Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Int Wound J ; 21(1): e14626, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38272816

ABSTRACT

Current microbiome investigations of patients with pressure ulcers (PU) are mainly based on wound swabs and/or biopsy sequencing, leaving the colonization scenario unclear. Urinary microbiota has been never studied. As a part of the prospective ESCAFLOR study, we studied urinary microbiota of spinal cord injury (SCI) patients with PU without any urinary tract infection at the inclusion, collected at two times (at admission [D0] and after 28 days [D28]) during the patient's care, investigated by 16S rDNA metagenomics next generation sequencing. Subgroup analyses were carried out between patients with wounds showing improved evolution versus stagnated/worsened wounds at D28. Analysis was done using EPISEQ® 16S and R software. Among the 12 studied patients, the urinary microbiota of patients with improved wound evolution at D28 (n = 6) presented a significant decrease of microbial diversity. This modification was associated with the presence of Proteobacteria phylum and an increase of Escherichia-Shigella (p = 0.005), as well as the presence of probiotic anaerobic bacteria Lactobacillus and Bifidobacterium. In contrast, Proteus abundance was significantly increased in urine of patients with stagnated/worsened wound evolution (n = 6) (p = 0.003). This study proposes urinary microbiota as a complementary factor indirectly associated with the wound evolution and patient cure. It opens new perspectives for further investigations based on multiple body microbiome comparison to describe the complete scenario of the transmission dynamics of wound-colonizing microorganisms.


Subject(s)
Microbiota , Pressure Ulcer , Spinal Cord Injuries , Humans , Pressure Ulcer/complications , Prospective Studies , Spinal Cord Injuries/complications
2.
Front Microbiol ; 13: 863777, 2022.
Article in English | MEDLINE | ID: mdl-35531285

ABSTRACT

Bone and joint infections (BJIs) are complex infections that require precise microbiological documentation to optimize antibiotic therapy. Currently, diagnosis is based on microbiological culture, sometimes complemented by amplification and sequencing of the 16S rDNA gene. Clinical metagenomics (CMg), that is, the sequencing of the entire nucleic acids in a sample, was previously shown to identify bacteria not detected by conventional methods, but its actual contribution to the diagnosis remains to be assessed, especially with regard to 16S rDNA sequencing. In the present study, we tested the performance of CMg in 34 patients (94 samples) with suspected BJIs, as compared to culture and 16S rDNA sequencing. A total of 94 samples from 34 patients with suspicion of BJIs, recruited from two sites, were analyzed by (i) conventional culture, (ii) 16S rDNA sequencing (Sanger method), and (iii) CMg (Illumina Technology). Two negative controls were also sequenced by CMg for contamination assessment. Based on the sequencing results of negative controls, 414 out of 539 (76.7%) bacterial species detected by CMg were considered as contaminants and 125 (23.2%) as truly present. For monomicrobial infections (13 patients), the sensitivity of CMg was 83.3% as compared to culture, and 100% as compared to 16S rDNA. For polymicrobial infections (13 patients), the sensitivity of CMg was 50% compared to culture, and 100% compared to 16S rDNA. For samples negative in culture (8 patients, 21 samples), CMg detected 11 bacteria in 10 samples from 5 different patients. In 5/34 patients, CMg brought a microbiological diagnosis where conventional methods failed, and in 16/34 patients, CMg provided additional information. Finally, 99 antibiotic resistance genes were detected in 24 patients (56 samples). Provided sufficient genome coverage (87.5%), a correct inference of antibiotic susceptibility was achieved in 8/8 bacteria (100%). In conclusion, our study demonstrated that the CMg provides complementary and potentially valuable data to conventional methods of BJIs diagnosis.

3.
Article in English | MEDLINE | ID: mdl-29914966

ABSTRACT

Multidrug-resistant Acinetobacter baumannii infection has recently emerged as a worldwide clinical problem, and colistin is increasingly being used as a last-resort therapy. Despite its favorable bacterial killing, resistance and heteroresistance (HR) to colistin have been described. The purpose of the present study was to investigate the role of the PmrAB regulatory pathway in laboratory-selected mutants representative of global epidemic strains. From three unrelated A. baumannii clinical strains (sequence types 2, 3, and 20), eight colistin-resistant mutants were selected. Half of the mutants showed HR to colistin according to the reference method (population analysis profiling), whereas the other half exhibited stable resistance. M12I mutation within pmrA and M308R, S144KLAGS, and P170L mutations for pmrB were associated with HR to colistin, while T235I, A226T, and P233S mutations within pmrB were associated with stable resistance. The transcript levels of the pmrCAB operon were upregulated in all the mutants. Compensatory mutations were explored for some mutants. A single mutant (T235I mutant) displayed a compensatory mutation through ISAba1 mobilization within the pmrB gene that was associated with the loss of colistin resistance. The mutant resistance phenotype associated with T235I was partially restored in a trans-complementation assay turning to HR. The level of colistin resistance was correlated with the level of expression of pmrC in the trans-complemented strains. This report shows the role of different mutations in the PmrAB regulatory pathway and warns of the development of colistin HR that could be present but not easily detected through routine testing.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Colistin/pharmacology , Drug Resistance, Bacterial/genetics , Transcription Factors/genetics , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Acinetobacter baumannii/genetics , Bacterial Proteins/metabolism , Base Sequence , Humans , Microbial Sensitivity Tests , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA , Transcription Factors/metabolism
4.
Front Microbiol ; 9: 511, 2018.
Article in English | MEDLINE | ID: mdl-29616014

ABSTRACT

The French National Reference Center for Staphylococci currently uses DNA arrays and spa typing for the initial epidemiological characterization of Staphylococcus aureus strains. We here describe the use of whole-genome sequencing (WGS) to investigate retrospectively four distinct and virulent S. aureus lineages [clonal complexes (CCs): CC1, CC5, CC8, CC30] involved in hospital and community outbreaks or sporadic infections in France. We used a WGS bioinformatics pipeline based on de novo assembly (reference-free approach), single nucleotide polymorphism analysis, and on the inclusion of epidemiological markers. We examined the phylogeographic diversity of the French dominant hospital-acquired CC8-MRSA (methicillin-resistant S. aureus) Lyon clone through WGS analysis which did not demonstrate evidence of large-scale geographic clustering. We analyzed sporadic cases along with two outbreaks of a CC1-MSSA (methicillin-susceptible S. aureus) clone containing the Panton-Valentine leukocidin (PVL) and results showed that two sporadic cases were closely related. We investigated an outbreak of PVL-positive CC30-MSSA in a school environment and were able to reconstruct the transmission history between eight families. We explored different outbreaks among newborns due to the CC5-MRSA Geraldine clone and we found evidence of an unsuspected link between two otherwise distinct outbreaks. Here, WGS provides the resolving power to disprove transmission events indicated by conventional methods (same sequence type, spa type, toxin profile, and antibiotic resistance profile) and, most importantly, WGS can reveal unsuspected transmission events. Therefore, WGS allows to better describe and understand outbreaks and (inter-)national dissemination of S. aureus lineages. Our findings underscore the importance of adding WGS for (inter-)national surveillance of infections caused by virulent clones of S. aureus but also substantiate the fact that technological optimization at the bioinformatics level is still urgently needed for routine use. However, the greatest limitation of WGS analysis is the completeness and the correctness of the reference database being used and the conversion of floods of data into actionable results. The WGS bioinformatics pipeline (EpiSeqTM) we used here can easily generate a uniform database and associated metadata for epidemiological applications.

5.
Int J Antimicrob Agents ; 50(2): 210-218, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28554735

ABSTRACT

Genetic determinants of antibiotic resistance (AR) have been extensively investigated. High-throughput sequencing allows for the assessment of the relationship between genotype and phenotype. A panel of 672 Pseudomonas aeruginosa strains was analysed, including representatives of globally disseminated multidrug-resistant and extensively drug-resistant clones; genomes and multiple antibiograms were available. This panel was annotated for AR gene presence and polymorphism, defining a resistome in which integrons were included. Integrons were present in >70 distinct cassettes, with In5 being the most prevalent. Some cassettes closely associated with clonal complexes, whereas others spread across the phylogenetic diversity, highlighting the importance of horizontal transfer. A resistome-wide association study (RWAS) was performed for clinically relevant antibiotics by correlating the variability in minimum inhibitory concentration (MIC) values with resistome data. Resistome annotation identified 147 loci associated with AR. These loci consisted mainly of acquired genomic elements and intrinsic genes. The RWAS allowed for correct identification of resistance mechanisms for meropenem, amikacin, levofloxacin and cefepime, and added 46 novel mutations. Among these, 29 were variants of the oprD gene associated with variation in meropenem MIC. Using genomic and MIC data, phenotypic AR was successfully correlated with molecular determinants at the whole-genome sequence level.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Genes, Bacterial , Genotype , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Genetic Loci , Humans , Interspersed Repetitive Sequences , Microbial Sensitivity Tests , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/isolation & purification
7.
Nat Microbiol ; 1: 16027, 2016 03 21.
Article in English | MEDLINE | ID: mdl-27572446

ABSTRACT

Together with plague, smallpox and typhus, epidemics of dysentery have been a major scourge of human populations for centuries(1). A previous genomic study concluded that Shigella dysenteriae type 1 (Sd1), the epidemic dysentery bacillus, emerged and spread worldwide after the First World War, with no clear pattern of transmission(2). This is not consistent with the massive cyclic dysentery epidemics reported in Europe during the eighteenth and nineteenth centuries(1,3,4) and the first isolation of Sd1 in Japan in 1897(5). Here, we report a whole-genome analysis of 331 Sd1 isolates from around the world, collected between 1915 and 2011, providing us with unprecedented insight into the historical spread of this pathogen. We show here that Sd1 has existed since at least the eighteenth century and that it swept the globe at the end of the nineteenth century, diversifying into distinct lineages associated with the First World War, Second World War and various conflicts or natural disasters across Africa, Asia and Central America. We also provide a unique historical perspective on the evolution of antibiotic resistance over a 100-year period, beginning decades before the antibiotic era, and identify a prevalent multiple antibiotic-resistant lineage in South Asia that was transmitted in several waves to Africa, where it caused severe outbreaks of disease.


Subject(s)
Dysentery, Bacillary/epidemiology , Dysentery, Bacillary/microbiology , Evolution, Molecular , Phylogeography , Serogroup , Shigella dysenteriae/classification , Shigella dysenteriae/isolation & purification , Drug Resistance, Bacterial , Dysentery, Bacillary/history , Genome, Bacterial , Global Health , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Molecular Epidemiology , Sequence Analysis, DNA , Shigella dysenteriae/genetics
8.
Future Microbiol ; 11(3): 427-39, 2016.
Article in English | MEDLINE | ID: mdl-26934540

ABSTRACT

The increasing burden of multidrug-resistant bacteria affects the management of several infections. In order to prescribe adequate antibiotics, clinicians facing severe infections such as hospital-acquired pneumonia (HAP) need to promptly identify the pathogens and know their antibiotic susceptibility profiles (AST), which with conventional microbiology currently requires 24 and 48 h, respectively. Clinical metagenomics, based on whole genome sequencing of clinical samples, could improve the diagnosis of HAP, however, many obstacles remain to be overcome, namely the turn-around time, the quantification of pathogens, the choice of antibiotic resistance determinants (ARDs), the inference of the AST from metagenomic data and the linkage between ARDs and their host. Here, we propose to tackle those issues in a bottom-up, clinically driven approach.


Subject(s)
Bacteria/genetics , Cross Infection/diagnosis , Disease Management , Metagenomics , Pneumonia, Ventilator-Associated/diagnosis , Anti-Bacterial Agents/therapeutic use , Bacteria/drug effects , Bacteria/isolation & purification , Computational Biology , Cross Infection/drug therapy , Cross Infection/microbiology , Drug Resistance, Multiple, Bacterial , Enterobacteriaceae/drug effects , Enterobacteriaceae/genetics , Enterobacteriaceae/isolation & purification , Genome, Bacterial , High-Throughput Nucleotide Sequencing , Humans , Pneumonia, Ventilator-Associated/drug therapy , Pneumonia, Ventilator-Associated/microbiology , Risk Factors
9.
PLoS One ; 9(1): e87194, 2014.
Article in English | MEDLINE | ID: mdl-24489870

ABSTRACT

The prediction of viral zoonosis epidemics has become a major public health issue. A profound understanding of the viral population in key animal species acting as reservoirs represents an important step towards this goal. Bats harbor diverse viruses, some of which are of particular interest because they cause severe human diseases. However, little is known about the diversity of the global population of viruses found in bats (virome). We determined the viral diversity of five different French insectivorous bat species (nine specimens in total) in close contact with humans. Sequence-independent amplification, high-throughput sequencing with Illumina technology and a dedicated bioinformatics analysis pipeline were used on pooled tissues (brain, liver and lungs). Comparisons of the sequences of contigs and unassembled reads provided a global taxonomic distribution of virus-related sequences for each sample, highlighting differences both within and between bat species. Many viral families were present in these viromes, including viruses known to infect bacteria, plants/fungi, insects or vertebrates, the most relevant being those infecting mammals (Retroviridae, Herpesviridae, Bunyaviridae, Poxviridae, Flaviviridae, Reoviridae, Bornaviridae, Picobirnaviridae). In particular, we detected several new mammalian viruses, including rotaviruses, gammaretroviruses, bornaviruses and bunyaviruses with the identification of the first bat nairovirus. These observations demonstrate that bats naturally harbor viruses from many different families, most of which infect mammals. They may therefore constitute a major reservoir of viral diversity that should be analyzed carefully, to determine the role played by bats in the spread of zoonotic viral infections.


Subject(s)
Bornaviridae/genetics , Chiroptera/virology , Gammaretrovirus/genetics , Nairovirus/genetics , Rotavirus/genetics , Animals , Bornaviridae/classification , Bornaviridae/isolation & purification , Female , France , Gammaretrovirus/classification , Gammaretrovirus/isolation & purification , High-Throughput Nucleotide Sequencing , Humans , Male , Metagenome , Molecular Sequence Data , Nairovirus/classification , Nairovirus/isolation & purification , Phylogeny , RNA, Viral/genetics , Rotavirus/classification , Rotavirus/isolation & purification , Sequence Analysis, RNA
10.
PLoS One ; 8(6): e67510, 2013.
Article in English | MEDLINE | ID: mdl-23818983

ABSTRACT

BACKGROUND: Clostridium botulinum and related clostridia express extremely potent toxins known as botulinum neurotoxins (BoNTs) that cause severe, potentially lethal intoxications in humans. These BoNT-producing bacteria are categorized in seven major toxinotypes (A through G) and several subtypes. The high diversity in nucleotide sequence and genetic organization of the gene cluster encoding the BoNT components poses a great challenge for the screening and characterization of BoNT-producing strains. METHODOLOGY/PRINCIPAL FINDINGS: In the present study, we designed and evaluated the performances of a resequencing microarray (RMA), the PathogenId v2.0, combined with an automated data approach for the simultaneous detection and characterization of BoNT-producing clostridia. The unique design of the PathogenID v2.0 array allows the simultaneous detection and characterization of 48 sequences targeting the BoNT gene cluster components. This approach allowed successful identification and typing of representative strains of the different toxinotypes and subtypes, as well as the neurotoxin-producing C. botulinum strain in a naturally contaminated food sample. Moreover, the method allowed fine characterization of the different neurotoxin gene cluster components of all studied strains, including genomic regions exhibiting up to 24.65% divergence with the sequences tiled on the arrays. CONCLUSIONS/SIGNIFICANCE: The severity of the disease demands rapid and accurate means for performing risk assessments of BoNT-producing clostridia and for tracing potentials sources of contamination in outbreak situations. The RMA approach constitutes an essential higher echelon component in a diagnostics and surveillance pipeline. In addition, it is an important asset to characterise potential outbreak related strains, but also environment isolates, in order to obtain a better picture of the molecular epidemiology of BoNT-producing clostridia.


Subject(s)
Botulinum Toxins/metabolism , Clostridium botulinum/genetics , Oligonucleotide Array Sequence Analysis/methods , Sequence Analysis, DNA/methods , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Typing Techniques/methods , Botulinum Toxins/classification , Botulism/diagnosis , Botulism/microbiology , Clostridium botulinum/classification , Clostridium botulinum/metabolism , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Food Contamination/analysis , Food Contamination/prevention & control , Food Microbiology/methods , Humans , Multigene Family/genetics , Reproducibility of Results , Sensitivity and Specificity
11.
PLoS One ; 7(5): e36995, 2012.
Article in English | MEDLINE | ID: mdl-22623967

ABSTRACT

Laboratory surveillance systems for salmonellosis should ideally be based on the rapid serotyping and subtyping of isolates. However, current typing methods are limited in both speed and precision. Using 783 strains and isolates belonging to 130 serotypes, we show here that a new family of DNA repeats named CRISPR (clustered regularly interspaced short palindromic repeats) is highly polymorphic in Salmonella. We found that CRISPR polymorphism was strongly correlated with both serotype and multilocus sequence type. Furthermore, spacer microevolution discriminated between subtypes within prevalent serotypes, making it possible to carry out typing and subtyping in a single step. We developed a high-throughput subtyping assay for the most prevalent serotype, Typhimurium. An open web-accessible database was set up, providing a serotype/spacer dictionary and an international tool for strain tracking based on this innovative, powerful typing and subtyping tool.


Subject(s)
Inverted Repeat Sequences/genetics , Polymorphism, Genetic/genetics , Population Surveillance/methods , Salmonella Infections/diagnosis , Salmonella typhimurium/genetics , Serotyping/methods , Databases, Genetic , Humans , Internet , Salmonella Infections/genetics
12.
J Clin Microbiol ; 50(3): 696-701, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22205819

ABSTRACT

Endemic strains of Legionella pneumophila sequence type 1 (ST1), in particular the ST1/Paris pulsotype, are dispersed worldwide and represent about 10% of culture-proven clinical cases of Legionnaires' disease in France. The high rate of isolation of this strain from both clinical and environmental samples makes identification of the source of infection difficult during epidemiological investigations. The full-length genome sequence of this strain was recently determined, and it revealed the presence of a CRISPR/cas complex. The aim of this study was to develop and evaluate a spoligotyping tool based on the diversity of this CRISPR locus that would allow the accurate subtyping of the L. pneumophila serogroup 1 ST1/Paris pulsotype. The CRISPR loci of 28 L. pneumophila ST1/Paris pulsotype isolates were sequenced, and 42 different spacers regions were characterized. A membrane-based spoligotyping method was developed and used to determine the subtypes of 406 L. pneumophila isolates, including 233 with the ST1/Paris pulsotype profile that were collected in France from 2000 to 2011. A total of 46 different spoligotypes were detected, and 41 of these were specifically identified in the ST1/Paris pulsotype isolates. In 27 of 33 epidemiological investigations, the environmental source of contamination was confirmed by comparing spoligotypes of clinical isolates with those of environmental isolates. With an index of discrimination of 79.72% (95% confidence interval, 75.82 to 83.63), spoligotyping of the L. pneumophila ST1/Paris pulsotype has the potential to be a useful complementary genotyping tool for discriminating isolates with undistinguishable pulsed-field gel electrophoresis (PFGE) and ST genotypes, which could help to identify environmental sources of infection.


Subject(s)
Legionella pneumophila/classification , Legionella pneumophila/genetics , Legionnaires' Disease/epidemiology , Legionnaires' Disease/microbiology , Molecular Typing/methods , Cluster Analysis , DNA Fingerprinting/methods , Electrophoresis, Gel, Pulsed-Field/methods , France/epidemiology , Genotype , Humans , Molecular Epidemiology/methods
13.
J Clin Microbiol ; 49(9): 3268-75, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21715589

ABSTRACT

High-throughput sequencing furnishes a large number of short sequence reads from uncloned DNA and has rapidly become a major tool for identifying viruses in biological samples, and in particular when the target sequence is undefined. In this study, we assessed the analytical sensitivity of a pipeline for detection of viruses in biological samples based on either the Roche-454 genome sequencer or Illumina genome analyzer platforms. We sequenced biological samples artificially spiked with a wide range of viruses with genomes composed of single or double-stranded DNA or RNA, including linear or circular single-stranded DNA. Viruses were added at a very low concentration most often corresponding to 3 or 0.8 times the validated level of detection of quantitative reverse transcriptase PCRs (RT-PCRs). For the viruses represented, or resembling those represented, in public nucleotide sequence databases, we show that the higher output of Illumina is associated with a much greater sensitivity, approaching that of optimized quantitative (RT-)PCRs. In this blind study, identification of viruses was achieved without incorrect identification. Nevertheless, at these low concentrations, the number of reads generated by the Illumina platform was too small to facilitate assembly of contigs without the use of a reference sequence, thus precluding detection of unknown viruses. When the virus load was sufficiently high, de novo assembly permitted the generation of long contigs corresponding to nearly full-length genomes and thus should facilitate the identification of novel viruses.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Virology/methods , Viruses/classification , Viruses/isolation & purification , DNA, Viral/genetics , Humans , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity , Viruses/genetics
14.
J Virol ; 84(18): 9557-74, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20610710

ABSTRACT

The rapid and accurate identification of pathogens is critical in the control of infectious disease. To this end, we analyzed the capacity for viral detection and identification of a newly described high-density resequencing microarray (RMA), termed PathogenID, which was designed for multiple pathogen detection using database similarity searching. We focused on one of the largest and most diverse viral families described to date, the family Rhabdoviridae. We demonstrate that this approach has the potential to identify both known and related viruses for which precise sequence information is unavailable. In particular, we demonstrate that a strategy based on consensus sequence determination for analysis of RMA output data enabled successful detection of viruses exhibiting up to 26% nucleotide divergence with the closest sequence tiled on the array. Using clinical specimens obtained from rabid patients and animals, this method also shows a high species level concordance with standard reference assays, indicating that it is amenable for the development of diagnostic assays. Finally, 12 animal rhabdoviruses which were currently unclassified, unassigned, or assigned as tentative species within the family Rhabdoviridae were successfully detected. These new data allowed an unprecedented phylogenetic analysis of 106 rhabdoviruses and further suggest that the principles and methodology developed here may be used for the broad-spectrum surveillance and the broader-scale investigation of biodiversity in the viral world.


Subject(s)
RNA, Viral/genetics , Rhabdoviridae Infections/diagnosis , Rhabdoviridae Infections/veterinary , Rhabdoviridae/classification , Rhabdoviridae/genetics , Sequence Analysis, DNA/methods , Virology/methods , Animals , Cluster Analysis , Genotype , Humans , Molecular Sequence Data , Phylogeny , Rhabdoviridae Infections/virology , Sensitivity and Specificity
15.
Antimicrob Agents Chemother ; 54(1): 333-40, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19884373

ABSTRACT

An oligonucleotide-based DNA microarray was developed to evaluate expression of genes for efflux pumps in Acinetobacter baumannii and to detect acquired antibiotic resistance determinants. The microarray contained probes for 205 genes, including those for 47 efflux systems, 55 resistance determinants, and 35 housekeeping genes. The microarray was validated by comparative analysis of mutants overexpressing or deficient in the pumps relative to the parental strain. The performance of the microarray was also evaluated using in vitro single-step mutants obtained on various antibiotics. Overexpression, confirmed by quantitative reverse transcriptase PCR, of RND efflux pumps AdeABC, due to a G30D substitution in AdeS in a multidrug-resistant (MDR) strain obtained on gentamicin, and AdeIJK, in two mutants obtained on cefotaxime or tetracycline, was detected. A new efflux pump, AdeFGH, was found to be overexpressed in a mutant obtained on chloramphenicol. Study of MDR clinical isolates, including the AYE strain, whose entire sequence has been determined, indicated overexpression of AdeABC and of the chromosomally encoded cephalosporinase as well as the presence of several acquired resistance genes. The overexpressed and acquired determinants detected by the microarray could account for nearly the entire MDR phenotype of the isolates. The microarray is potentially useful for detection of resistance in A. baumannii and should allow detection of new efflux systems associated with antibiotic resistance.


Subject(s)
Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Oligonucleotide Array Sequence Analysis , Acinetobacter baumannii/metabolism , Anti-Bacterial Agents/metabolism , DNA, Bacterial/genetics , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Drug Resistance, Multiple, Bacterial/genetics , Genes, MDR/genetics , Metals, Heavy/pharmacology , Microbial Sensitivity Tests , Mutation/genetics , RNA, Bacterial/genetics , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
16.
BMC Genomics ; 9: 560, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-19036134

ABSTRACT

BACKGROUND: Extraintestinal pathogenic Escherichia coli (ExPEC) strains represent a huge public health burden. Knowledge of their clonal diversity and of the association of clones with genomic content and clinical features is a prerequisite to recognize strains with a high invasive potential. In order to provide an unbiased view of the diversity of E. coli strains responsible for bacteremia, we studied 161 consecutive isolates from patients with positive blood culture obtained during one year in two French university hospitals. We collected precise clinical information, multilocus sequence typing (MLST) data and virulence gene content for all isolates. A subset representative of the clonal diversity was subjected to comparative genomic hybridization (CGH) using 2,324 amplicons from the flexible gene pool of E. coli. RESULTS: Recombination-insensitive phylogenetic analysis of MLST data in combination with the ECOR collection revealed that bacteremic E. coli isolates were highly diverse and distributed into five major lineages, corresponding to the classical E. coli phylogroups (A+B1, B2, D and E) and group F, which comprises strains previously assigned to D. Compared to other strains of phylogenetic group B2, strains belonging to MLST-derived clonal complexes (CCs) CC1 and CC4 were associated (P < 0.05) with a urinary origin. In contrast, no CC appeared associated with severe sepsis or unfavorable outcome of the bacteremia. CGH analysis revealed genomic characteristics of the distinct CCs and identified genomic regions associated with CC1 and/or CC4. CONCLUSION: Our results demonstrate that human bacteremia strains distribute over the entire span of E. coli phylogenetic diversity and that CCs represent important phylogenetic units for pathogenesis and comparative genomics.


Subject(s)
Escherichia coli/classification , Escherichia coli/genetics , Phylogeny , Bacteremia/microbiology , Bacterial Typing Techniques , Cluster Analysis , Comparative Genomic Hybridization , DNA, Bacterial/genetics , Escherichia coli Infections/microbiology , Genome, Bacterial , Genotype , Humans , Polymorphism, Genetic
17.
Microbes Infect ; 10(14-15): 1582-6, 2008.
Article in English | MEDLINE | ID: mdl-18977453

ABSTRACT

The aim of this study was to analyze Bordetella pertussis isolates circulating, between 1991 and 1995, in Niakhar, Senegal area where the pertussis vaccination coverage was low and to compare them with those circulating in France, before and after introduction of generalized pertussis vaccination for toddlers in 1959. We carried out bacteriological analyses, typing, genotyping and DNA microarray analyses. We found that the isolates circulating in Senegal between 1991 and 1995 are part of the same Pulsed Field Gel Electrophoresis Group, express the same antigens and possess the same gene deletions than isolates circulating in France before the introduction of vaccination, but are different from those circulating in 1991-1995. We confirmed the influence of pertussis vaccination on circulating isolates and that isolates vary with the pertussis cycle.


Subject(s)
Bordetella pertussis/classification , Bordetella pertussis/genetics , Genome, Bacterial , Whooping Cough/microbiology , Antigens, Bacterial/genetics , Bacterial Typing Techniques , Bordetella pertussis/isolation & purification , DNA Fingerprinting , DNA, Bacterial/genetics , Electrophoresis, Gel, Pulsed-Field , France , Genotype , Humans , Microarray Analysis , Pertussis Vaccine/administration & dosage , Senegal , Sequence Deletion
18.
BMC Genomics ; 9: 388, 2008 Aug 18.
Article in English | MEDLINE | ID: mdl-18706115

ABSTRACT

BACKGROUND: Translation of the genome sequence of Plasmodium sp. into biologically relevant information relies on high through-put genomics technology which includes transcriptome analysis. However, few studies to date have used this powerful approach to explore transcriptome alterations of P. falciparum parasites exposed to antimalarial drugs. RESULTS: The rapid action of artesunate allowed us to study dynamic changes of the parasite transcriptome in synchronous parasite cultures exposed to the drug for 90 minutes and 3 hours. Developmentally regulated genes were filtered out, leaving 398 genes which presented altered transcript levels reflecting drug-exposure. Few genes related to metabolic pathways, most encoded chaperones, transporters, kinases, Zn-finger proteins, transcription activating proteins, proteins involved in proteasome degradation, in oxidative stress and in cell cycle regulation. A positive bias was observed for over-expressed genes presenting a subtelomeric location, allelic polymorphism and encoding proteins with potential export sequences, which often belonged to subtelomeric multi-gene families. This pointed to the mobilization of processes shaping the interface between the parasite and its environment. In parallel, pathways were engaged which could lead to parasite death, such as interference with purine/pyrimidine metabolism, the mitochondrial electron transport chain, proteasome-dependent protein degradation or the integrity of the food vacuole. CONCLUSION: The high proportion of over-expressed genes encoding proteins exported from the parasite highlight the importance of extra-parasitic compartments as fields for exploration in drug research which, to date, has mostly focused on the parasite itself rather than on its intra and extra erythrocytic environment. Further work is needed to clarify which transcriptome alterations observed reflect a specific response to overcome artesunate toxicity or more general perturbations on the path to cellular death.


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Analysis of Variance , Animals , Artesunate , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genes, Protozoan/drug effects , Life Cycle Stages , Oligonucleotide Array Sequence Analysis , Plasmodium falciparum/growth & development , RNA, Protozoan/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
19.
PLoS One ; 3(6): e2437, 2008 Jun 18.
Article in English | MEDLINE | ID: mdl-18560590

ABSTRACT

BACKGROUND: The objective of the study was to analyse the evolution of Bordetella pertussis population and the influence of herd immunity in different areas of the world where newborns and infants are highly vaccinated. METHODOLOGY: The analysis was performed using DNA microarray on 15 isolates, PCR on 111 isolates as well as GS-FLX sequencing technology on 3 isolates and the B. pertussis reference strain, Tohama I. PRINCIPAL FINDINGS: Our analyses demonstrate that the current circulating isolates are continuing to lose genetic material as compared to isolates circulating during the pre-vaccine era whatever the area of the world considered. The lost genetic material does not seem to be important for virulence. Our study confirms that the use of whole cell vaccines has led to the control of isolates that were similar to vaccine strains. GS-FLX sequencing technology shows that current isolates did not acquire any additional material when compared with vaccine strains or with isolates of the pre-vaccine era and that the sequenced strain Tohama I is not representative of the isolates. Furthermore, this technology allowed us to observe that the number of Insertion Sequence elements contained in the genome of the isolates is temporally increasing or varying between isolates. CONCLUSIONS: B. pertussis adaptation to humans is still in progress by losing genetic material via Insertion Sequence elements. Furthermore, recent isolates did not acquire any additional material when compared with vaccine strains or with isolates of the pre-vaccine era. Herd immunity, following intensive vaccination of infants and children with whole cell vaccines, has controlled isolates similar to the vaccine strains without modifying significantly the virulence of the isolates. With the replacement of whole cell vaccines by subunit vaccines, containing only few bacterial antigens targeting the virulence of the bacterium, one could hypothesize the circulation of isolates expressing less or modified vaccine antigens.


Subject(s)
Bordetella pertussis/genetics , Bordetella pertussis/immunology , Genome, Bacterial , Pertussis Vaccine/administration & dosage , Humans , Immunization Programs , Infant , Infant, Newborn , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
20.
Cell Microbiol ; 10(8): 1676-86, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18419774

ABSTRACT

Entamoeba histolytica is the protozoan parasite responsible for human amoebiasis. During invasive amoebiasis, migration is an essential process and it has previously been shown that the pro-inflammatory compound tumour necrosis factor (TNF) is produced and that it has a migratory effect on E. histolytica. This paper focuses on the analysis of parasite signalling and cytoskeleton changes leading to directional motility. TNF-induced signalling was PI3K-dependent and could lead to modifications in the polarization of certain cytoskeleton-related proteins. To analyse the effect of TNF signalling on gene expression, we used microarray analysis to screen for genes encoding proteins that were potentially important during chemotaxis towards TNF. Interestingly, we found that elements of the galactose/N-acetylgalactosamine lectin (Gal/GalNAc lectin) were upregulated during chemotaxis as well as genes encoding proteins involved in cytoskeleton dynamics. The alpha-actinin protein appeared to be an important candidate to link the Gal/GalNAc lectin to the cytoskeleton during chemotaxis signalling. Dominant negative parasites blocked for Gal/GalNAc lectin signalling were no longer able to chemotax towards TNF. These results have given us an insight on how E. histolytica changes its cytoskeleton dynamics during chemotaxis and revealed the capital role of PI3K and Gal/GalNAc lectin signalling in chemotaxis.


Subject(s)
Chemotaxis , Entamoeba histolytica/cytology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Tumor Necrosis Factors/immunology , Acetylgalactosamine/metabolism , Androstadienes/pharmacology , Animals , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Entamoeba histolytica/immunology , Entamoeba histolytica/metabolism , Galactose/metabolism , Gene Expression , Humans , Lectins/immunology , Lectins/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protozoan Proteins/analysis , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...