vibrio cholerae serogroup O37 based on unverified metadata associated with three particular strains from the American Type Culture Collection. Here, by sequencing the original strain preserved at the Czech National Collection of Type Cultures since 1966, we show that the strain responsible for this outbreak was actually a V. cholerae O5 that lacks the genes encoding the cholera toxin, the toxin-coregulated pilus protein and Vibrio pathogenicity islands present in V. cholerae O37 strains.


Subject(s)
Cholera , Disease Outbreaks , Gastroenteritis , Vibrio cholerae , Gastroenteritis/microbiology , Gastroenteritis/epidemiology , Gastroenteritis/history , Humans , Vibrio cholerae/genetics , Vibrio cholerae/classification , Czechoslovakia , Cholera/epidemiology , Cholera/microbiology , Cholera/history , Cholera Toxin/genetics , Genomic Islands , Serogroup
5.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273334

ABSTRACT

Listeria pathogenicity island 1 (LIPI-1) is a genetic region containing a cluster of genes essential for virulence of the bacterial pathogen Listeria monocytogenes. Main virulence factors in LIPI-1 include long 5' untranslated regions (5'UTRs), among which is Rli51, a small RNA (sRNA) in the 5'UTR of the Zn-metalloprotease-coding mpl. So far, Rli51 function and molecular mechanisms have remained obscure. Here, we show that Rli51 exhibits a dual mechanism of regulation, functioning as a cis- and as a trans-acting sRNA. Under nutrient-rich conditions, rli51-mpl transcription is prematurely terminated, releasing a short 121-nucleotide-long sRNA. Rli51 is predicted to function as a transcription attenuator that can fold into either a terminator or a thermodynamically more stable antiterminator. We show that the sRNA Rli21/RliI binds to a single-stranded RNA loop in Rli51, which is essential to mediate premature transcription termination, suggesting that sRNA binding could stabilize the terminator fold. During intracellular infection, rli51 transcription is increased, which generates a higher abundance of the short Rli51 sRNA and allows for transcriptional read-through into mpl. Comparative intracellular bacterial transcriptomics in rli51-null mutants and the wild-type reference strain EGD-e suggests that Rli51 upregulates iron-scavenging proteins and downregulates virulence factors from LIPI-1. MS2 affinity purification confirmed that Rli51 binds transcripts of the heme-binding protein Lmo2186 and Lmo0937 in vivo. These results prove that Rli51 functions as a trans-acting sRNA in intracellular bacteria. Our research shows a growth condition-dependent mechanism of regulation for Rli51, preventing unintended mpl transcription in extracellular bacteria and regulating genes important for virulence in intracellular bacteria.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial , Listeria monocytogenes , RNA, Bacterial , RNA, Small Untranslated , Listeria monocytogenes/pathogenicity , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Genomic Islands/genetics , Transcription, Genetic , 5' Untranslated Regions , Virulence/genetics , Virulence Factors/genetics , Virulence Factors/metabolism , Humans , Listeriosis/microbiology
6.
Proc Natl Acad Sci U S A ; 121(39): e2409843121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39288183

ABSTRACT

The opportunistic pathogen Acinetobacter baumannii, carries variants of A. baumannii resistance islands (AbaR)-type genomic islands conferring multidrug resistance. Their pervasiveness in the species has remained enigmatic. The dissemination of AbaRs is intricately linked to their horizontal transfer via natural transformation, a process through which bacteria can import and recombine exogenous DNA, effecting allelic recombination, genetic acquisition, and deletion. In experimental populations of the closely related pathogenic Acinetobacter nosocomialis, we quantified the rates at which these natural transformation events occur between individuals. When integrated into a model of population dynamics, they lead to the swift removal of AbaRs from the population, contrasting with the high prevalence of AbaRs in genomes. Yet, genomic analyses show that nearly all AbaRs specifically disrupt comM, a gene encoding a helicase critical for natural transformation. We found that such disruption impedes gene acquisition, and deletion, while moderately impacting acquisition of single nucleotide polymorphism. A mathematical evolutionary model demonstrates that AbaRs inserted into comM gain a selective advantage over AbaRs inserted in sites that do not inhibit or completely inhibit transformation, in line with the genomic observations. The persistence of AbaRs can be ascribed to their integration into a specific gene, diminishing the likelihood of their removal from the bacterial genome. This integration preserves the acquisition and elimination of alleles, enabling the host bacterium-and thus its AbaR-to adapt to unpredictable environments and persist over the long term. This work underscores how manipulation of natural transformation by mobile genetic elements can drive the prevalence of multidrug resistance.


Subject(s)
Acinetobacter baumannii , Genomic Islands , Acinetobacter baumannii/genetics , Acinetobacter baumannii/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Anti-Bacterial Agents/pharmacology , Gene Transfer, Horizontal , Transformation, Bacterial , Polymorphism, Single Nucleotide , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
7.
Microb Pathog ; 195: 106900, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39208964

ABSTRACT

Campylobacter jejuni (C. jejuni) is a major cause of gastroenteritis and rarely cause bloodstream infection. Herein, we characterized a multidrug-resistant C. jejuni strain LZCJ isolated from a tumor patient with bloodstream infection. LZCJ was resistant to norfloxacin, ampicillin, ceftriaxone, ciprofloxacin and tetracycline. It showed high survival rate in serum and acidic environment. Whole genome sequencing (WGS) analysis revealed that strain LZCJ had a single chromosome of 1,629,078 bp (30.6 % G + C content) and belonged to the ST137 lineage. LZCJ shared the highest identity of 99.66 % with the chicken-derived C. jejuni MTVDSCj20. Four antimicrobial resistance genes (ARGs) were detected, blaOXA-61, tet(O), gyrA (T86I), and cmeR (G144D and S207G). In addition, a 12,746 bp genomic island GI_LZCJ carrying 15 open reading frames (ORFs) including the resistance gene tet(O) was identified. Sequence analysis found that the GI_LZCJ was highly similar to the duck-derived C. jejuni ZS004, but with an additional ISChh1-like sequence. 137 non-synonymous mutations in motility related genes (flgF, fapR, flgS), capsular polysaccharide (CPS) coding genes (kpsE, kpsF, kpsM, kpsT), metabolism associated genes (nuoF, nuoG, epsJ, holB), and transporter related genes (comEA, gene0911) were confirmed in LZCJ compared with the best closed chicken-derived strain MTVDSCj20. Our study showed that C. jejuni strain LZCJ was highly similar to the chicken-derived strain MTVDSCj20 but with a lot of SNPs involved in motility, CPS and metabolism coding genes. This strain possessed a tet(O)-positive genomic island GI_LZCJ, which was closed to duck-derived C. jejuni ZS004, but with an additional ISChh1-like sequence. The above data indicated that the LZCJ strain may originate from foodborne bacteria on animals and the importance of continuous surveillance for the spread of foodborne bacteria.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Campylobacter Infections , Campylobacter jejuni , Drug Resistance, Multiple, Bacterial , Genomic Islands , Microbial Sensitivity Tests , Whole Genome Sequencing , Campylobacter jejuni/genetics , Campylobacter jejuni/drug effects , Campylobacter jejuni/isolation & purification , Drug Resistance, Multiple, Bacterial/genetics , Humans , Genomic Islands/genetics , Campylobacter Infections/microbiology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Genome, Bacterial , Base Composition , Bacteremia/microbiology , Animals , Phylogeny , Open Reading Frames , Carrier Proteins
8.
Nat Commun ; 15(1): 7137, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164300

ABSTRACT

The histone-like nucleoid structuring (H-NS) protein is a DNA binding factor, found in gammaproteobacteria, with functional equivalents in diverse microbes. Universally, such proteins are understood to silence transcription of horizontally acquired genes. Here, we identify transposon capture as a major overlooked function of H-NS. Using genome-scale approaches, we show that H-NS bound regions are transposition "hotspots". Since H-NS often interacts with pathogenicity islands, such targeting creates clinically relevant phenotypic diversity. For example, in Acinetobacter baumannii, we identify altered motility, biofilm formation, and interactions with the human immune system. Transposon capture is mediated by the DNA bridging activity of H-NS and, if absent, more ubiquitous transposition results. Consequently, transcribed and essential genes are disrupted. Hence, H-NS directs transposition to favour evolutionary outcomes useful for the host cell.


Subject(s)
Acinetobacter baumannii , Bacterial Proteins , DNA Transposable Elements , DNA-Binding Proteins , DNA Transposable Elements/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Acinetobacter baumannii/genetics , Acinetobacter baumannii/metabolism , Humans , Biofilms/growth & development , Gene Expression Regulation, Bacterial , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Genome, Bacterial , Genomic Islands
9.
Appl Environ Microbiol ; 90(9): e0020124, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39136487

ABSTRACT

Salmonella enterica serovar Typhimurium, which is a common foodborne pathogen, causes both intestinal and systemic infections in hosts. Salmonella has a complex pathogenic mechanism that involves invasive capacity and intracellular survivability, which hampers research on virulence of Salmonella. The virulence of Salmonella is primarily studied through Salmonella pathogenicity islands (SPIs). However, there are also genes outside these SPIs that significantly impact virulence. Macrophage survival gene msgA is positioned at a region independent of the SPIs and conserved in Salmonella. However, there has been limited research on msgA to date. This study aims to investigate the virulent function of msgA to deepen our understanding of Salmonella virulence. Proteomic and RT-qPCR analyses reveal that MsgA influences multiple metabolic pathways and the expression of SPIs. The depletion of msgA led to the significantly reduced invasive capacity and intracellular survivability, and thus the decreased virulence of Salmonella. In conclusion, our study suggests that MsgA is an important regulator that mainly regulates virulence. Further research into the function of MsgA will enhance the understanding of Salmonella pathogenesis and promote the application of Salmonella for medical treatment. IMPORTANCE: Salmonella enterica serovar Typhimurium is a common foodborne pathogen, it has a complex pathogenic mechanism that involves invasive capacity and intracellular survivability. The virulence of Salmonella is primarily studied through its pathogenicity islands. In contrast, virulence genes located outside the Salmonella pathogenicity islands (SPIs) have received less attention. Macrophage survival gene (MsgA) is positioned at a region independent of the SPIs and conserved in Salmonella. Our research indicates that MsgA is a novel global regulator influencing the metabolic pathways and SPIs. Further research into the function of MsgA will enhance the understanding of Salmonella pathogenesis and promote the application of Salmonella for medical treatment.


Subject(s)
Bacterial Proteins , Salmonella typhimurium , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/genetics , Salmonella typhimurium/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mice , Virulence , Animals , Genomic Islands , Macrophages/microbiology , Salmonella Infections/microbiology , Humans , RAW 264.7 Cells , Gene Expression Regulation, Bacterial , Carbon-Oxygen Lyases
10.
Biomedica ; 44(2): 258-276, 2024 05 30.
Article in English, Spanish | MEDLINE | ID: mdl-39088536

ABSTRACT

In Salmonella enterica serovar Typhimurium (Typhimurium), multidrug resistance is associated with integrons carrying resistance genes dispersed by mobile genetic elements. This exploratory systematic review sought to identify integron types and their resistance genes in multidrug resistance Typhimurium isolates. We used Medline, PubMed, SciELO, ScienceDirect, Redalyc, and Google Scholar as motor searchers for articles in Spanish or English published between 2012 and 2020, including the keywords "integrons", "antibiotic resistance", and "Salmonella Typhimurium". We included 38 articles reporting multidrug resistance up to five antibiotic families. Class 1 integrons with aadA2 and blaPSE-1 gene cassettes were predominant, some probably related to the Salmonella genomic island 1. We did not find studies detailing class 1 and 2 integrons in the same isolate, nor class 3 integrons reported. The presence of integrons largely explains the resistance profiles found in isolates from different sources in 15 countries.


La multirresistencia a los antibióticos en Salmonella enterica serovar Typhimurium (Typhimurium) se asocia con integrones que portan genes de resistencia y que son dispersados por elementos genéticos móviles. En esta revisión sistemática exploratoria, se buscó identificar los tipos de integrones y sus genes de resistencia en aislamientos de Typhimurium multirresistentes a antibióticos. Se realizó una búsqueda de artículos en Medline, PubMed, SciELO, ScienceDirect, Redalyc y Google Académico, publicados entre el 2012 y el 2020, en español o inglés, con las palabras claves: "integrons", "antibiotic resistance" y "Salmonella Typhimurium". En el análisis se incluyeron 38 artículos que reportaron multirresistencia a cinco familias de antibióticos. Los integrones de clase 1 con casetes de genes aadA2 y blaPSE-1 fueron los predominantes, algunos probablemente relacionados con la isla genómica de Salmonella 1. No se encontraron integrones de clase 1 y 2 en un mismo aislamiento, ni se reportaron integrones de clase 3. La presencia de integrones explica en gran medida los perfiles de resistencia encontrados en aislamientos de diferentes fuentes de 15 países.


Subject(s)
Drug Resistance, Multiple, Bacterial , Integrons , Salmonella typhimurium , Integrons/genetics , Drug Resistance, Multiple, Bacterial/genetics , Salmonella typhimurium/genetics , Salmonella typhimurium/drug effects , Humans , Anti-Bacterial Agents/pharmacology , Salmonella Infections/microbiology , Salmonella Infections/epidemiology , Genomic Islands , Animals
11.
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
12.
J Appl Microbiol ; 135(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39165105

ABSTRACT

AIMS: Characterize global genomic features of 86 genomes of Salmonella Gallinarum (SG) and Pullorum (SP), which are important pathogens causing systemic infections in poultry. METHODS AND RESULTS: All genomes harbored efflux pump encoding gene mdsA and gold tolerance genes golS and golT. Aminoglycoside (aac(6')-Ib, aadA5, aph(6)-Id, aph(3'')-Ib, ant(2'')-Ia), beta-lactam (blaTEM-1, blaTEM-135), efflux pump (mdsB), fosfomycin (fosA3), sulfonamide (sul1, sul2), tetracycline [tet(A)], trimethoprim (dfrA17), acid (asr), and disinfectant (qacEdelta1) resistance genes, gyrA, gyrB, and parC quinolone resistance point mutations, and mercury tolerance genes (mer) were found in different frequencies. Additionally, 310 virulence genes, pathogenicity islands (including SPI-1, 2, 3, 4, 5, 6, 9, 10, 12, 13, and 14), plasmids [IncFII(S), ColpVC, IncX1, IncN, IncX2, and IncC], and prophages (Fels-2, ST104, 500465-1, pro483, Gifsy-2, 103 203_sal5, Fels-1, RE-2010, vB_SenS-Ent2, and L-413C) were detected. MLST showed biovar-specific sequence types, and core genome MLST showed country-specific and global-related clusters. CONCLUSION: SG and SP global strains carry many virulence factors and important antimicrobial resistance genes. The diverse plasmids and prophages suggest genetic variability. MLST and cgMLST differentiated biovars and showed profiles occurring locally or worldwide.


Subject(s)
Genome, Bacterial , Poultry Diseases , Salmonella enterica , Serogroup , Salmonella enterica/genetics , Salmonella enterica/drug effects , Animals , Poultry Diseases/microbiology , Anti-Bacterial Agents/pharmacology , Genomic Islands/genetics , Salmonella Infections, Animal/microbiology , Drug Resistance, Multiple, Bacterial/genetics , Virulence Factors/genetics , Plasmids/genetics , Chickens/microbiology , Genomics , Microbial Sensitivity Tests , Drug Resistance, Bacterial/genetics
13.
World J Microbiol Biotechnol ; 40(10): 289, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39102038

ABSTRACT

This study presents the empirical findings of an in-depth genomic analysis of Enterococcus faecalis and Enterococcus lactis isolates from South Africa. It offers valuable insights into their genetic characteristics and their significant implications for public health. The study uncovers nuanced variations in the gene content of these isolates, despite their similar GC contents, providing a comprehensive view of the evolutionary diversity within the species. Genomic islands are identified, particularly in E. faecalis, emphasizing its propensity for horizontal gene transfer and genetic diversity, especially in terms of antibiotic resistance genes. Pangenome analysis reveals the existence of a core genome, accounting for a modest proportion of the total genes, with 2157 core genes, 1164 shell genes, and 4638 cloud genes out of 7959 genes in 52 South African E. faecalis genomes (2 from this study, 49 south Africa genomes downloaded from NCBI, and E. faecalis reference genome). Detecting large-scale genomic rearrangements, including chromosomal inversions, underscores the dynamic nature of bacterial genomes and their role in generating genetic diversity. The study uncovers an array of antibiotic resistance genes, with trimethoprim, tetracycline, glycopeptide, and multidrug resistance genes prevalent, raising concerns about the effectiveness of antibiotic treatment. Virulence gene profiling unveils a diverse repertoire of factors contributing to pathogenicity, encompassing adhesion, biofilm formation, stress resistance, and tissue damage. These empirical findings provide indispensable insights into these bacteria's genomic dynamics, antibiotic resistance mechanisms, and virulence potential, underlining the pressing need to address antibiotic resistance and implement robust control measures.


Subject(s)
Anti-Bacterial Agents , Enterococcus faecalis , Genetic Variation , Genome, Bacterial , Virulence Factors , South Africa , Enterococcus faecalis/genetics , Enterococcus faecalis/drug effects , Enterococcus faecalis/pathogenicity , Enterococcus faecalis/isolation & purification , Virulence/genetics , Anti-Bacterial Agents/pharmacology , Virulence Factors/genetics , Humans , Drug Resistance, Bacterial/genetics , Genomic Islands/genetics , Gram-Positive Bacterial Infections/microbiology , Enterococcus/genetics , Enterococcus/drug effects , Enterococcus/pathogenicity , Enterococcus/isolation & purification , Enterococcus/classification , Phylogeny , Gene Transfer, Horizontal , Genomics , Microbial Sensitivity Tests
14.
Sci Rep ; 14(1): 19804, 2024 08 27.
Article in English | MEDLINE | ID: mdl-39191804

ABSTRACT

Fusobacterium necrophorum is a Gram-negative anaerobic bacterium responsible for localized infections of the oropharynx that can evolve into bacteremia and/or septic thrombophlebitis of the jugular vein or peritonsillar vein, called Lemierre's syndrome. To identify microbial genetic determinants associated with the severity of this life-threatening disease, 70 F. necrophorum strains were collected and grouped into two categories according to the clinical presentation: (i) localized infection, (ii) bacteremia with/without Lemierre's syndrome. Comparative genomic analyses revealed two clades with distinct genetic content, one clade being significantly enriched with isolates from subjects with bacteremia. To identify genetic determinants contributing to F. necrophorum pathogenicity, genomic islands and virulence factor orthogroups (OVFs) were predicted. The presence/absence profiles of OVFs did not group isolates according to their clinical category, but rather according to their phylogeny. However, a variant of lktA, a key virulence factor, with a frameshift deletion that results in two open reading frames, was associated with bacteremia. Moreover, a genome-wide association study identified three orthogroups associated with bacteremic strains: (i) cas8a1, (ii) a sodium/solute symporter, and (iii) a POP1 domain-containing protein. Further studies must be performed to assess the functional impact of lktA mutation and of these orthogroups on the physiopathological mechanisms of F. necrophorum infections.


Subject(s)
Bacteremia , Fusobacterium necrophorum , Lemierre Syndrome , Virulence Factors , Fusobacterium necrophorum/genetics , Fusobacterium necrophorum/isolation & purification , Humans , Lemierre Syndrome/microbiology , Bacteremia/microbiology , Virulence Factors/genetics , Male , Female , Phylogeny , Adult , Genome-Wide Association Study , Middle Aged , Bacterial Proteins/genetics , Fusobacterium Infections/microbiology , Fusobacterium Infections/complications , Aged , Genomic Islands/genetics , Hemolysin Proteins
15.
J Bacteriol ; 206(9): e0014524, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39133004

ABSTRACT

In response to predation by bacteriophages and invasion by other mobile genetic elements such as plasmids, bacteria have evolved specialized defense systems that are often clustered together on genomic islands. The O1 El Tor strains of Vibrio cholerae responsible for the ongoing seventh cholera pandemic (7PET) contain a characteristic set of genomic islands involved in host colonization and disease, many of which contain defense systems. Notably, Vibrio pathogenicity island 2 contains several characterized defense systems as well as a putative type I restriction-modification (T1RM) system, which, interestingly, is interrupted by two genes of unknown function. Here, we demonstrate that the T1RM system is active, methylates the host genomes of a representative set of 7PET strains, and identify a specific recognition sequence that targets non-methylated plasmids for restriction. We go on to show that the two genes embedded within the T1RM system encode a novel two-protein modification-dependent restriction system related to the GmrSD family of type IV restriction enzymes. Indeed, we show that this system has potent anti-phage activity against diverse members of the Tevenvirinae, a subfamily of bacteriophages with hypermodified genomes. Taken together, these results expand our understanding of how this highly conserved genomic island contributes to the defense of pandemic V. cholerae against foreign DNA. IMPORTANCE: Defense systems are immunity systems that allow bacteria to counter the threat posed by bacteriophages and other mobile genetic elements. Although these systems are numerous and highly diverse, the most common types are restriction enzymes that can specifically recognize and degrade non-self DNA. Here, we show that the Vibrio pathogenicity island 2, present in the pathogen Vibrio cholerae, encodes two types of restriction systems that use distinct mechanisms to sense non-self DNA. The first system is a classical Type I restriction-modification system, and the second is a novel modification-dependent type IV restriction system that recognizes hypermodified cytosines. Interestingly, these systems are embedded within each other, suggesting that they are complementary to each other by targeting both modified and non-modified phages.


Subject(s)
Genomic Islands , Vibrio cholerae , Vibrio cholerae/genetics , Vibrio cholerae/virology , Plasmids/genetics , Bacteriophages/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cholera/microbiology , DNA Methylation , DNA Restriction-Modification Enzymes/genetics , DNA Restriction-Modification Enzymes/metabolism
16.
PLoS Biol ; 22(8): e3002796, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39208359

ABSTRACT

Mobile genetic elements shape microbial gene repertoires and populations. Recent results reveal that many, possibly most, microbial mobile genetic elements require helpers to transfer between genomes, which we refer to as Hitcher Genetic Elements (hitchers or HGEs). They may be a large fraction of pathogenicity and resistance genomic islands, whose mechanisms of transfer have remained enigmatic for decades. Together with their helper elements and their bacterial hosts, hitchers form tripartite networks of interactions that evolve rapidly within a parasitism-mutualism continuum. In this emerging view of microbial genomes as communities of mobile genetic elements many questions arise. Which elements are being moved, by whom, and how? How often are hitchers costly hyper-parasites or beneficial mutualists? What is the evolutionary origin of hitchers? Are there key advantages associated with hitchers' lifestyle that justify their unexpected abundance? And why are hitchers systematically smaller than their helpers? In this essay, we start answering these questions and point ways ahead for understanding the principles, origin, mechanisms, and impact of hitchers in bacterial ecology and evolution.


Subject(s)
Gene Transfer, Horizontal , Interspersed Repetitive Sequences/genetics , Bacteria/genetics , Evolution, Molecular , Genomic Islands , Symbiosis/genetics , Genome, Bacterial
17.
Sci Rep ; 14(1): 15216, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956138

ABSTRACT

Here, we present the whole genome sequence of Bt S2160-1, a potential alternative to the mosquitocidal model strain, Bti. One chromosome genome and four mega-plasmids were contained in Bt S2160-1, and 13 predicted genes encoding predicted insecticidal crystal proteins were identified clustered on one plasmid pS2160-1p2 containing two pathogenic islands (PAIs) designed as PAI-1 (Cry54Ba, Cry30Ea4, Cry69Aa-like, Cry50Ba2-like, Cry4Ca1-like, Cry30Ga2, Cry71Aa-like, Cry72Aa-like, Cry70Aa-like, Cyt1Da2-like and Vpb4C1-like) and PAI-2 (Cyt1Aa-like, and Tpp80Aa1-like). The clusters appear to represent mosquitocidal toxin islands similar to pathogenicity islands. Transcription/translation of 10 of the 13 predicted genes was confirmed by whole-proteome analysis using LTQ-Orbitrap LC-MS/MS. In summary, the present study identified the existence of a mosquitocidal toxin island in Bacillus thuringiensis, and provides important genomic information for understanding the insecticidal mechanism of B. thuringiensis.


Subject(s)
Bacillus thuringiensis , Bacterial Proteins , Insecticides , Proteomics , Bacillus thuringiensis/genetics , Bacillus thuringiensis/metabolism , Proteomics/methods , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Insecticides/pharmacology , Whole Genome Sequencing/methods , Genome, Bacterial , Endotoxins/genetics , Bacillus thuringiensis Toxins , Genomic Islands , Proteome , Plasmids/genetics , Tandem Mass Spectrometry , Animals , Hemolysin Proteins/genetics
18.
BMC Genomics ; 25(1): 724, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39060998

ABSTRACT

BACKGROUND: The obligate intracellular bacterial family Chlamydiaceae comprises a number of different species that cause disease in various vertebrate hosts including humans. Chlamydia suis, primarily found in the gastrointestinal tract of pigs, is the only species of the Chlamydiaceae family to have naturally gained tetracycline resistance (TetR), through a genomic island (Tet-island), integrated into the middle of chromosomal invasin-like gene inv. Previous studies have hypothesised that the uptake of the Tet-island from a host outside the Chlamydiaceae family was a unique event, followed by spread among C. suis through homologous recombination. In vitro recombination studies have shown that Tet-island exchange between C. suis strains is possible. Our aim in this study was to gain a deeper understanding of the interclade recombination of the Tet-island, among currently circulating C. suis field strains compared to in vitro-generated recombinants, using published whole genome sequences of C. suis field strains (n = 35) and in vitro-generated recombinants (n = 63). RESULTS: We found that the phylogeny of inv better reflected the phylogeny of the Tet-island than that of the whole genome, supporting recombination rather than site-specific insertion as the means of transfer. There were considerable differences between the distribution of recombinations within in vitro-generated strains compared to that within the field strains. These differences are likely because in vitro-generated recombinants were selected for a tetracycline and rifamycin/rifampicin resistant background, leading to the largest peak of recombination across the Tet-island. Finally, we found that interclade recombinations across the Tet-island were more variable in length downstream of the Tet-island than upstream. CONCLUSIONS: Our study supports the hypothesis that the occurrence of TetR strains in both clades of C. suis came about through interclade recombination after a single ancestral horizontal gene transfer event.


Subject(s)
Chlamydia , Genomic Islands , Phylogeny , Recombination, Genetic , Tetracycline Resistance , Chlamydia/genetics , Tetracycline Resistance/genetics , Animals , Swine , Gene Transfer, Horizontal , Genome, Bacterial
19.
Environ Microbiol ; 26(8): e16684, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39080854

ABSTRACT

The knowledge of the different population-level processes operating within a species, and the genetic variability of the individual prokaryotic genomes, is key to understanding the adaptability of microbial populations. Here, we characterized the flexible genome of ammonia-oxidizing archaeal (AOA) populations using a metagenomic recruitment approach and long-read (PacBio HiFi) metagenomic sequencing. In the lower photic zone of the western Mediterranean Sea (75 m deep), the genomes Nitrosopelagicus brevis CN25 and Nitrosopumilus catalinensis SPOT1 had the highest recruitment values among available complete AOA genomes. They were used to analyse the diversity of flexible genes (variable from strain to strain) by examining the long-reads located within the flexible genomic islands (fGIs) identified by their under-recruitment. Both AOA genomes had a large fGI involved in the glycosylation of exposed structures, highly variable, and rich in glycosyltransferases. N. brevis had two fGIs related to the transport of phosphorus and ammonium respectively. N. catalinensis had fGIs involved in phosphorus transportation and metal uptake. A fGI5 previously reported as 'unassigned function' in N. brevis could be associated with defense. These findings demonstrate that the microdiversity of marine microbe populations, including AOA, can be effectively characterized using an approach that incorporates third-generation sequencing metagenomics.


Subject(s)
Ammonia , Archaea , Genome, Archaeal , Metagenome , Oxidation-Reduction , Seawater , Mediterranean Sea , Archaea/genetics , Archaea/metabolism , Archaea/classification , Ammonia/metabolism , Seawater/microbiology , Metagenomics , Phylogeny , Genetic Variation , Genomic Islands , Biodiversity
20.
Emerg Infect Dis ; 30(8): 1729-1732, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39043427

ABSTRACT

Vibrio mimicus bacteria have caused sporadic cases and outbreaks of cholera-like diarrhea throughout the world, but the association of lineages with such events is unexplored. Genomic analyses revealed V. mimicus lineages carrying the virulence factors cholera toxin and toxin coregulated pilus, one of which has persisted for decades in China and the United States.


Subject(s)
Cholera Toxin , Genomic Islands , Vibrio mimicus , China/epidemiology , Humans , Vibrio mimicus/genetics , Vibrio mimicus/pathogenicity , United States/epidemiology , Cholera Toxin/genetics , Cholera/microbiology , Cholera/epidemiology , Phylogeny , Vibrio Infections/microbiology , Vibrio Infections/epidemiology , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL