78 results on '"Christophe Ginevra"'
Search Results
2. TNF-α response in macrophages depends on clinical Legionella pneumophila isolates genotypes
- Author
-
Johann Guillemot, Christophe Ginevra, Camille Allam, Elisabeth Kay, Christophe Gilbert, Patricia Doublet, Sophie Jarraud, and Annelise Chapalain
- Subjects
legionella pneumophila ,clinical isolates ,sequence type ,st1 ,st47 ,tnf-α ,immune response ,macrophages ,cell death ,Infectious and parasitic diseases ,RC109-216 - Abstract
Legionnaires’ Disease (LD) is a severe pneumonia mainly caused in Europe by Legionella pneumophila serogroup 1 (Lp1). Sequence-based typing methods reveal that some sequence types (ST) are overrepresented in clinical samples such as ST1 and ST47, suggesting that some strains are more fit for infection than others. In the present study, a collection of 108 Lp1 clinical isolates were used to evaluate the strain-dependent immune responses from human macrophages. Clinical Lp1 isolates induced differential TNFα secretion from macrophages. ST1 isolates induced a significantly higher TNF-α secretion than non-ST1, whereas ST47 isolates induced a significantly lower TNF-α secretion than non-ST47 isolates. ST1 isolates induced a significantly higher cell death than ST47 isolates evaluated by lactate dehydrogenase activity (cytotoxicity) and caspase-3 activity (apoptosis). Treatment of macrophages with anti-TNF-α antibodies significantly reduced the cell death in macrophages infected with ST1 or ST47 strains. The TNF-α secretion was neither explained by a differential bacterial replication nor by the number or type (bystander or infected) of TNF-α producing cells following infection but by a differential response from macrophages. The Paris ST1 reference strain elicited a significantly higher TNF-α gene transcription and a higher induction of NF-κB signaling pathway than the Lorraine ST47 reference strain. Clinical Lp1 isolates induce a diverse immune response and cell death, which could be related to the genotype. The two predominant sequence-types ST1 and ST47 trigger opposite inflammatory response that could be related to the host susceptibility.
- Published
- 2022
- Full Text
- View/download PDF
3. Hyper-inflammatory profile and immunoparalysis in patients with severe Legionnaires’ disease
- Author
-
Camille Allam, William Mouton, Hugo Testaert, Christophe Ginevra, Noémie Fessy, Marine Ibranosyan, Ghislaine Descours, Laetitia Beraud, Johann Guillemot, Annelise Chapalain, Chloé Albert-Vega, Jean-Christophe Richard, Laurent Argaud, Arnaud Friggeri, Vanessa Labeye, Yvan Jamilloux, Nathalie Freymond, Fabienne Venet, Gérard Lina, Patricia Doublet, Florence Ader, Sophie Trouillet-Assant, and Sophie Jarraud
- Subjects
Legionnaires’ disease ,hyper-inflammation ,immunoparalysis ,immune functional assays ,severity ,LPS stimulation ,Microbiology ,QR1-502 - Abstract
IntroductionSevere Legionnaires’ disease (LD) can lead to multi-organ failure or death in 10%–30% of patients. Although hyper-inflammation and immunoparalysis are well described in sepsis and are associated with high disease severity, little is known about the immune response in LD. This study aimed to evaluate the immune status of patients with LD and its association with disease severity.MethodsA total of 92 hospitalized LD patients were included; 19 plasmatic cytokines and pulmonary Legionella DNA load were measured in 84 patients on the day of inclusion (day 0, D0). Immune functional assays (IFAs) were performed from whole blood samples collected at D2 and stimulated with concanavalin A [conA, n = 19 patients and n = 21 healthy volunteers (HV)] or lipopolysaccharide (LPS, n = 14 patients and n = 9 HV). A total of 19 cytokines (conA stimulation) and TNF-α (LPS stimulation) were quantified from the supernatants. The Sequential Organ Failure Assessment (SOFA) severity score was recorded at D0 and the mechanical ventilation (MV) status was recorded at D0 and D8.ResultsAmong the 84 patients, a higher secretion of plasmatic MCP-1, MIP1-β, IL-6, IL-8, IFN-γ, TNF-α, and IL-17 was observed in the patients with D0 and D8 MV. Multiparametric analysis showed that these seven cytokines were positively associated with the SOFA score. Upon conA stimulation, LD patients had a lower secretion capacity for 16 of the 19 quantified cytokines and a higher release of IL-18 and MCP-1 compared to HV. IL-18 secretion was higher in D0 and D8 MV patients. TNF-α secretion, measured after ex vivo LPS stimulation, was significantly reduced in LD patients and was associated with D8 MV status.DiscussionThe present findings describe a hyper-inflammatory phase at the initial phase of Legionella pneumonia that is more pronounced in patients with severe LD. These patients also present an immunoparalysis for a large number of cytokines, except IL-18 whose secretion is increased. An assessment of the immune response may be relevant to identify patients eligible for future innovative host-directed therapies.
- Published
- 2023
- Full Text
- View/download PDF
4. Bacterial persistence in Legionella pneumophila clinical isolates from patients with recurring legionellosis
- Author
-
Xanthe Adams-Ward, Annelise Chapalain, Christophe Ginevra, Sophie Jarraud, Patricia Doublet, and Christophe Gilbert
- Subjects
L. pneumophila ,virulence ,bacterial persistence ,antibiotic tolerance ,recurring legionellosis ,amoeba ,Microbiology ,QR1-502 - Abstract
Bacterial persisters are a transient subpopulation of non-growing, antibiotic-tolerant cells. There is increasing evidence that bacterial persisters play an important role in treatment failure leading to recurring infections and promoting the development of antibiotic resistance. Current research reveals that recurring legionellosis is often the result of relapse rather than reinfection and suggests that the mechanism of bacterial persistence may play a role. The development of single-cell techniques such as the Timerbac system allows us to identify potential persister cells and investigate their physiology. Here, we tested the persister forming capacity of 7 pairs of Legionella pneumophila (Lp) clinical isolates, with isolate pairs corresponding to two episodes of legionellosis in the same patient. We distinguished non-growing subpopulations from their replicating counterparts during infection in an amoeba model. Imaging flow cytometry allowed us to identify single non-growing bacteria within amoeba cells 17 h post-infection, thus corresponding to this subpopulation of potential persister cells. Interestingly the magnitude of this subpopulation varies between the 7 pairs of Lp clinical isolates. Biphasic killing kinetics using ofloxacin stress confirmed the persister development capacity of ST1 clinical isolates, highlighting enhanced persister formation during the host cell infection. Thus, persister formation appears to be strain or ST (sequence type) dependent. Genome sequence analysis was carried out between ST1 clinical isolates and ST1 Paris. No genetic microevolution (SNP) linked to possible increase of persistence capacity was revealed among all the clones tested, even in clones issued from two persistence cycle experiments, confirming the transient reversible phenotypic status of persistence. Treatment failure in legionellosis is a serious issue as infections have a 5-10% mortality rate, and investigations into persistence in a clinical context and the mechanisms involved may allow us to combat this issue.
- Published
- 2023
- Full Text
- View/download PDF
5. Co-infection with Legionella and SARS-CoV-2, France, March 2020
- Author
-
Camille Allam, Alexandre Gaymard, Ghislaine Descours, Christophe Ginevra, Laurence Josset, Maud Bouscambert, Laetitia Beraud, Marine Ibranosyan, Camille Golfier, Arnaud Friggeri, Bruno Lina, Christine Campèse, Florence Ader, and Sophie Jarraud
- Subjects
coronavirus disease ,COVID-19 ,Legionnaires’ disease ,Legionella ,pneumonia ,respiratory infections ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
We describe a March 2020 co-occurrence of Legionnaires’ disease (LD) and coronavirus disease in France. Severe acute respiratory syndrome coronavirus 2 co-infections were identified in 7 of 49 patients from LD case notifications. Most were elderly men with underlying conditions who had contracted severe pneumonia, illustrating the relevance of co-infection screening.
- Published
- 2021
- Full Text
- View/download PDF
6. Improving the Diagnosis of Bacterial Infections: Evaluation of 16S rRNA Nanopore Metagenomics in Culture-Negative Samples
- Author
-
Coralie Bouchiat, Christophe Ginevra, Yvonne Benito, Tiphaine Gaillard, Hélène Salord, Olivier Dauwalder, Frédéric Laurent, and François Vandenesch
- Subjects
metagenomics ,bacterial infections ,molecular diagnosis ,Nanopore sequencing ,16S rRNA ,Microbiology ,QR1-502 - Abstract
While 16S rRNA PCR-Sanger sequencing has paved the way for the diagnosis of culture-negative bacterial infections, it does not provide the composition of polymicrobial infections. We aimed to evaluate the performance of the Nanopore-based 16S rRNA metagenomic approach, using both partial and full-length amplification of the gene, and to explore its feasibility and suitability as a routine diagnostic tool for bacterial infections in a clinical laboratory. Thirty-one culture-negative clinical samples from mono- and polymicrobial infections based on Sanger-sequencing results were sequenced on MinION using both the in-house partial amplification and the Nanopore dedicated kit for the full-length amplification of the 16S rRNA gene. Contamination, background noise definition, bacterial identification, and time-effectiveness issues were addressed. Cost optimization was also investigated with the miniaturized version of the flow cell (Flongle). The partial 16S approach had a greater sensitivity compared to the full-length kit that detected bacterial DNA in only 24/31 (77.4%) samples. Setting a threshold of 1% of total reads overcame the background noise issue and eased the interpretation of clinical samples. Results were obtained within 1 day, discriminated polymicrobial samples, and gave accurate bacterial identifications compared to Sanger-based results. We also found that multiplexing and using Flongle flow cells was a cost-effective option. The results confirm that Nanopore technology is user-friendly as well as cost- and time-effective. They also indicate that 16S rRNA targeted metagenomics is a suitable approach to be implemented for the routine diagnosis of culture-negative samples in clinical laboratories.
- Published
- 2022
- Full Text
- View/download PDF
7. Enterobacter cloacae colonisation and infection in a neonatal intensive care unit: retrospective investigation of preventive measures implemented after a multiclonal outbreak
- Author
-
Alexandrine Ferry, Frank Plaisant, Christophe Ginevra, Yann Dumont, Jacqueline Grando, Olivier Claris, François Vandenesch, and Marine Butin
- Subjects
Enterobacter cloacae ,NICU ,Outbreak ,Cephalosporins ,Biocleaning ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background Enterobacter cloacae species is responsible for nosocomial outbreaks in vulnerable patients in neonatal intensive care units (NICU). The environment can constitute the reservoir and source of infection in NICUs. Herein we report the impact of preventive measures implemented after an Enterobacter cloacae outbreak inside a NICU. Methods This retrospective study was conducted in one level 3 NICU in Lyon, France, over a 6 year-period (2012–2018). After an outbreak of Enterobacter cloacae infections in hospitalized neonates in 2013, several measures were implemented including intensive biocleaning and education of medical staff. Clinical and microbiological characteristics of infected patients and evolution of colonization/infection with Enterobacter spp. in this NICU were retrieved. Moreover, whole genome sequencing was performed on 6 outbreak strains. Results Enterobacter spp. was isolated in 469 patients and 30 patients developed an infection including 2 meningitis and 12 fatal cases. Preventive measures and education of medical staff were not associated with a significant decrease in patient colonisation but led to a persistent decreased use of cephalosporin in the NICU. Infection strains were genetically diverse, supporting the hypothesis of multiple hygiene defects rather than the diffusion of a single clone. Conclusions Grouped cases of infections inside one setting are not necessarily related to a single-clone outbreak and could reveal other environmental and organisational problematics. The fight against implementation and transmission of Enterobacter spp. in NICUs remains a major challenge.
- Published
- 2020
- Full Text
- View/download PDF
8. Bacterial Long-Range Warfare: Aerial Killing of Legionella pneumophila by Pseudomonas fluorescens
- Author
-
Marie-Hélène Corre, Anne Mercier, Mathilde Bouteiller, Alix Khalil, Christophe Ginevra, Ségolène Depayras, Charly Dupont, Meg Rouxel, Mathias Gallique, Laettitia Grac, Sophie Jarraud, David Giron, Annabelle Merieau, Jean-Marc Berjeaud, and Julien Verdon
- Subjects
Legionella ,Pseudomonas ,volatile organic compounds ,1-undecene ,antibacterial activity ,SPME ,Microbiology ,QR1-502 - Abstract
ABSTRACT Legionella pneumophila, the causative agent of Legionnaires’ disease, is mostly found in man-made water systems and is one of the most closely monitored waterborne pathogens. With the aim of finding natural ways to control waterborne pathogens and thus further reduce the impact of disinfection by-products on human health, some studies have demonstrated the ability of bacteria to kill Legionella through the production of secondary metabolites or antimicrobial compounds. Here, we describe an unexpected growth inhibition of L. pneumophila when exposed to a physically separated strain of Pseudomonas fluorescens, designated as MFE01. Most of the members of the Legionellaceae family are sensitive to the volatile substances emitted by MFE01, unlike other bacteria tested. Using headspace solid-phase microextraction GC-MS strategy, a volatilome comparison revealed that emission of 1-undecene, 2-undecanone, and 2-tridecanone were mainly reduced in a Tn5-transposon mutant unable to inhibit at distance the growth of L. pneumophila strain Lens. We showed that 1-undecene was mainly responsible for the inhibition at distance in vitro, and led to cell lysis in small amounts, as determined by gas chromatography-mass spectrometry (GC-MS). Collectively, our results provide new insights into the mode of action of bacterial volatiles and highlight them as potent anti-Legionella agents to focus research on novel strategies to fight legionellosis. IMPORTANCE Microbial volatile compounds are molecules whose activities are increasingly attracting the attention of researchers. Indeed, they can act as key compounds in long-distance intrakingdom and interkingdom communication, but also as antimicrobials in competition and predation. In fact, most studies to date have focused on their antifungal activities and only a few have reported on their antibacterial properties. Here, we describe that 1-undecene, naturally produced by P. fluorescens, is a volatile with potent activity against bacteria of the genus Legionella. In small amounts, it is capable of inducing cell lysis even when the producing strain is physically separated from the target. This is the first time that such activity is described. This molecule could therefore constitute an efficient compound to counter bacterial pathogens whose treatment may fail, particularly in pulmonary diseases. Indeed, inhalation of these volatiles should be considered as a possible route of therapy in addition to antibiotic treatment.
- Published
- 2021
- Full Text
- View/download PDF
9. Transmission of Legionnaires’ Disease through Toilet Flushing
- Author
-
Jeanne Couturier, Christophe Ginevra, Didier Nesa, Marine Adam, Cyril Gouot, Ghislaine Descours, Christine Campèse, Giorgia Battipaglia, Eolia Brissot, Laetitia Beraud, Anne-Gaëlle Ranc, Sophie Jarraud, and Frédéric Barbut
- Subjects
Bacteria ,healthcare-associated infections ,Legionnaires’ disease ,nosocomial infections ,phylogeny ,pneumophilia ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
We describe 2 cases of healthcare-associated Legionnaires’ disease in patients in France hospitalized 5 months apart in the same room. Whole-genome sequencing analyses showed that clinical isolates from the patients and isolates from the room’s toilet clustered together. Toilet contamination by Legionella pneumophila could lead to a risk for exposure through flushing.
- Published
- 2020
- Full Text
- View/download PDF
10. The clinical presentation of Legionella arthritis reveals the mode of infection and the bacterial species: case report and literature review
- Author
-
Marine Ibranosyan, Laetitia Beraud, Hélène Lemaire, Anne-Gaëlle Ranc, Christophe Ginevra, Sophie Jarraud, and Ghislaine Descours
- Subjects
Legionella bozemanii ,Arthritis ,Immunosuppression ,Corticosteroid ,Tocilizumab ,Methotrexate ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background While Legionella is a common cause of pneumonia, extrapulmonary infections like arthritis are scarce. Here, we describe a case of monoarthritis due to Legionella bozemanii, with no history of pneumonia. We provide a literature review of the 9 previously published Legionella arthritis and highlight a dichotomous epidemiology suggesting different physiopathological pathways leading to joint infection. Case presentation A 56-year old woman under immunosuppressive treatment by oral and intra-articular corticosteroids, methotrexate, and tocilizumab for an anti-synthetase syndrome was hospitalized for worsening pain and swelling of the left wrist for 3 days. Clinical examination showed left wrist synovitis and no fever. The arthritis occurred a few days after an accidental fall on wet asphalt responsible for a cutaneous wound followed by a corticosteroid intra-articular injection. Due to both the negativity of conventional culture of articular fluid and suspicion of infection, 16S rRNA and specific PCRs were performed leading to the identification of L. bozemanii. Legionella-specific culture of the articular fluid was performed retrospectively and isolated L. bozemanii. The empiric antibiotic therapy was switched for oral levofloxacin and rifampin and the patient recovered after a 12-week treatment. Conclusion We report a case of L. bozemanii monoarthritis in an immunosuppressed woman, following a fall on wet asphalt and intra-articular corticosteroid injection. The review of the literature found that the clinical presentation reveals the mode of infection and the bacterial species. Monoarthritis more likely occurred after inoculation in patients under immunosuppressive therapy and were associated with non-Legionella pneumophila serogroup 1 (Lp1) strains that predominate in the environment. Polyarthritis were more likely secondary legionellosis localizations after blood spread of Lp1, the most frequently found in pneumonia. In both settings, 16S rRNA and Legionella-specific PCR were key factors for the diagnosis.
- Published
- 2019
- Full Text
- View/download PDF
11. Rifampicin exposure reveals within-host Mycobacterium tuberculosis diversity in patients with delayed culture conversion.
- Author
-
Charlotte Genestet, Elisabeth Hodille, Alexia Barbry, Jean-Luc Berland, Jonathan Hoffmann, Emilie Westeel, Fabiola Bastian, Michel Guichardant, Samuel Venner, Gérard Lina, Christophe Ginevra, Florence Ader, Sylvain Goutelle, and Oana Dumitrescu
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Mycobacterium tuberculosis (Mtb) genetic micro-diversity in clinical isolates may underline mycobacterial adaptation to tuberculosis (TB) infection and provide insights to anti-TB treatment response and emergence of resistance. Herein we followed within-host evolution of Mtb clinical isolates in two cohorts of TB patients, either with delayed Mtb culture conversion (> 2 months), or with fast culture conversion (< 2 months). We captured the genetic diversity of Mtb isolates obtained in each patient, by focusing on minor variants detected as unfixed single nucleotide polymorphisms (SNPs). To unmask antibiotic tolerant sub-populations, we exposed these isolates to rifampicin (RIF) prior to whole genome sequencing (WGS) analysis. Thanks to WGS, we detected at least 1 unfixed SNP within the Mtb isolates for 9/15 patients with delayed culture conversion, and non-synonymous (ns) SNPs for 8/15 patients. Furthermore, RIF exposure revealed 9 additional unfixed nsSNP from 6/15 isolates unlinked to drug resistance. By contrast, in the fast culture conversion cohort, RIF exposure only revealed 2 unfixed nsSNP from 2/20 patients. To better understand the dynamics of Mtb micro-diversity, we investigated the variant composition of a persistent Mtb clinical isolate before and after controlled stress experiments mimicking the course of TB disease. A minor variant, featuring a particular mycocerosates profile, became enriched during both RIF exposure and macrophage infection. The variant was associated with drug tolerance and intracellular persistence, consistent with the pharmacological modeling predicting increased risk of treatment failure. A thorough study of such variants not necessarily linked to canonical drug-resistance, but which are prone to promote anti-TB drug tolerance, may be crucial to prevent the subsequent emergence of resistance. Taken together, the present findings support the further exploration of Mtb micro-diversity as a promising tool to detect patients at risk of poorly responding to anti-TB treatment, ultimately allowing improved and personalized TB management.
- Published
- 2021
- Full Text
- View/download PDF
12. Persistent Legionnaires’ Disease and Associated Antibiotic Treatment Engender a Highly Disturbed Pulmonary Microbiome Enriched in Opportunistic Microorganisms
- Author
-
Ana Elena Pérez-Cobas, Christophe Ginevra, Christophe Rusniok, Sophie Jarraud, and Carmen Buchrieser
- Subjects
Legionella pneumophila ,antibiotic resistance ,pneumonia ,pulmonary microbiome ,Microbiology ,QR1-502 - Abstract
ABSTRACT Despite the importance of pneumonia to public health, little is known about the composition of the lung microbiome during infectious diseases, such as pneumonia, and how it evolves during antibiotic therapy. To study the possible relation of the pulmonary microbiome to the severity and outcome of this respiratory disease, we analyzed the dynamics of the pathogen and the human lung microbiome during persistent infections caused by the bacterium Legionella pneumophila and their evolution during antimicrobial treatment. We collected 10 bronchoalveolar lavage fluid samples from three patients during long-term hospitalization due to pneumonia and performed a unique longitudinal study of the interkingdom microbiome, analyzing the samples for presence of bacteria, archaea, fungi, and protozoa by high-throughput Illumina sequencing of marker genes. The lung microbiome of the patients was characterized by a strong predominance of the pathogen, a low diversity of the bacterial fraction, and an increased presence of opportunistic microorganisms. The fungal fraction was more stable than the bacterial fraction. During long-term treatment, no genomic changes or antibiotic resistance-associated mutations that could explain the persistent infection occurred, according to whole-genome sequencing analyses of the pathogen. After antibiotic treatment, the microbiome did not recover rapidly but was mainly constituted of antibiotic-resistant species and enriched in bacteria, archaea, fungi, or protozoa associated with pathogenicity. The lung microbiome seems to contribute to nonresolving Legionella pneumonia, as it is strongly disturbed during infection and enriched in opportunistic and/or antibiotic-resistant bacteria and microorganisms, including fungi, archaea, and protozoa that are often associated with infections. IMPORTANCE The composition and dynamics of the lung microbiome during pneumonia are not known, although the lung microbiome might influence the severity and outcome of this infectious disease, similar to what was shown for the microbiome at other body sites. Here we report the findings of a comprehensive analysis of the lung microbiome composition of three patients with long-term pneumonia due to L. pneumophila and its evolution during antibiotic treatment. This work adds to our understanding of how the microbiome changes during disease and antibiotic treatment and points to microorganisms and their interactions that might be beneficial. In addition to bacteria and fungi, our analyses included archaea and eukaryotes (protozoa), showing that both are present in the pulmonary microbiota and that they might also play a role in the response to the microbiome disturbance.
- Published
- 2020
- Full Text
- View/download PDF
13. Polymorphisms of a Collagen-Like Adhesin Contributes to Legionella pneumophila Adhesion, Biofilm Formation Capacity and Clinical Prevalence
- Author
-
Mena Abdel-Nour, Han Su, Carla Duncan, Shaopei Li, Deepa Raju, Feras Shamoun, Marine Valton, Christophe Ginevra, Sophie Jarraud, Cyril Guyard, Kagan Kerman, and Mauricio R. Terebiznik
- Subjects
collagen-like adhesion ,biofilm ,Legionella pneumophila ,cell–cell adhesion ,clinical prevalence ,adhesion ,Microbiology ,QR1-502 - Abstract
Legionellosis is a severe respiratory illness caused by the inhalation of aerosolized water droplets contaminated with the opportunistic pathogen Legionella pneumophila. The ability of L. pneumophila to produce biofilms has been associated with its capacity to colonize and persist in human-made water reservoirs and distribution systems, which are the source of legionellosis outbreaks. Nevertheless, the factors that mediate L. pneumophila biofilm formation are largely unknown. In previous studies we reported that the adhesin Legionella collagen-like protein (Lcl), is required for auto-aggregation, attachment to multiple surfaces and the formation of biofilms. Lcl structure contains three distinguishable regions: An N-terminal region with a predicted signal sequence, a central region containing tandem collagen-like repeats (R-domain) and a C-terminal region (C-domain) with no significant homology to other known proteins. Lcl R-domain encodes tandem repeats of the collagenous tripeptide Gly-Xaa-Yaa (GXY), a motif that is key for the molecular organization of mammalian collagen and mediates the binding of collagenous proteins to different cellular and environmental ligands. Interestingly, Lcl is polymorphic in the number of GXY tandem repeats. In this study, we combined diverse biochemical, genetic, and cellular approaches to determine the role of Lcl domains and GXY repeats polymorphisms on the structural and functional properties of Lcl, as well as on bacterial attachment, aggregation and biofilm formation. Our results indicate that the R-domain is key for assembling Lcl collagenous triple-helices and has a more preponderate role over the C-domain in Lcl adhesin binding properties. We show that Lcl molecules oligomerize to form large supramolecular complexes to which both, R and C-domains are required. Furthermore, we found that the number of GXY tandem repeats encoded in Lcl R-domain correlates positively with the binding capabilities of Lcl and with the attachment and biofilm production capacity of L. pneumophila strains. Accordingly, the number of GXY tandem repeats in Lcl influences the clinical prevalence of L. pneumophila strains. Therefore, the number of Lcl tandem repeats could be considered as a potential predictor for virulence in L. pneumophila isolates.
- Published
- 2019
- Full Text
- View/download PDF
14. Detection of Legionella Anisa in Water from Hospital Dental Chair Units and Molecular Characterization by Whole-Genome Sequencing
- Author
-
Giuseppe Fleres, Natacha Couto, Mariette Lokate, Luc W. M. van der Sluis, Christophe Ginevra, Sophie Jarraud, Ruud H. Deurenberg, John W. Rossen, Silvia García-Cobos, and Alex W. Friedrich
- Subjects
Legionella spp. ,whole-genome sequencing (WGS) ,core-genome multi-locus sequence typing (cgMLST) ,whole-genome multi-locus sequence typing (wgMLST) ,next-generation sequencing (NGS) ,Biology (General) ,QH301-705.5 - Abstract
This study aims to assess contamination with Legionella spp. in water from dental chair units (DCUs) of a hospital dental ward and to perform its molecular characterization by whole-genome sequencing (WGS). We collect eight water samples (250 mL) from four DCUs (sink and water-syringe). Samples are tested for the presence of Legionella spp. (CFUs/mL) by culturing according to the Nederland Norm (NEN) 6265. Three DCUs are found positive for Legionella anisa, and four isolates are cultured (sink n = 2, water-syringe n = 1; two isolates from the same chair) with 1 × 102 CFU/mL. Whole-genome multi-locus sequence typing (wgMLST) results indicate that all strains belong to the same cluster with two to four allele differences. Classical culture combined with WGS allows the identification of a unique clone of L. anisa in several DCUs in the same hospital dental ward. This may indicate a common contamination source in the dental unit waterlines, which was fixed by replacing the chairs and main pipeline of the unit. Our results reveal tap water contamination in direct contact with patients and the usefulness of WGS to investigate bacterial molecular epidemiology.
- Published
- 2018
- Full Text
- View/download PDF
15. Lorraine Strain of Legionella pneumophila Serogroup 1, France
- Author
-
Christophe Ginevra, Françoise Forey, Christine Campèse, Monique Reyrolle, Didier Che, Jerome Etienne, and Sophie Jarraud
- Subjects
Legionella ,epidemiology ,endemic strain ,emergence ,letter ,France ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Published
- 2008
- Full Text
- View/download PDF
16. Fungal Integrated Histomolecular Diagnosis Using Targeted Next-Generation Sequencing on Formalin-Fixed Paraffin-Embedded Tissues
- Author
-
Alexis Trecourt, Meja Rabodonirina, Claire Mauduit, Alexandra Traverse-Glehen, Mojgan Devouassoux-Shisheboran, David Meyronet, Frédérique Dijoud, Christophe Ginevra, Emmanuelle Chapey-Picq, Emilie Josse, Patricia Martins-Simoes, Abderrazzak Bentaher, Damien Dupont, Charline Miossec, Florence Persat, Martine Wallon, Tristan Ferry, Félix Pham, Bruno Simon, and Jean Menotti
- Subjects
Microbiology (medical) - Abstract
Histopathology is the gold standard for fungal infection (FI) diagnosis, but it does not provide a genus and/or species identification. The objective of the present study was to develop targeted next-generation sequencing (NGS) on formalin-fixed tissue samples (FTs) to achieve a fungal integrated histomolecular diagnosis.
- Published
- 2023
- Full Text
- View/download PDF
17. Rapid adaptations of Legionella pneumophila to the human host
- Author
-
Daniël Leenheer, Anaísa B. Moreno, Kiran Paranjape, Susan Murray, Sophie Jarraud, Christophe Ginevra, and Lionel Guy
- Subjects
Evolutionsbiologi ,Legionnaires? disease ,Mikrobiologi ,Evolutionary Biology ,host-specific adaptations ,molecular evolution ,Mikrobiologi inom det medicinska området ,comparative genomics ,General Medicine ,Microbiology ,Legionella pneumophila ,Microbiology in the medical area - Abstract
Legionella pneumophila are host-adapted bacteria that infect and reproduce primarily in amoeboid protists. Using similar infection mechanisms, they infect human macrophages, and cause Legionnaires’ disease, an atypical pneumonia, and the milder Pontiac fever. We hypothesized that, despite the similarities in infection mechanisms, the hosts are different enough that there exist high-selective value mutations that would dramatically increase the fitness of Legionella inside the human host. By comparing a large number of isolates from independent infections, we identified two genes, mutated in three unrelated patients, despite the short duration of the incubation period (2–14 days). One is a gene coding for an outer membrane protein (OMP) belonging to the OmpP1/FadL family. The other is a gene coding for an EAL-domain-containing protein involved in cyclic-di-GMP regulation, which in turn modulates flagellar activity. The clinical strain, carrying the mutated EAL-domain-containing homologue, grows faster in macrophages than the wild-type strain, and thus appears to be better adapted to the human host. As human-to-human transmission is very rare, fixation of these mutations into the population and spread into the environment is unlikely. Therefore, parallel evolution – here mutations in the same genes observed in independent human infections – could point to adaptations to the accidental human host. These results suggest that despite the ability of L. pneumophila to infect, replicate in and exit from macrophages, its human-specific adaptations are unlikely to be fixed in the population.
- Published
- 2023
- Full Text
- View/download PDF
18. Co-infection with Legionella and SARS-CoV-2, France, March 2020
- Author
-
Laetitia Beraud, Marine Ibranosyan, C. Campèse, Camille Allam, Alexandre Gaymard, Camille Golfier, Florence Ader, Sophie Jarraud, Arnaud Friggeri, Ghislaine Descours, Laurence Josset, Maud Bouscambert, Christophe Ginevra, and Bruno Lina
- Subjects
Microbiology (medical) ,Male ,2019-20 coronavirus outbreak ,Epidemiology ,Legionella ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Disease ,macromolecular substances ,Infectious and parasitic diseases ,RC109-216 ,medicine.disease_cause ,respiratory infections ,medicine ,Humans ,pneumonia ,viruses ,Coronavirus ,Original Research ,Aged ,biology ,business.industry ,SARS-CoV-2 ,Coinfection ,Dispatch ,COVID-19 ,Co-infection with Legionella and SARS-CoV-2, France, March 2020 ,medicine.disease ,biology.organism_classification ,Virology ,zoonoses ,Pneumonia ,Infectious Diseases ,coronavirus disease ,Medicine ,Legionnaires' disease ,France ,business ,Co infection ,severe acute respiratory syndrome coronavirus 2 ,Legionnaires’ disease - Abstract
We describe a March 2020 co-occurrence of Legionnaires' disease (LD) and coronavirus disease in France. Severe acute respiratory syndrome coronavirus 2 co-infections were identified in 7 of 49 patients from LD case notifications. Most were elderly men with underlying conditions who had contracted severe pneumonia, illustrating the relevance of co-infection screening.
- Published
- 2021
19. P454 Massive parallel fungal sequencing on formalin-fixed tissues: development and contribution in integrated histomolecular diagnosis
- Author
-
Alexis Trecourt, Meja Rabodonirina, Emilie Josse, M. Christophe Ginevra, Emmanuelle Chapey-Picq, Damien Dupont, Charline Miossec, Florence Persat, Claire Mauduit, Alexandra Traverse-Glehen, David Meyronet, Martine Wallon, Brunio Simon, and Jean Menotti
- Subjects
Infectious Diseases ,General Medicine - Abstract
Poster session 3, September 23, 2022, 12:30 PM - 1:30 PM Objectives Histopathology is the gold standard for distinguishing between colonization and fungal infection, but it does not provide a precise diagnosis of genera/species. However, if the culture is negative or if no specimen is sent to the Mycology laboratory, only the specimen sent to the Pathology department is available. Formalin fixation and paraffin embedding (FFPE) cause DNA damage, making it difficult to perform molecular techniques. The objective was to develop and evaluate the contribution of massive parallel sequencing (MPS) to FFPE tissues. Methods Histopathological review of all cases was performed. Then, DNA extraction from FFPE tissues was optimized by: (1) macrodissecting the fungal-rich area on the paraffin block; (2) comparing the efficiency of two DNA extraction kits (QIAamp DNAmicro-kit, QIAGEN; Maxwell 16 LEVRNA FFPE Purification kit, Promega, optimized for RNA and DNA extraction), by comparison of Aspergillus fumigatus and Mucorale specific PCR results for 30 cases. For 124 other cases, the sensitivity of two primer pairs (ITS3/4 and MITS2A/2B) was tested for identification by Sanger sequencing and then MPS. Finally, a histomolecular comparison was performed. This work was funded by the Société de Pathologie Infectieuse de Langue Française (SPILF). Results To optimize extraction, DNA was extracted by both kits from samples of 16 mucormycoses and 14 A. fumigatus infections. PCR sensitivity was better with the QIAGEN extraction kit [100% (30/30)] compared to the Promega kit [86.7% (26/30)]. PCR amplification of fungal DNA from an additional 124 FFPE samples was performed. The primer pairs ITS3/4 and MITS2A/2B, allowed: (1) identification by Sanger sequencing-histopathological analysis in 38.7% (48/124) of the cases in total, and more specifically 33% (41/124) of cases with the ITS3/4 primers and 32.3% (40/124) of cases with the MITS2A/B primers; and (2) identification by integrated SMP-histopathological analysis in 75% (93/124) of all cases (primers ITS3/4 and MITS2A/2B), and more specifically 66.1% (82/124) for ITS3/4 and 62.1% (77/124) for MITS2A/B (both primer pairs did not detect/amplify the same fungal genera/species). The combination of all results from Sanger sequencing and MPS led to fungal identification in 75.8% (94/124) of cases. In total, the addition of NGS to Sanger sequencing increased the diagnostic proportion by 36.3% (45/124; P Conclusion The development of the fungal MPS on FFPE tissues is innovative and unprecedented for the achievement of an integrated histomolecular diagnosis in fungal pathology. It increases significantly the diagnostic proportion by 36.3%. This strategy can be used in hospitals and could improve patient management, especially when no sample is sent to the Mycology laboratory or when the culture is negative.
- Published
- 2022
- Full Text
- View/download PDF
20. Rapid, in-patient adaptations of Legionella pneumophila to the human host
- Author
-
Daniël Leenheer, Anaísa B. Moreno, Susan Murray, Sophie Jarraud, Christophe Ginevra, and Lionel Guy
- Abstract
Legionella pneumophila are host-adapted bacteria that infect and reproduce primarily in amoeboid protists. Using similar infection mechanisms, they infect human macrophages, and cause Legionnaires’ disease, an atypical pneumonia, and the milder Pontiac fever. We hypothesized that, despite these similarities, the hosts are different enough so that there exist high-selective value mutations that would dramatically increase the fitness of Legionella inside the human host. By comparing a large number of isolates from independent infections, we identified two genes, mutated in three unrelated patients, despite the short duration of the incubation period (2-14 days). One is a gene coding for an outer membrane protein (OMP) belonging to the OmpP1/FadL family. The clinical strain, carrying the mutated OMP homolog, grows faster in macrophages than the wild type strain, and thus appears to be better adapted to the human host. The other is a gene coding for a protein involved in cyclic-di-GMP regulation, which in turn modulates flagellar activity. As human-to-human transmission is very rare, fixation of these mutations into the population and spread into the environment is unlikely. Therefore, convergent evolution – here mutations in the same genes observed in independent human infections – could point to adaptations to the accidental human host. These results suggest that despite its ability to infect, replicate, and disperse from amoebae, L. pneumophila is not well adapted to the human host.Impact statementLegionella pneumophila is primarily infecting amoeboid protists, but occasionally infects human lung macrophages, causing Legionnaires’ disease, an atypical pneumonia. By comparing 171 isolates from patients to their probable environmental source, we identified 119 mutations that presumably occurred in-patient. Among these, several mutations occurred in the gene. In particular, two genes were mutated thrice, significantly more often than expected by chance alone, and are likely to represent adaptations to the human host. We experimentally show that, for one mutation at least, the mutated strain grows faster in human macrophages than in amoebae. By specifically investigating in-patient mutations, we were able to identify two genes that might be involved in human host-specific adaptations of L. pneumophila. This result suggests that L. pneumophila is not particularly well adapted to the human host, as mutations get fixed in-patient, during the short course of an infection (2-14 days), indicating a very high selective value.Data SummaryThe sequencing data generated in this study are available in the NCBI database under the BioProject accession number: PRJEB52976.
- Published
- 2022
- Full Text
- View/download PDF
21. Detection of highly macrolide-resistant Legionella pneumophila strains from a hotel water network using systematic whole-genome sequencing
- Author
-
Christophe Ginevra, Laetitia Beraud, Isabelle Pionnier, Kassandra Sallabery, Houcine Bentayeb, Bruno Simon, Camille Allam, Joelle Chastang, Marine Ibranosyan, Véronique Decroix, Christine Campese, Sophie Jarraud, and Ghislaine Descours
- Subjects
Pharmacology ,Microbiology (medical) ,Infectious Diseases ,Drug Resistance, Bacterial ,Water ,Pharmacology (medical) ,Macrolides ,Microbial Sensitivity Tests ,Anti-Bacterial Agents ,Legionella pneumophila - Abstract
Objectives Implementation of an antibiotic resistance detection tool in Legionella daily surveillance at the French National Reference Centre for Legionella. Methods Systematic WGS of Legionella pneumophila isolates and bioinformatics detection of specific mutations linked to antibiotic resistance. Phenotypic validation of antibiotic resistance detected by WGS was performed by the broth microdilution method. Results More than 3000 L. pneumophila strains were screened for antibiotic resistance. A macrolide resistance-associated A2052G mutation in the 23S rRNA gene was identified in the genome of eight isolates from a hotel water network. High-level macrolide resistance (i.e. MICs of 1024–2048 mg/L for azithromycin and erythromycin) with no cross-resistance to other antimicrobials was phenotypically confirmed by antimicrobial susceptibility testing for the eight isolates. Conclusions Systematic WGS of L. pneumophila is a powerful tool for first-line high-throughput screening of antibiotic resistance before phenotypic validation.
- Published
- 2022
22. Genome analysis of Legionella pneumophila ST23 from various countries reveals highly similar strains
- Author
-
Maria Luisa Ricci, Silvia Fillo, Andrea Ciammaruconi, Florigio Lista, Christophe Ginevra, Sophie Jarraud, Antonietta Girolamo, Fabrizio Barbanti, Maria Cristina Rota, Diane Lindsay, Jamie Gorzynski, Søren A Uldum, Sharmin Baig, Marina Foti, Giancarlo Petralito, Stefania Torri, Marino Faccini, Maira Bonini, Gabriella Gentili, Sabrina Senatore, Anna Lamberti, Joao André Carrico, Maria Scaturro, and Repositório da Universidade de Lisboa
- Subjects
Ecology ,Health, Toxicology and Mutagenesis ,Plant Science ,Biochemistry, Genetics and Molecular Biology (miscellaneous) - Abstract
© 2022 Ricci et al. This article is available under a CreativeCommons License (Attribution 4.0 International, as described at https://creativecommons.org/licenses/by/4.0/)., Legionella pneumophila serogroup 1 (Lp1) sequence type (ST) 23 is one of the most commonly detected STs in Italy where it currently causes all investigated outbreaks. ST23 has caused both epidemic and sporadic cases between 1995 and 2018 and was analysed at genomic level and compared with ST23 isolated in other countries to determine possible similarities and differences. A core genome multi-locus sequence typing (cgMLST), based on a previously described set of 1,521 core genes, and single-nucleotide polymorphisms (SNPs) approaches were applied to an ST23 collection including genomes from Italy, France, Denmark and Scotland. DNAs were automatically extracted, libraries prepared using NextEra library kit and MiSeq sequencing performed. Overall, 63 among clinical and environmental Italian Lp1 isolates and a further seven and 11 ST23 from Denmark and Scotland, respectively, were sequenced, and pangenome analysed. Both cgMLST and SNPs analyses showed very few loci and SNP variations in ST23 genomes. All the ST23 causing outbreaks and sporadic cases in Italy and elsewhere, were phylogenetically related independent of year, town or country of isolation. Distances among the ST23s were further shortened when SNPs due to horizontal gene transfers were removed. The Lp1 ST23 isolated in Italy have kept their monophyletic origin, but they are phylogenetically close also to ST23 from other countries. The ST23 are quite widespread in Italy, and a thorough epidemiological investigation is compelled to determine sources of infection when this ST is identified in both LD sporadic cases and outbreaks.
- Published
- 2022
23. A Community Outbreak of Legionnaires' Disease with Two Strains of
- Author
-
Cyril, Rousseau, Christophe, Ginevra, Leslie, Simac, Noel, Fiard, Karine, Vilhes, Anne-Gaëlle, Ranc, Sophie, Jarraud, Hervé, Gornes, Damien, Mouly, and Christine, Campese
- Subjects
Aquatic Therapy ,Humans ,Legionnaires' Disease ,Serogroup ,Disease Outbreaks ,Legionella pneumophila - Abstract
An outbreak of Legionnaires' disease affected 18 people in Montpellier, a town of the south of France, between December 2016 and July 2017. All cases were diagnosed by a positive urinary antigen test. No deaths were reported. Epidemiological, environmental and genomic investigations (nested Sequence-Based Typing (nSBT) and whole genome sequencing) were undertaken. For the cases for which we had information, four had a new isolate (ST2471), one had a different new isolate (ST2470), one had a genomic pattern compatible with the ST2471 identified by nSBT (
- Published
- 2021
24. Bacterial Long-Range Warfare: Aerial Killing of Legionella pneumophila by Pseudomonas fluorescens
- Author
-
Christophe Ginevra, Mathilde Bouteiller, Laettitia Grac, David Giron, Annabelle Merieau, Meg Rouxel, Ségolène Depayras, Anne Mercier, Marie-Hélène Corre, Mathias Gallique, Julien Verdon, Jean-Marc Berjeaud, S. Jarraud, Alix Khalil, Charly Dupont, Ecologie et biologie des interactions (EBI), Université de Poitiers-Centre National de la Recherche Scientifique (CNRS), Microbiologie de l'Eau (MDE), Université de Poitiers-Centre National de la Recherche Scientifique (CNRS)-Université de Poitiers-Centre National de la Recherche Scientifique (CNRS), Institut de recherche sur la biologie de l'insecte UMR7261 (IRBI), Université de Tours (UT)-Centre National de la Recherche Scientifique (CNRS), and Université de Tours-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Microbiology (medical) ,Physiology ,Legionella ,medicine.drug_class ,[SDV]Life Sciences [q-bio] ,SPME ,Antibiotics ,Pseudomonas fluorescens ,Microbiology ,Legionella pneumophila ,03 medical and health sciences ,1-undecene ,antibacterial activity ,Pseudomonas ,Genetics ,medicine ,Humans ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,030304 developmental biology ,Volatile Organic Compounds ,0303 health sciences ,General Immunology and Microbiology ,Ecology ,biology ,030306 microbiology ,Chemistry ,Cell Biology ,Antimicrobial ,biology.organism_classification ,QR1-502 ,Anti-Bacterial Agents ,[SDV.MP]Life Sciences [q-bio]/Microbiology and Parasitology ,Infectious Diseases ,Legionnaires' Disease ,[SDE.BE]Environmental Sciences/Biodiversity and Ecology ,Legionellaceae ,Bacteria ,Research Article - Abstract
Legionella pneumophila, the causative agent of Legionnaires’ disease, is mostly found in man-made water systems and is one of the most closely monitored waterborne pathogens. With the aim of finding natural ways to control waterborne pathogens and thus further reduce the impact of disinfection by-products on human health, some studies have demonstrated the ability of bacteria to kill Legionella through the production of secondary metabolites or antimicrobial compounds. Here, we describe an unexpected growth inhibition of L. pneumophila when exposed to a physically separated strain of Pseudomonas fluorescens, designated as MFE01. Most of the members of the Legionellaceae family are sensitive to the volatile substances emitted by MFE01, unlike other bacteria tested. Using headspace solid-phase microextraction GC-MS strategy, a volatilome comparison revealed that emission of 1-undecene, 2-undecanone, and 2-tridecanone were mainly reduced in a Tn5-transposon mutant unable to inhibit at distance the growth of L. pneumophila strain Lens. We showed that 1-undecene was mainly responsible for the inhibition at distance in vitro, and led to cell lysis in small amounts, as determined by gas chromatography-mass spectrometry (GC-MS). Collectively, our results provide new insights into the mode of action of bacterial volatiles and highlight them as potent anti-Legionella agents to focus research on novel strategies to fight legionellosis. IMPORTANCE Microbial volatile compounds are molecules whose activities are increasingly attracting the attention of researchers. Indeed, they can act as key compounds in long-distance intrakingdom and interkingdom communication, but also as antimicrobials in competition and predation. In fact, most studies to date have focused on their antifungal activities and only a few have reported on their antibacterial properties. Here, we describe that 1-undecene, naturally produced by P. fluorescens, is a volatile with potent activity against bacteria of the genus Legionella. In small amounts, it is capable of inducing cell lysis even when the producing strain is physically separated from the target. This is the first time that such activity is described. This molecule could therefore constitute an efficient compound to counter bacterial pathogens whose treatment may fail, particularly in pulmonary diseases. Indeed, inhalation of these volatiles should be considered as a possible route of therapy in addition to antibiotic treatment.
- Published
- 2021
- Full Text
- View/download PDF
25. Transmission of Legionnaires’ Disease through Toilet Flushing
- Author
-
Laetitia Beraud, C. Campèse, Giorgia Battipaglia, Didier Nesa, Ghislaine Descours, Cyril Gouot, Jeanne Couturier, Christophe Ginevra, Sophie Jarraud, Anne-Gaëlle Ranc, Frédéric Barbut, Eolia Brissot, Marine Adam, Couturier, J., Ginevra, C., Nesa, D., Adam, M., Gouot, C., Descours, G., Campese, C., Battipaglia, G., Brissot, E., Beraud, L., Ranc, A. -G., Jarraud, S., and Barbut, F.
- Subjects
Microbiology (medical) ,Healthcare associated infections ,medicine.medical_specialty ,toilet ,pneumophilia ,Epidemiology ,030231 tropical medicine ,lcsh:Medicine ,phylogeny ,Legionella pneumophila ,Transmission of Legionnaires’ Disease through Toilet Flushing ,lcsh:Infectious and parasitic diseases ,respiratory infections ,03 medical and health sciences ,0302 clinical medicine ,fluids and secretions ,healthcare-associated infection ,respiratory infection ,Internal medicine ,nosocomial infections ,Humans ,Medicine ,In patient ,lcsh:RC109-216 ,030212 general & internal medicine ,Toilet ,Cross Infection ,biology ,Bacteria ,business.industry ,Transmission (medicine) ,lcsh:R ,Dispatch ,transmission ,biology.organism_classification ,medicine.disease ,digestive system diseases ,Infectious Diseases ,healthcare-associated infections ,nosocomial infection ,Bathroom Equipment ,Legionnaires' disease ,France ,Toilet flushing ,Legionnaires' Disease ,business ,Legionnaires’ disease - Abstract
We describe 2 cases of healthcare-associated Legionnaires’ disease in patients in France hospitalized 5 months apart in the same room. Whole-genome sequencing analyses showed that clinical isolates from the patients and isolates from the room’s toilet clustered together. Toilet contamination by Legionella pneumophila could lead to a risk for exposure through flushing.
- Published
- 2020
26. Diverse conjugative elements silence natural transformation in Legionella species
- Author
-
S. Jarraud, Pierre-Alexandre Juan, Kevin Picq, Isabelle Durieux, Laetitia Attaiech, Xavier Charpentier, Christophe Ginevra, Transfert de gène horizontal chez les bactéries pathogènes – Horizontal gene transfer in bacterial pathogens, Centre International de Recherche en Infectiologie - UMR (CIRI), Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS), Pathogenèse des légionelles- Legionella pathogenesis (LegioPath), Centre National de Référence des Légionelles (CNR), ANR-17-CE11-0009,RNAchap,Etude d'un nouveau type de chaperonne à ARN(2017), ANR-11-LABX-0048,ECOFECT,Dynamiques eco-évolutives des maladies infectieuses(2011), ANR-11-IDEX-0007,Avenir L.S.E.,PROJET AVENIR LYON SAINT-ETIENNE(2011), Attaiech, Laetitia, Etude d'un nouveau type de chaperonne à ARN - - RNAchap2017 - ANR-17-CE11-0009 - AAPG2017 - VALID, Dynamiques eco-évolutives des maladies infectieuses - - ECOFECT2011 - ANR-11-LABX-0048 - LABX - VALID, PROJET AVENIR LYON SAINT-ETIENNE - - Avenir L.S.E.2011 - ANR-11-IDEX-0007 - IDEX - VALID, École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Small RNA ,Gene Transfer, Horizontal ,Bacterial genome size ,[SDV.BBM.BM] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,Microbiology ,natural transformation ,Legionella pneumophila ,noncoding RNA ,03 medical and health sciences ,chemistry.chemical_compound ,Plasmid ,Bacterial Proteins ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Gene Silencing ,Gene ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Genetics ,0303 health sciences ,Multidisciplinary ,biology ,030306 microbiology ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,Biological Sciences ,biology.organism_classification ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,DNA-Binding Proteins ,Transformation (genetics) ,chemistry ,Horizontal gene transfer ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,horizontal gene transfer ,RNA ,RNA, Small Untranslated ,Transformation, Bacterial ,[SDV.MP.BAC] Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,Mobile genetic elements ,conjugative element ,DNA ,Genome-Wide Association Study ,Molecular Chaperones ,Plasmids - Abstract
Significance Natural transformation is a major mechanism of horizontal gene transfer. Although the genes required for natural transformation are nearly ubiquitous in bacteria, it is commonly reported that some isolates of transformable species fail to transform. In Legionella pneumophila, we show that the inability of multiple clinical isolates to transform is caused by a conjugative element that shuts down expression of genes required for transformation. Diverse conjugative elements in the Legionella genus have adopted the same inhibition strategy. We propose that inhibition of natural transformation by episomal and integrated conjugative elements can explain the lack of transformability of isolates and also the apparent lack of natural transformation in some species., Natural transformation (i.e., the uptake of DNA and its stable integration in the chromosome) is a major mechanism of horizontal gene transfer in bacteria. Although the vast majority of bacterial genomes carry the genes involved in natural transformation, close relatives of naturally transformable species often appear not competent for natural transformation. In addition, unexplained extensive variations in the natural transformation phenotype have been reported in several species. Here, we addressed this phenomenon by conducting a genome-wide association study (GWAS) on a panel of isolates of the opportunistic pathogen Legionella pneumophila. GWAS revealed that the absence of the transformation phenotype is associated with the conjugative plasmid pLPL. The plasmid inhibits transformation by simultaneously silencing the genes required for DNA uptake and recombination. We identified a small RNA (sRNA), RocRp, as the sole plasmid-encoded factor responsible for the silencing of natural transformation. RocRp is homologous to the highly conserved and chromosome-encoded sRNA RocR which controls the transient expression of the DNA uptake system. Assisted by the ProQ/FinO-domain RNA chaperone RocC, RocRp acts as a substitute of RocR, ensuring that the bacterial host of the conjugative plasmid does not become naturally transformable. Distinct homologs of this plasmid-encoded sRNA are found in diverse conjugative elements in other Legionella species. Their low to high prevalence may result in the lack of transformability of some isolates up to the apparent absence of natural transformation in the species. Generally, our work suggests that conjugative elements obscure the widespread occurrence of natural transformability in bacteria.
- Published
- 2019
- Full Text
- View/download PDF
27. Slowly or Nonresolving Legionnaires’ Disease: Case Series and Literature Review
- Author
-
Anne Bergeron-Lafaurie, Anne-Gaëlle Ranc, Nicolas Magand, Cécile Pouderoux, Florence Ader, Ghislaine Descours, Laetitia Beraud, Anne Conrad, S. Jarraud, Sandrine Boisset, and Christophe Ginevra
- Subjects
0301 basic medicine ,Microbiology (medical) ,medicine.medical_specialty ,medicine.drug_class ,Legionella ,030106 microbiology ,Antibiotics ,Lung abscess ,Real-Time Polymerase Chain Reaction ,Gastroenterology ,Legionella pneumophila ,03 medical and health sciences ,Interquartile range ,Internal medicine ,medicine ,Humans ,Retrospective Studies ,biology ,business.industry ,Retrospective cohort study ,medicine.disease ,biology.organism_classification ,Empyema ,Anti-Bacterial Agents ,030104 developmental biology ,Infectious Diseases ,Cardiothoracic surgery ,Legionnaires' disease ,Legionnaires' Disease ,business - Abstract
Background Rarely, Legionnaires’ disease (LD) can progress into a slowly or nonresolving form. Methods A nationwide retrospective study was conducted by the French National Reference Center for Legionella (2013–2017) including cases of slowly or nonresolving LD defined as persistent clinical symptoms, computed tomography (CT) scan abnormalities, and Legionella detection in lower respiratory tract specimens by culture and/or real-time (RT) polymerase chain reaction (PCR) >30 days after symptom onset. Results Twelve cases of community-acquired slowly or nonresolving LD were identified among 1686 cases of culture-positive LD. Median (interquartile range [IQR]) age was 63 (29–82) years. Ten (83.3%) patients had ≥1 immunosuppressive factor. Clinically, 9 patients transiently recovered before further deterioration (median [IQR] symptom-free interval, 30 [18–55] days), 3 patients had uniformly persistent symptoms (median [IQR] time, 48 [41.5–54] days). Two patients had >2 recurrences. CT scan imagery found lung abscess in 5 (41.6%) cases. Slowly or nonresolving LD was diagnosed on positive Legionella cultures (n = 10, 83.3%) at 49.5 (IQR, 33.7–79) days. Two cases were documented through positive Legionella RT PCR at 52 and 53 days (cycle threshold detection of 21.5 and 33.7, respectively). No genomic microevolution and no Legionella resistance to antibiotics were detected. The median (IQR) duration of treatment was 46.5 (21–92.5) days. Two empyema cases required thoracic surgery. At a median (IQR) follow-up of 26 (14–41.5) months, LD-attributable mortality was 16.6% (n = 2). Conclusions Slowly or nonresolving LD may occur in immunocompromised patients, possibly leading to lung abscess and empyema.
- Published
- 2019
- Full Text
- View/download PDF
28. Rifampicin exposure reveals within-host Mycobacterium tuberculosis diversity in patients with delayed culture conversion
- Author
-
Sylvain Goutelle, Gerard Lina, Charlotte Genestet, Alexia Barbry, Jonathan Hoffmann, Jean-Luc Berland, Christophe Ginevra, Emilie Westeel, Florence Ader, Elisabeth Hodille, Fabiola Bastian, Samuel Venner, Michel Guichardant, Oana Dumitrescu, Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut des Agents Infectieux [Lyon] (IAI), Hospices Civils de Lyon (HCL), Laboratoire des pathogènes émergents -- Emerging Pathogens Laboratory (LPE-Fondation Mérieux), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Plateforme DTAMB de biologie moléculaire, Cardiovasculaire, métabolisme, diabétologie et nutrition (CarMeN), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Hospices Civils de Lyon (HCL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Laboratoire de Biométrie et Biologie Evolutive - UMR 5558 (LBBE), Université de Lyon-Université de Lyon-Institut National de Recherche en Informatique et en Automatique (Inria)-VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS)-Centre National de la Recherche Scientifique (CNRS), Service des Maladies Infectieuses et Tropicales [Lyon], Hôpital de la Croix-Rousse [CHU - HCL], Hospices Civils de Lyon (HCL)-Hospices Civils de Lyon (HCL), ANR-11-LABX-0048,ECOFECT,Dynamiques eco-évolutives des maladies infectieuses(2011), ANR-11-IDEX-0007,Avenir L.S.E.,PROJET AVENIR LYON SAINT-ETIENNE(2011), ROSSI, Sabine, Dynamiques eco-évolutives des maladies infectieuses - - ECOFECT2011 - ANR-11-LABX-0048 - LABX - VALID, PROJET AVENIR LYON SAINT-ETIENNE - - Avenir L.S.E.2011 - ANR-11-IDEX-0007 - IDEX - VALID, Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL)
- Subjects
Bacterial Diseases ,Extensively Drug-Resistant Tuberculosis ,Antibiotics ,Apoptosis ,Drug resistance ,Pathology and Laboratory Medicine ,White Blood Cells ,Medical Conditions ,Drug tolerance ,Animal Cells ,Culture conversion ,Medicine and Health Sciences ,Biology (General) ,[SDV.MP.VIR] Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,0303 health sciences ,Cell Death ,Antimicrobials ,Drugs ,Bacterial Pathogens ,Actinobacteria ,Infectious Diseases ,Intracellular Pathogens ,[SDV.MP]Life Sciences [q-bio]/Microbiology and Parasitology ,Cell Processes ,Medical Microbiology ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Rifampin ,Cellular Types ,Pathogens ,medicine.drug ,Research Article ,Tuberculosis ,medicine.drug_class ,QH301-705.5 ,Immune Cells ,Immunology ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Microbiology ,Mycobacterium tuberculosis ,03 medical and health sciences ,Virology ,Microbial Control ,Drug Resistance, Bacterial ,Genetics ,medicine ,Humans ,[SDV.MP] Life Sciences [q-bio]/Microbiology and Parasitology ,Molecular Biology ,Antibiotics, Antitubercular ,Microbial Pathogens ,030304 developmental biology ,Pharmacology ,Blood Cells ,Bacteria ,030306 microbiology ,Macrophages ,Organisms ,Extensively drug-resistant tuberculosis ,Biology and Life Sciences ,Mycobacteria ,Cell Biology ,RC581-607 ,medicine.disease ,biology.organism_classification ,Tropical Diseases ,Parasitology ,Immunologic diseases. Allergy ,Rifampicin - Abstract
Mycobacterium tuberculosis (Mtb) genetic micro-diversity in clinical isolates may underline mycobacterial adaptation to tuberculosis (TB) infection and provide insights to anti-TB treatment response and emergence of resistance. Herein we followed within-host evolution of Mtb clinical isolates in two cohorts of TB patients, either with delayed Mtb culture conversion (> 2 months), or with fast culture conversion (< 2 months). We captured the genetic diversity of Mtb isolates obtained in each patient, by focusing on minor variants detected as unfixed single nucleotide polymorphisms (SNPs). To unmask antibiotic tolerant sub-populations, we exposed these isolates to rifampicin (RIF) prior to whole genome sequencing (WGS) analysis. Thanks to WGS, we detected at least 1 unfixed SNP within the Mtb isolates for 9/15 patients with delayed culture conversion, and non-synonymous (ns) SNPs for 8/15 patients. Furthermore, RIF exposure revealed 9 additional unfixed nsSNP from 6/15 isolates unlinked to drug resistance. By contrast, in the fast culture conversion cohort, RIF exposure only revealed 2 unfixed nsSNP from 2/20 patients. To better understand the dynamics of Mtb micro-diversity, we investigated the variant composition of a persistent Mtb clinical isolate before and after controlled stress experiments mimicking the course of TB disease. A minor variant, featuring a particular mycocerosates profile, became enriched during both RIF exposure and macrophage infection. The variant was associated with drug tolerance and intracellular persistence, consistent with the pharmacological modeling predicting increased risk of treatment failure. A thorough study of such variants not necessarily linked to canonical drug-resistance, but which are prone to promote anti-TB drug tolerance, may be crucial to prevent the subsequent emergence of resistance. Taken together, the present findings support the further exploration of Mtb micro-diversity as a promising tool to detect patients at risk of poorly responding to anti-TB treatment, ultimately allowing improved and personalized TB management., Author summary Tuberculosis (TB) is caused by Mycobacterium tuberculosis (Mtb), bacteria that are able to persist inside the patient for many months or years, thus requiring long antibiotic treatments. Here we focused on TB patients with delayed response to treatment and we performed genetic characterization of Mtb isolates to search for sub-populations that may tolerate anti-TB drugs. We found that Mtb cultured from 9/15 patients contained different sub-populations, and in vitro drug exposure revealed Mtb sub-populations in 6/15 isolates, none related to known drug-resistance mechanisms. By contrast, drug exposure revealed Mtb sup-populations in 2/20 isolates in the control cohort of patients with fast culture conversion. Furthermore, we characterized a Mtb variant isolated from a sub-population growing in the presence of rifampicin (RIF), a major anti-TB drug. We found that this variant featured a modified lipidic envelope, and that it was able to develop in the presence of RIF and inside human macrophage cells. We performed pharmacological modelling and found that this kind of variant may be related to a poor response to treatment. In conclusion, searching for particular Mtb sub-populations may help to detect patients at risk of treatment failure and provide additional guidance for TB management.
- Published
- 2021
- Full Text
- View/download PDF
29. Genome analysis of
- Author
-
Maria Luisa, Ricci, Silvia, Fillo, Andrea, Ciammaruconi, Florigio, Lista, Christophe, Ginevra, Sophie, Jarraud, Antonietta, Girolamo, Fabrizio, Barbanti, Maria Cristina, Rota, Diane, Lindsay, Jamie, Gorzynski, Søren A, Uldum, Sharmin, Baig, Marina, Foti, Giancarlo, Petralito, Stefania, Torri, Marino, Faccini, Maira, Bonini, Gabriella, Gentili, Sabrina, Senatore, Anna, Lamberti, Joao André, Carrico, and Maria, Scaturro
- Subjects
Humans ,Legionnaires' Disease ,Serogroup ,Disease Outbreaks ,Legionella pneumophila ,Multilocus Sequence Typing - Published
- 2021
30. Evaluation of NGS-based approaches for SARS-CoV-2 whole genome characterisation
- Author
-
Caroline Charre, Christophe Ginevra, Marina Sabatier, Hadrien Regue, Grégory Destras, Solenne Brun, Gwendolyne Burfin, Caroline Scholtes, Florence Morfin, Martine Valette, Bruno Lina, Antonin Bal, and Laurence Josset
- Subjects
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,AcademicSubjects/SCI01130 ,AcademicSubjects/SCI02285 ,Genomics ,Computational biology ,Biology ,Genome ,Microbiology ,DNA sequencing ,Metagenomics ,Virology ,Respiratory virus ,AcademicSubjects/MED00860 ,Nanopore sequencing ,Illumina dye sequencing ,Rapid Communication - Abstract
Since the beginning of the COVID-19 outbreak, SARS-CoV-2 whole-genome sequencing (WGS) has been performed at unprecedented rate worldwide with the use of very diverse Next Generation Sequencing (NGS) methods. Herein, we compare the performance of four NGS-based approaches for SARS-CoV-2 WGS. Twenty four clinical respiratory samples with a large scale of Ct values (from 10.7 to 33.9) were sequenced with four methods. Three used Illumina sequencing: an in-house metagenomic NGS (mNGS) protocol and two newly commercialized kits including a hybridization capture method developed by Illumina (DNA Prep with Enrichment kit and Respiratory Virus Oligo Panel, RVOP) and an amplicon sequencing method developed by Paragon Genomics (CleanPlex SARS-CoV-2 kit). We also evaluated the widely used amplicon sequencing protocol developed by ARTIC Network and combined with Oxford Nanopore Technologies (ONT) sequencing. All four methods yielded near-complete genomes (>99%) for high viral loads samples, with mNGS and RVOP producing the most complete genomes. For mid viral loads, 2/8 and 1/8 genomes were incomplete (vsthe other methods, due to the dedicated bioinformatics pipeline setting a high threshold to call SNP compared to reference sequence. Importantly, all methods correctly identified a newly observed 34-nt deletion in ORF6 but required specific bioinformatic validation for RVOP. Finally, as a major warning for targeted techniques, a default of coverage in any given region of the genome should alert to a potential rearrangement or a SNP in primer annealing or probe-hybridizing regions and would require regular updates of the technique according to SARS-CoV-2 evolution.
- Published
- 2020
31. Persistent Legionnaires' Disease and Associated Antibiotic Treatment Engender a Highly Disturbed Pulmonary Microbiome Enriched in Opportunistic Microorganisms
- Author
-
Christophe Rusniok, Christophe Ginevra, Ana Elena Pérez-Cobas, Sophie Jarraud, Carmen Buchrieser, Biologie des Bactéries intracellulaires - Biology of Intracellular Bacteria, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Pathogenèse des légionelles- Legionella pathogenesis (LegioPath), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre National de Référence des Légionelles (CNR), This work was supported by the Institut Pasteur and the Agence National de Recherche (grant number ANR-15-CE17-0014-03 and grant number ANR-10-LABX-62IBEID to C.B. and A.E.P.-C.). Work in the S.J. laboratory is financed by Santé Publique France and the Agence National de Recherche (grant number ANR-15-CE17-0014-01). C.G. and S.J. are part of the French Laboratory of Excellence project ECOFECT (grant number ANR-11-LABX-0048)., We thank the Institut Pasteur Biomics Pole (Christiane Bouchier and Laurence Ma) for sequencing of the samples reported in this study., ANR-15-CE17-0014,ProgLegio,Biomarqueurs bactériens et humains d'intérêt pronostic pour les légionelloses sévères(2015), ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-11-LABX-0048,ECOFECT,Dynamiques eco-évolutives des maladies infectieuses(2011), Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Adult ,Male ,Lung microbiome ,antibiotic resistance ,Drug Resistance ,Legionella Pneumonia ,Opportunistic Infections ,Microbiology ,Legionella pneumophila ,pulmonary microbiome ,Host-Microbe Biology ,03 medical and health sciences ,Antibiotic resistance ,Virology ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Pneumonia, Bacterial ,medicine ,Humans ,pneumonia ,Longitudinal Studies ,Microbiome ,Lung ,Pathogen ,Aged ,030304 developmental biology ,0303 health sciences ,Bacteria ,Whole Genome Sequencing ,biology ,030306 microbiology ,Microbiota ,Fungi ,Eukaryota ,High-Throughput Nucleotide Sequencing ,Genomics ,biology.organism_classification ,medicine.disease ,QR1-502 ,Anti-Bacterial Agents ,3. Good health ,respiratory tract diseases ,Female ,Legionnaires' disease ,Legionnaires' Disease ,Bronchoalveolar Lavage Fluid ,Research Article - Abstract
The composition and dynamics of the lung microbiome during pneumonia are not known, although the lung microbiome might influence the severity and outcome of this infectious disease, similar to what was shown for the microbiome at other body sites. Here we report the findings of a comprehensive analysis of the lung microbiome composition of three patients with long-term pneumonia due to L. pneumophila and its evolution during antibiotic treatment. This work adds to our understanding of how the microbiome changes during disease and antibiotic treatment and points to microorganisms and their interactions that might be beneficial. In addition to bacteria and fungi, our analyses included archaea and eukaryotes (protozoa), showing that both are present in the pulmonary microbiota and that they might also play a role in the response to the microbiome disturbance., Despite the importance of pneumonia to public health, little is known about the composition of the lung microbiome during infectious diseases, such as pneumonia, and how it evolves during antibiotic therapy. To study the possible relation of the pulmonary microbiome to the severity and outcome of this respiratory disease, we analyzed the dynamics of the pathogen and the human lung microbiome during persistent infections caused by the bacterium Legionella pneumophila and their evolution during antimicrobial treatment. We collected 10 bronchoalveolar lavage fluid samples from three patients during long-term hospitalization due to pneumonia and performed a unique longitudinal study of the interkingdom microbiome, analyzing the samples for presence of bacteria, archaea, fungi, and protozoa by high-throughput Illumina sequencing of marker genes. The lung microbiome of the patients was characterized by a strong predominance of the pathogen, a low diversity of the bacterial fraction, and an increased presence of opportunistic microorganisms. The fungal fraction was more stable than the bacterial fraction. During long-term treatment, no genomic changes or antibiotic resistance-associated mutations that could explain the persistent infection occurred, according to whole-genome sequencing analyses of the pathogen. After antibiotic treatment, the microbiome did not recover rapidly but was mainly constituted of antibiotic-resistant species and enriched in bacteria, archaea, fungi, or protozoa associated with pathogenicity. The lung microbiome seems to contribute to nonresolving Legionella pneumonia, as it is strongly disturbed during infection and enriched in opportunistic and/or antibiotic-resistant bacteria and microorganisms, including fungi, archaea, and protozoa that are often associated with infections.
- Published
- 2020
- Full Text
- View/download PDF
32. A Community Outbreak of Legionnaires’ Disease with Two Strains of L. pneumophila Serogroup 1 Linked to an Aquatic Therapy Centre
- Author
-
Cyril Rousseau, Christophe Ginevra, Leslie Simac, Noel Fiard, Karine Vilhes, Anne-Gaëlle Ranc, Sophie Jarraud, Hervé Gornes, Damien Mouly, and Christine Campese
- Subjects
outbreak ,Legionella pneumophila ,aquatic therapy centre ,sequence typing ,Health, Toxicology and Mutagenesis ,Public Health, Environmental and Occupational Health ,Medicine - Abstract
An outbreak of Legionnaires’ disease affected 18 people in Montpellier, a town of the south of France, between December 2016 and July 2017. All cases were diagnosed by a positive urinary antigen test. No deaths were reported. Epidemiological, environmental and genomic investigations (nested Sequence-Based Typing (nSBT) and whole genome sequencing) were undertaken. For the cases for which we had information, four had a new isolate (ST2471), one had a different new isolate (ST2470), one had a genomic pattern compatible with the ST2471 identified by nSBT (flaA = 3), and one had a genomic pattern not compatible with two previous identified STs (pilE = 6). The analysis conducted on the pool of an aquatic therapy center revealed seven isolates of Legionella pneumophila. Whole genome analysis confirmed the link between the environmental and clinical isolates for both ST2470 and ST2471. As the outbreak occurred slowly, with several weeks between new cases, it was not possible to immediately identify a common source. The sixth case was the first to report having aquatic therapy care. Of the 18 cases, eight had attended the aquatic therapy center and the other 10 were inhabitants who lived, worked or walked close to the center. The main cause for this outbreak was the lack of facility maintenance. This investigation highlights the risk to public health of aquatic therapy centers for users and nearby populations, and emphasizes the need for risk reduction measures with specific guidelines to improve health and safety in aquatic facilities.
- Published
- 2022
- Full Text
- View/download PDF
33. Seeding and Establishment of Legionella pneumophila in Hospitals: Implications for Genomic Investigations of Nosocomial Legionnaires’ Disease
- Author
-
Isabelle Podglajen, S. Jarraud, M. Mentasti, Simon R. Harris, Timothy G. Harrison, Julian Parkhill, Baharak Afshar, Victoria J. Chalker, Sophia David, Christophe Ginevra, The Wellcome Trust Sanger Institute [Cambridge], Public Health England [London], European Programme for Public Health Microbiology Training (EUPHEM), European Centre for Disease Prevention and Control (ECDC), Pathogenèse des légionelles- Legionella pathogenesis (LegioPath), Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre National de Référence Legionella, Service de Microbiologie [CHU GeorgesPompidou], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Parkhill, Julian [0000-0002-7069-5958], Apollo - University of Cambridge Repository, European Centre for Disease Prevention and Control [Stockholm, Sweden] (ECDC), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,bacterial genomics ,Diving ,Disease ,Bioinformatics ,Legionella pneumophila ,Genotype ,Phylogeny ,education.field_of_study ,Molecular Epidemiology ,Cross Infection ,biology ,Genomics ,Hospitals ,3. Good health ,Infectious Diseases ,whole-genome sequencing ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,nosocomial infections ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Legionnaires' disease ,Legionnaires\textquoteright disease ,Legionnaires' Disease ,Water Microbiology ,Pneumonia (non-human) ,Sequence Analysis ,Legionnaires’ disease ,Microbiology (medical) ,Legionella ,030106 microbiology ,Population ,Microbiology ,03 medical and health sciences ,medicine ,Major Article ,Humans ,education ,Molecular epidemiology ,business.industry ,Computational Biology ,Sequence Analysis, DNA ,DNA ,biology.organism_classification ,medicine.disease ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,respiratory tract diseases ,Molecular Typing ,Editor's Choice ,business - Abstract
Summary Whole-genome sequencing can be used to support or refute suspected links between hospital water systems and Legionnaires’ disease cases. However, caveats regarding the interpretation of genomic data from Legionella pneumophila are described that should be considered in future investigations., Background. Legionnaires’ disease is an important cause of hospital-acquired pneumonia and is caused by infection with the bacterium Legionella. Because current typing methods often fail to resolve the infection source in possible nosocomial cases, we aimed to determine whether whole-genome sequencing (WGS) could be used to support or refute suspected links between cases and hospitals. We focused on cases involving a major nosocomial-associated strain, L. pneumophila sequence type (ST) 1. Methods. WGS data from 229 L. pneumophila ST1 isolates were analyzed, including 99 isolates from the water systems of 17 hospitals and 42 clinical isolates from patients with confirmed or suspected hospital-acquired infections, as well as isolates obtained from or associated with community-acquired sources of Legionnaires’ disease. Results. Phylogenetic analysis demonstrated that all hospitals from which multiple isolates were obtained have been colonized by 1 or more distinct ST1 populations. However, deep sampling of 1 hospital also revealed the existence of substantial diversity and ward-specific microevolution within the population. Across all hospitals, suspected links with cases were supported with WGS, although the degree of support was dependent on the depth of environmental sampling and available contextual information. Finally, phylogeographic analysis revealed that hospitals have been seeded with L. pneumophila via both local and international spread of ST1. Conclusions. WGS can be used to support or refute suspected links between hospitals and Legionnaires’ disease cases. However, deep hospital sampling is frequently required due to the potential coexistence of multiple populations, existence of substantial diversity, and similarity of hospital isolates to local populations.
- Published
- 2017
- Full Text
- View/download PDF
34. Legionella pneumophila infection and antibiotic treatment engenders a highly disturbed pulmonary microbiome with decreased microbial diversity
- Author
-
Christophe Rusniok, S. Jarraud, Ana Elena Pérez-Cobas, Carmen Buchrieser, and Christophe Ginevra
- Subjects
0303 health sciences ,Lung microbiome ,medicine.diagnostic_test ,biology ,030306 microbiology ,Firmicutes ,Bacterial pneumonia ,biology.organism_classification ,medicine.disease ,Legionella pneumophila ,3. Good health ,Microbiology ,respiratory tract diseases ,03 medical and health sciences ,Bronchoalveolar lavage ,medicine ,Microbiome ,Pathogen ,Bacteria ,030304 developmental biology - Abstract
BackgroundLung microbiome analyses have shown that the healthy lung is not sterile but it is colonized like other body sites by bacteria, fungi and viruses. However, little is known about the microbial composition of the lung microbiome during infectious diseases such as pneumonia and how it evolves during antibiotic therapy. To better understand the impact of the composition of the pulmonary microbiome on severity and outcome of pneumonia we analysed the composition and evolution of the human lung microbiome during pneumonia caused by the bacteriumLegionella pneumophila.ResultsWe collected 10 bronchoalveolar lavage (BAL) samples from three patients during long-term hospitalisation due to severe pneumonia and performed a longitudinal in-depth study of the composition of their lung microbiome by high-throughput Illumina sequencing of the 16S rRNA gene (bacteria and archaea), ITS region (fungi) and 18S rRNA gene (eukaryotes). We found that the composition of the bacterial lung microbiome during pneumonia is hugely disturbed containing a very high percentage of the pathogen, a very low bacterial diversity, and an increased presence of opportunistic microorganisms such as species belonging to Staphylococcaceae and Streptococcaceae. The microbiome of antibiotic treated patients cured from pneumonia represented a different perturbation state with a higher abundance of resistant bacteria (mainly Firmicutes) and a significantly different bacterial composition as that found in healthy individuals. In contrast, the mycobiome remains more stable during pneumonia and antimicrobial therapy. Interestingly we identified possible cooperation within and between both communities. Furthermore, archaea (Methanobrevibacter) and protozoa (AcanthamoebaandTrichomonas) were detected.ConclusionsBacterial pneumonia leads to a collapse of the healthy microbiome and a strongly disturbed bacterial composition of the pulmonary microbiome that is dominated by the pathogen. Antibiotic treatment allows some bacteria to regrow or recolonize the lungs but the restoration of a healthy lung microbiome composition is only regained a certain time after the antibiotic treatment. Archaea and protozoa should also be considered, as they might be important but yet overseen members of the lung microbiome. Interactions between the micro- and the mycobiome might play a role in the restoration of the microbiome and the clinical evolution of the disease.
- Published
- 2019
- Full Text
- View/download PDF
35. Subcultured Mycobacterium tuberculosis isolates on different growth media are fully representative of bacteria within clinical samples
- Author
-
Florence Ader, Juliet E. Bryant, Gerard Lina, Christophe Ginevra, Jean-Luc Berland, Elisabeth Hodille, Emilie Westeel, Charlotte Genestet, Samuel Venner, and Oana Dumitrescu
- Subjects
0301 basic medicine ,Microbiology (medical) ,DNA, Bacterial ,Tuberculosis ,030106 microbiology ,Immunology ,Single-nucleotide polymorphism ,Biology ,Microbiology ,Polymorphism, Single Nucleotide ,Mycobacterium tuberculosis ,03 medical and health sciences ,Tuberculosis diagnosis ,Predictive Value of Tests ,Genetic variation ,medicine ,Humans ,Tuberculosis, Pulmonary ,Whole genome sequencing ,Genetics ,Bacteriological Techniques ,Whole Genome Sequencing ,Sputum ,Reproducibility of Results ,biology.organism_classification ,medicine.disease ,3. Good health ,030104 developmental biology ,Infectious Diseases ,Subculture (biology) ,Bacteria - Abstract
Mycobacterium tuberculosis (Mtb) whole genome sequencing (WGS) plays an increasingly important role in tuberculosis diagnosis and research. WGS is typically performed on biobanked isolates obtained by subculture during diagnosis. Genetic variation upon culturing is known to occur in other bacterial species. However, little is understood regarding the impact of different subculture media on genome-wide diversity and variant selection in Mtb. Here we compared WGS derived from direct sequencing of sputa samples to WGS sequences from isolates subcultured on 3 different media. Based on analysis of single nucleotide polymorphisms (SNPs), there was no evidence of variant selection caused by the different culture media used, indicating that subcultured clinical strains can be reliably used to explore genetic determinants of Mtb pathogenesis and epidemiological features.
- Published
- 2019
36. Whole-genome sequencing in drug susceptibility testing of Mycobacterium tuberculosis in routine practice in Lyon, France
- Author
-
Gerard Lina, Jean-Luc Berland, Elisabeth Hodille, Juliet E. Bryant, Oana Dumitrescu, Charlotte Genestet, Christophe Ginevra, Florence Ader, Centre International de Recherche en Infectiologie - UMR (CIRI), Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Microbiology (medical) ,Tuberculosis ,[SDV]Life Sciences [q-bio] ,030106 microbiology ,Antitubercular Agents ,Microbial Sensitivity Tests ,Drug resistance ,Mycobacterium tuberculosis ,03 medical and health sciences ,0302 clinical medicine ,Bacterial Proteins ,Drug Resistance, Multiple, Bacterial ,Tuberculosis, Multidrug-Resistant ,Isoniazid ,medicine ,Humans ,Pharmacology (medical) ,Mycobacteria growth indicator tube ,Pentosyltransferases ,030212 general & internal medicine ,Tuberculosis, Pulmonary ,Ethambutol ,Whole Genome Sequencing ,biology ,DNA-Directed RNA Polymerases ,General Medicine ,Pyrazinamide ,bacterial infections and mycoses ,biology.organism_classification ,medicine.disease ,Virology ,3. Good health ,Infectious Diseases ,France ,Rifampin ,Rifampicin ,medicine.drug - Abstract
Rapid and correct determination of Mycobacterium tuberculosis (MTB) drug susceptibility is a challenge for tuberculosis (TB) management. Phenotypic drug susceptibility testing (DST) remains the reference method but is time consuming. In this study, genotypic prediction of the first-line drug susceptibility profile obtained by whole-genome sequencing (WGS) was compared with that obtained by phenotypic DST and the line probe assay (LPA). All MTB strains isolated from patients during routine practice at the mycobacteria laboratory of Lyon University Hospital, France, between November 2016 and July 2019 were included (n = 274). Isolates were tested for the first-line drugs using phenotypic DST (Mycobacteria Growth Indicator Tube) and for genotypic prediction of the susceptibility profile with LPA and WGS. Considering phenotypic DST as the reference, WGS predicted resistance to rifampicin, isoniazid, ethambutol and pyrazinamide with sensitivities of 100%, 100%, 100% and 93.8%, respectively, and susceptibility to these drugs with specificities of 99.6%, 100%, 98.5% and 100%, respectively. Performance of the LPA was poorer, with sensitivity of 83.3% for rifampicin and 85.7% for isoniazid resistance. Five isolates were classified as susceptible according to phenotypic DST (1 for rifampicin, 4 for ethambutol) while WGS detected resistance mutations in rpoB and embB genes. WGS, used under appropriate quality-control conditions, has good performance to predict the resistance profile for the four first-line drugs and can correct phenotypic DST results. This study highlights the need for future guidelines recommending WGS as the initial tool in routine practice in areas where the prevalences of TB and drug-resistant MTB are low.
- Published
- 2020
- Full Text
- View/download PDF
37. Polymorphisms of a Collagen-Like Adhesin Contributes to
- Author
-
Mena, Abdel-Nour, Han, Su, Carla, Duncan, Shaopei, Li, Deepa, Raju, Feras, Shamoun, Marine, Valton, Christophe, Ginevra, Sophie, Jarraud, Cyril, Guyard, Kagan, Kerman, and Mauricio R, Terebiznik
- Subjects
stomatognathic diseases ,adhesion ,collagen-like adhesion ,hemic and lymphatic diseases ,otorhinolaryngologic diseases ,cell–cell adhesion ,Microbiology ,biofilm ,Original Research ,Legionella pneumophila ,clinical prevalence - Abstract
Legionellosis is a severe respiratory illness caused by the inhalation of aerosolized water droplets contaminated with the opportunistic pathogen Legionella pneumophila. The ability of L. pneumophila to produce biofilms has been associated with its capacity to colonize and persist in human-made water reservoirs and distribution systems, which are the source of legionellosis outbreaks. Nevertheless, the factors that mediate L. pneumophila biofilm formation are largely unknown. In previous studies we reported that the adhesin Legionella collagen-like protein (Lcl), is required for auto-aggregation, attachment to multiple surfaces and the formation of biofilms. Lcl structure contains three distinguishable regions: An N-terminal region with a predicted signal sequence, a central region containing tandem collagen-like repeats (R-domain) and a C-terminal region (C-domain) with no significant homology to other known proteins. Lcl R-domain encodes tandem repeats of the collagenous tripeptide Gly-Xaa-Yaa (GXY), a motif that is key for the molecular organization of mammalian collagen and mediates the binding of collagenous proteins to different cellular and environmental ligands. Interestingly, Lcl is polymorphic in the number of GXY tandem repeats. In this study, we combined diverse biochemical, genetic, and cellular approaches to determine the role of Lcl domains and GXY repeats polymorphisms on the structural and functional properties of Lcl, as well as on bacterial attachment, aggregation and biofilm formation. Our results indicate that the R-domain is key for assembling Lcl collagenous triple-helices and has a more preponderate role over the C-domain in Lcl adhesin binding properties. We show that Lcl molecules oligomerize to form large supramolecular complexes to which both, R and C-domains are required. Furthermore, we found that the number of GXY tandem repeats encoded in Lcl R-domain correlates positively with the binding capabilities of Lcl and with the attachment and biofilm production capacity of L. pneumophila strains. Accordingly, the number of GXY tandem repeats in Lcl influences the clinical prevalence of L. pneumophila strains. Therefore, the number of Lcl tandem repeats could be considered as a potential predictor for virulence in L. pneumophila isolates.
- Published
- 2018
38. Detection of Legionella anisa in Water from Hospital Dental Chair Units and Molecular Characterization by Whole-Genome Sequencing
- Author
-
Alexander W. Friedrich, Natacha Couto, Mariëtte Lokate, Ruud H. Deurenberg, John W. A. Rossen, Sophie Jarraud, Giuseppe Fleres, Christophe Ginevra, Luc W M van der Sluis, Silvia García-Cobos, University Medical Center Groningen [Groningen] (UMCG), Pathogenèse des légionelles- Legionella pathogenesis (LegioPath), Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Personalized Healthcare Technology (PHT), Microbes in Health and Disease (MHD), Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Microbiology (medical) ,Legionella ,030106 microbiology ,whole-genome multi-locus sequence typing (wgMLST) ,Microbiology ,whole-genome sequencing (WGS) ,ANNOTATION ,03 medical and health sciences ,PNEUMOPHILA ,Tap water ,core-genome multi-locus sequence typing (cgMLST) ,Virology ,QUALITY ,Typing ,next-generation sequencing (NGS) ,lcsh:QH301-705.5 ,Legionella spp ,Whole genome sequencing ,biology ,Molecular epidemiology ,biology.organism_classification ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,3. Good health ,Legionella anisa ,030104 developmental biology ,lcsh:Biology (General) ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Dental chair ,RESISTANCE - Abstract
This study aims to assess contamination with Legionella spp. in water from dental chair units (DCUs) of a hospital dental ward and to perform its molecular characterization by whole-genome sequencing (WGS). We collect eight water samples (250 mL) from four DCUs (sink and water-syringe). Samples are tested for the presence of Legionella spp. (CFUs/mL) by culturing according to the Nederland Norm (NEN) 6265. Three DCUs are found positive for Legionella anisa, and four isolates are cultured (sink n = 2, water-syringe n = 1, two isolates from the same chair) with 1 ×, 102 CFU/mL. Whole-genome multi-locus sequence typing (wgMLST) results indicate that all strains belong to the same cluster with two to four allele differences. Classical culture combined with WGS allows the identification of a unique clone of L. anisa in several DCUs in the same hospital dental ward. This may indicate a common contamination source in the dental unit waterlines, which was fixed by replacing the chairs and main pipeline of the unit. Our results reveal tap water contamination in direct contact with patients and the usefulness of WGS to investigate bacterial molecular epidemiology.
- Published
- 2018
- Full Text
- View/download PDF
39. Detection of
- Author
-
Giuseppe, Fleres, Natacha, Couto, Mariette, Lokate, Luc W M, van der Sluis, Christophe, Ginevra, Sophie, Jarraud, Ruud H, Deurenberg, John W, Rossen, Silvia, García-Cobos, and Alex W, Friedrich
- Subjects
Legionella spp ,core-genome multi-locus sequence typing (cgMLST) ,whole-genome multi-locus sequence typing (wgMLST) ,next-generation sequencing (NGS) ,whole-genome sequencing (WGS) ,Article - Abstract
This study aims to assess contamination with Legionella spp. in water from dental chair units (DCUs) of a hospital dental ward and to perform its molecular characterization by whole-genome sequencing (WGS). We collect eight water samples (250 mL) from four DCUs (sink and water-syringe). Samples are tested for the presence of Legionella spp. (CFUs/mL) by culturing according to the Nederland Norm (NEN) 6265. Three DCUs are found positive for Legionella anisa, and four isolates are cultured (sink n = 2, water-syringe n = 1; two isolates from the same chair) with 1 × 102 CFU/mL. Whole-genome multi-locus sequence typing (wgMLST) results indicate that all strains belong to the same cluster with two to four allele differences. Classical culture combined with WGS allows the identification of a unique clone of L. anisa in several DCUs in the same hospital dental ward. This may indicate a common contamination source in the dental unit waterlines, which was fixed by replacing the chairs and main pipeline of the unit. Our results reveal tap water contamination in direct contact with patients and the usefulness of WGS to investigate bacterial molecular epidemiology.
- Published
- 2018
40. Minimum inhibitory concentration (MIC) distribution among wild-type strains of Legionella pneumophila identifies a subpopulation with reduced susceptibility to macrolides owing to efflux pump genes
- Author
-
Max Maurin, Marine Vandewalle-Capo, Pierre Alain Billy, Christophe Gilbert, Clémence Massip, Christophe Ginevra, Gerard Lina, S. Jarraud, Joelle Chastang, Joséphine Charavit, Sandrine Boisset, Ghislaine Descours, Pathogenèse des légionelles- Legionella pathogenesis (LegioPath), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre National de Référence des Légionelles (CNR), Thérapeutique Recombinante Expérimentale (TIMC-IMAG-TheREx), Techniques de l'Ingénierie Médicale et de la Complexité - Informatique, Mathématiques et Applications, Grenoble - UMR 5525 (TIMC-IMAG), Institut polytechnique de Grenoble - Grenoble Institute of Technology (Grenoble INP )-VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS)-Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes [2016-2019] (UGA [2016-2019])-Institut polytechnique de Grenoble - Grenoble Institute of Technology (Grenoble INP )-VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS)-Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes [2016-2019] (UGA [2016-2019]), Pathogénie des Staphylocoques – Staphylococcal Pathogenesis (StaPath), Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Microbiology (medical) ,medicine.drug_class ,030106 microbiology ,Antibiotics ,Drug Resistance ,Microbial Sensitivity Tests ,Azithromycin ,Legionella pneumophila ,Microbiology ,Macrolide Antibiotics ,03 medical and health sciences ,Minimum inhibitory concentration ,Drug Resistance, Bacterial ,medicine ,Humans ,Pharmacology (medical) ,Antiinfective agent ,biology ,Efflux pump ,Broth microdilution ,Bacterial ,LpeAB ,General Medicine ,biology.organism_classification ,medicine.disease ,bacterial infections and mycoses ,Virology ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,3. Good health ,Anti-Bacterial Agents ,Infectious Diseases ,Genes ,Genes, Bacterial ,Susceptibility ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Legionnaires' disease ,Efflux ,Macrolides ,Legionnaires' Disease ,Rifampin ,Fluoroquinolones - Abstract
International audience; Legionnaires' disease is a severe pneumonia mainly caused by Legionella pneumophila that is treated by antibiotics. The purpose of this study was to describe the susceptibility of clinical strains of L. pneumophila to eight antibiotics used for treatment of legionellosis. The minimum inhibitory concentrations (MICs) of 109 well-characterised clinical strains of L. pneumophila serogroup 1 were determined by the broth microdilution method without charcoal and were compared with antibiotic-resistant strains selected in vitro. All strains were inhibited by low concentrations of fluoroquinolones, macrolides and rifampicin. The epidemiological cut-off values (ECOFFs) were 0.064 mg/L for ciprofloxacin, 0.064 mg/L for moxifloxacin, 0.032 mg/L for levofloxacin, 1 mg/L for erythromycin, 2 mg/L for azithromycin, 0.064 mg/L for clarithromycin, 2 mg/L for doxycycline and 0.001 mg/L for rifampicin. However, MIC distributions revealed a subpopulation of strains displaying reduced susceptibility to some macrolides (especially azithromycin), which correlated with the presence of the lpeAB genes encoding a macrolide efflux pump found specifically in sequence type (ST) ST1, ST701 and closely related STs. Thus, all isolates could be considered susceptible to the tested antibiotics, although macrolides were less active against some strains harbouring a specific efflux system.
- Published
- 2017
- Full Text
- View/download PDF
41. Legionella pneumophila LPS to evaluate urinary antigen tests
- Author
-
Margot Carpentier, Anne-Gaëlle Ranc, Gerard Lina, Elodie Maisonneuve, Ghislaine Descours, Laetitia Beraud, Jean-Marc Berjeaud, Sophie Jarraud, Julien Verdon, Christophe Ginevra, Centre National de Référence des Légionelles (CNR), Pathogenèse des légionelles- Legionella pathogenesis (LegioPath), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Microbiologie de l'Eau (MDE), Ecologie et biologie des interactions (EBI), Université de Poitiers-Centre National de la Recherche Scientifique (CNRS)-Université de Poitiers-Centre National de la Recherche Scientifique (CNRS), Université de Poitiers-Centre National de la Recherche Scientifique (CNRS), Pathogénie des Staphylocoques – Staphylococcal Pathogenesis (StaPath), Centre International de Recherche en Infectiologie - UMR (CIRI), Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Microbiology (medical) ,congenital, hereditary, and neonatal diseases and abnormalities ,LPS ,Urinary system ,[SDV]Life Sciences [q-bio] ,030106 microbiology ,Fluorescent Antibody Technique ,Sensitivity and Specificity ,Legionella pneumophila ,Microbiology ,Immunoenzyme Techniques ,03 medical and health sciences ,Antigen ,Limit of Detection ,Humans ,[SDV.EE]Life Sciences [q-bio]/Ecology, environment ,Antigens, Bacterial ,biology ,mAb3/1 ,General Medicine ,Urinary antigen ,biology.organism_classification ,bacterial infections and mycoses ,Virology ,eye diseases ,Pontiac ,Infectious Diseases ,[SDV.MP]Life Sciences [q-bio]/Microbiology and Parasitology ,Reagent Kits, Diagnostic ,Legionnaires' Disease - Abstract
International audience; Three urinary antigen tests were compared using purified Legionella pneumophila (Lp) LPS. For Lp serogroup1, Sofia®FIA and Binax®EIA limits of detection (LOD) were similar; that of BinaxNOW® lower. For all tests the LOD was higher with LPS from non-Pontiac compared to Pontiac-strains. The LOD was variable for other Lp serogroups.
- Published
- 2017
- Full Text
- View/download PDF
42. Rapid detection and evolutionary analysis of Legionella pneumophila serogroup 1 sequence type 47
- Author
-
Laura Gomez-Valero, M. Mentasti, Carmen Buchrieser, Baharak Afshar, Sophia David, Timothy G. Harrison, S. Jarraud, P. Cassier, Christophe Ginevra, Anthony Underwood, Vicki Chalker, Jerome Etienne, Julian Parkhill, Public Health England [London], Centre National de Référence Legionella, The Wellcome Trust Sanger Institute [Cambridge], Pathogenèse des légionelles- Legionella pathogenesis (LegioPath), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Biologie des Bactéries intracellulaires - Biology of Intracellular Bacteria, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), European Programme for Public Health Microbiology Training (EUPHEM), European Centre for Disease Prevention and Control [Stockholm, Sweden] (ECDC), Pathogénie des Staphylocoques – Staphylococcal Pathogenesis (StaPath), Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), and European Centre for Disease Prevention and Control (ECDC)
- Subjects
0301 basic medicine ,Microbiology (medical) ,Legionella ,Evolution ,030106 microbiology ,Biology ,Real-Time Polymerase Chain Reaction ,Serogroup ,Genome ,Legionella pneumophila ,Polymorphism, Single Nucleotide ,Sensitivity and Specificity ,Evolution, Molecular ,03 medical and health sciences ,Genetic ,Phylogenetics ,ST47 ,Humans ,Typing ,Legionella pneumophila Serogroup 1 ,Polymorphism ,Phylogeny ,ST62 ,Comparative genomics ,Genetics ,Recombination, Genetic ,Phylogenetic tree ,Bacterial ,Molecular ,General Medicine ,Sequence Analysis, DNA ,Single Nucleotide ,DNA ,biology.organism_classification ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,Recombination ,Molecular Typing ,030104 developmental biology ,Infectious Diseases ,PCR ,L.~pneumophila ,ST109 ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Sequence Analysis ,Genome, Bacterial ,WGS - Abstract
International audience; OBJECTIVES: Legionella pneumophila serogroup 1 (Lp1) sequence type 47 is the leading cause of legionellosis in north-western Europe, but, surprisingly, it is rarely isolated from environmental samples. Comparative genomics was applied to develop a PCR assay and to better understand the evolution of this strain. METHODS: Comparative analysis of 36 genomes representative of the Lp species was used to identify specific PCR targets, which were then evaluated in silico on 545 sequenced genomes and in~vitro on 436 Legionella strains, 106 respiratory samples, and three environmental samples from proven ST47 sources. Phylogenetic analyses were performed to understand the evolution of ST47. RESULTS: The gene LPO\₁073 was characterized as being 100% conserved in all 129 ST47 genomes analysed. A real-time PCR designed to detect LPO\₁073 was positive for all 110 ST47 strains tested and agreed with culture and typing results previously obtained for 106 respiratory samples. The three environmental samples were also positive. Surprisingly, 26 of the 44 ST109 strains tested among 342 non-ST47 strains scored positive for LPO\₁073. SNP-based phylogenetic analysis was undertaken to understand this result: the PCR-positive ST109 genomes were almost identical to ST47 genomes, with the exception of a recombined region probably acquired by ST47 from a ST62(-like) strain. CONCLUSION: The genomic analysis allowed the design of a highly specific PCR assay for rapid detection of ST47 strains. Furthermore, it allowed us to uncover the evolution of ST47 strains from ST109 by homologous recombination with ST62. We hypothesize that this recombination generated the leading cause of legionellosis in north-western Europe.
- Published
- 2017
- Full Text
- View/download PDF
43. Ribosomal Mutations Conferring Macrolide Resistance in Legionella pneumophila
- Author
-
Françoise Forey, Ghislaine Descours, Sophie Jarraud, Jerome Etienne, Joelle Chastang, Nathalie Jacotin, Christophe Ginevra, Gerard Lina, Elisabeth Kay, Patricia Doublet, Pathogenèse des légionelles- Legionella pathogenesis (LegioPath), Centre International de Recherche en Infectiologie - UMR (CIRI), Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS), Centre National de Référence Legionella, Pathogénie des Staphylocoques – Staphylococcal Pathogenesis (StaPath), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Ribosomal Proteins ,Lineage (genetic) ,030106 microbiology ,Drug Resistance ,Erythromycin ,Microbial Sensitivity Tests ,ribosomal mutations ,Azithromycin ,medicine.disease_cause ,Legionella pneumophila ,Microbiology ,resistance ,03 medical and health sciences ,Antibiotic resistance ,Bacterial Proteins ,Ribosomal protein ,23S ribosomal RNA ,Mechanisms of Resistance ,Drug Resistance, Bacterial ,medicine ,Protein Isoforms ,Pharmacology (medical) ,Pharmacology ,Ribosomal ,Mutation ,biology ,Bacterial ,High-Throughput Nucleotide Sequencing ,23S rRNA ,macrolide ,Ribosomal RNA ,biology.organism_classification ,23S ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,3. Good health ,Anti-Bacterial Agents ,Clone Cells ,RNA, Ribosomal, 23S ,Infectious Diseases ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,RNA ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,medicine.drug - Abstract
Monitoring the emergence of antibiotic resistance is a recent issue in the treatment of Legionnaires' disease. Macrolides are recommended as first-line therapy, but resistance mechanisms have not been studied in Legionella species. Our aim was to determine the molecular basis of macrolide resistance in L. pneumophila . Twelve independent lineages from a common susceptible L. pneumophila ancestral strain were propagated under conditions of erythromycin or azithromycin pressure to produce high-level macrolide resistance. Whole-genome sequencing was performed on 12 selected clones, and we investigated mutations common to all lineages. We reconstructed the dynamics of mutation for each lineage and demonstrated their involvement in decreased susceptibility to macrolides. The resistant mutants were produced in a limited number of passages to obtain a 4,096-fold increase in erythromycin MICs. Mutations affected highly conserved 5-amino-acid regions of L4 and L22 ribosomal proteins and of domain V of 23S rRNA (G2057, A2058, A2059, and C2611 nucleotides). The early mechanisms mainly affected L4 and L22 proteins and induced a 32-fold increase in the MICs of the selector drug. Additional mutations related to 23S rRNA mostly occurred later and were responsible for a major increase of macrolide MICs, depending on the mutated nucleotide, the substitution, and the number of mutated genes among the three rrl copies. The major mechanisms of the decreased susceptibility to macrolides in L. pneumophila and their dynamics were determined. The results showed that macrolide resistance could be easily selected in L. pneumophila and warrant further investigations in both clinical and environmental settings.
- Published
- 2017
- Full Text
- View/download PDF
44. The Legionella pneumophila Collagen-Like Protein Mediates Sedimentation, Autoaggregation, and Pathogen-Phagocyte Interactions
- Author
-
Donald E. Low, Carla Duncan, Mena Abdel-Nour, Mauricio R. Terebiznik, Akriti Prashar, Christophe Ginevra, Alexander W. Ensminger, Chitong Rao, Cyril Guyard, and Sophie Jarraud
- Subjects
Phagocyte ,Cations, Divalent ,Legionella ,Biology ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Legionella pneumophila ,Bacterial Adhesion ,Microbiology ,Bacterial Proteins ,hemic and lymphatic diseases ,otorhinolaryngologic diseases ,Environmental Microbiology ,Escherichia coli ,medicine ,Lobosea ,Gene ,Pathogen ,Acanthamoeba castellanii ,Phagocytes ,Ecology ,Biofilm ,biology.organism_classification ,respiratory tract diseases ,stomatognathic diseases ,medicine.anatomical_structure ,Host-Pathogen Interactions ,bacteria ,Gene Deletion ,Food Science ,Biotechnology - Abstract
Although only partially understood, multicellular behavior is relatively common in bacterial pathogens. Bacterial aggregates can resist various host defenses and colonize their environment more efficiently than planktonic cells. For the waterborne pathogen Legionella pneumophila , little is known about the roles of autoaggregation or the parameters which allow cell-cell interactions to occur. Here, we determined the endogenous and exogenous factors sufficient to allow autoaggregation to take place in L. pneumophila . We show that isolates from Legionella species which do not produce the Legionella collagen-like protein (Lcl) are deficient in autoaggregation. Targeted deletion of the Lcl-encoding gene ( lpg2644 ) and the addition of Lcl ligands impair the autoaggregation of L. pneumophila . In addition, Lcl-induced autoaggregation requires divalent cations. Escherichia coli producing surface-exposed Lcl is able to autoaggregate and shows increased biofilm production. We also demonstrate that L. pneumophila infection of Acanthamoeba castellanii and Hartmanella vermiformis is potentiated under conditions which promote Lcl dependent autoaggregation. Overall, this study shows that L. pneumophila is capable of autoaggregating in a process that is mediated by Lcl in a divalent-cation-dependent manner. It also reveals that Lcl potentiates the ability of L. pneumophila to come in contact, attach, and infect amoebae.
- Published
- 2014
- Full Text
- View/download PDF
45. Macrolide resistance in Legionella pneumophila: the role of LpeAB efflux pump
- Author
-
Ghislaine Descours, S. Jarraud, Clémence Massip, Christophe Ginevra, Christophe Gilbert, Patricia Doublet, Pathogenèse des légionelles- Legionella pathogenesis (LegioPath), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre National de Référence Legionella, Centre International de Recherche en Infectiologie - UMR (CIRI), Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Microbiology (medical) ,Operon ,030106 microbiology ,Drug Resistance ,Drug resistance ,Microbial Sensitivity Tests ,Biology ,Azithromycin ,medicine.disease_cause ,Legionella pneumophila ,Microbiology ,03 medical and health sciences ,Bacterial Proteins ,Drug Resistance, Multiple, Bacterial ,medicine ,Pharmacology (medical) ,Gene ,Escherichia coli ,Pharmacology ,Ribosomal ,Mutation ,Bacterial ,Membrane Transport Proteins ,Ribosomal RNA ,biology.organism_classification ,23S ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,3. Good health ,Anti-Bacterial Agents ,Erythromycin ,RNA, Ribosomal, 23S ,Infectious Diseases ,Genes ,Genes, Bacterial ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,bacteria ,RNA ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Efflux ,Macrolides ,Multiple - Abstract
International audience; Objectives: A previous study on 12 in vitro -selected azithromycin-resistant Legionella~pneumophila lineages showed that ribosomal mutations were major macrolide resistance determinants. In addition to these mechanisms that have been well described in many species, mutations upstream of lpeAB operon, homologous to acrAB in Escherichia~coli , were identified in two lineages. In this study, we investigated the role of LpeAB and of these mutations in macrolide resistance of L.~pneumophila . Methods: The role of LpeAB was studied by testing the antibiotic susceptibility of WT, deleted and complemented L.~pneumophila Paris strains. Translational fusion experiments using GFP as a reporter were conducted to investigate the consequences of the mutations observed in the upstream sequence of lpeAB operon. Results: We demonstrated the involvement of LpeAB in an efflux pump responsible for a macrolide-specific reduced susceptibility of L.~pneumophila Paris strain. Mutations in the upstream sequence of lpeAB operon were associated with an increased protein expression. Increased expression was also observed under sub-inhibitory macrolide concentrations in strains with both mutated and WT promoting regions. Conclusions: LpeAB are components of an efflux pump, which is a macrolide resistance determinant in L.~pneumophila Paris strain. Mutations observed in the upstream sequence of lpeAB operon in resistant lineages led to an overexpression of this efflux pump. Sub-inhibitory concentrations of macrolides themselves participated in upregulating this efflux and could constitute a first step in the acquisition of a high macrolide resistance level.
- Published
- 2017
- Full Text
- View/download PDF
46. Molecular epidemiology, phylogeny and evolution of Legionella
- Author
-
A. Khodr, Christophe Ginevra, Laura Gomez-Valero, S. Jarraud, Carmen Buchrieser, E. Kay, Patricia Doublet, Biologie des Bactéries intracellulaires - Biology of Intracellular Bacteria, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Pathogenèse des légionelles- Legionella pathogenesis (LegioPath), Centre International de Recherche en Infectiologie - UMR (CIRI), École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre National de Référence des Légionelles (CNR), Work in SJ and PD laboratory is performed within the framework of the LABEX ECOFECT (ANR-11-LABX-0042) of Université de Lyon, within the program 'Investissements d'Avenir' (ANR-11-IDEX-0007) operated by the French National Research Agency (ANR) and supported by the Institut national de la santé et de la recherche médicale (INSERM), the Fondation pour la Recherche Médicale (FRM) grant n° DBI20131228 568, the Hospices Civils de Lyon and the General Direction for Health (DGS)., Work in CB laboratory is financed by the Institut Pasteur, the Centre National de Recherche Scientifique (CNRS), the Institut Carnot-Pasteur MI, the French Region Ile de France (DIM Malinf), the grant n°ANR-10-LABX-62-IBEID, the Infect-ERA project EUGENPATH (ANR-13-IFEC- 0003-02) and the Fondation pour la Recherche Médicale (FRM) grant N° DEQ20120323697., ANR-11-LABX-0048,ECOFECT,Dynamiques eco-évolutives des maladies infectieuses(2011), ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-13-IFEC-0003,EUGENPATH,Eukaryotic genes in vacuolar pathogens and symbionts - Implications for virulence, metabolism and ecology(2013), Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Centre International de Recherche en Infectiologie (CIRI), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Microbiology (medical) ,Genotyping Techniques ,Legionella ,Virulence Factors ,Epidemiology ,ICEs ,030106 microbiology ,Virulence ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Microbiology ,Legionella pneumophila ,Evolution, Molecular ,03 medical and health sciences ,Genetics ,medicine ,Humans ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Phylogeny ,Experimental evolution ,Molecular Epidemiology ,Host cell cytosol ,Molecular epidemiology ,biology ,HGT ,Sequence Analysis, DNA ,medicine.disease ,biology.organism_classification ,bacterial infections and mycoses ,Adaptation, Physiological ,3. Good health ,respiratory tract diseases ,Infectious Diseases ,bacteria ,Legionnaires' disease ,Mobile genetic elements ,Legionnaires' Disease ,T4ASS - Abstract
International audience; Legionella are opportunistic pathogens that develop in aquatic environments where they multiply in protozoa. When infected aerosols reach the human respiratory tract they may accidentally infect the alveolar macrophages leading to a severe pneumonia called Legionnaires' disease (LD). The ability of Legionella to survive within host-cells is strictly dependent on the Dot/Icm Type 4 Secretion System that translocates a large repertoire of effectors into the host cell cytosol. Although Legionella is a large genus comprising nearly 60 species that are worldwide distributed, only about half of them have been involved in LD cases. Strikingly, the species Legionella pneumophila alone is responsible for 90% of all LD cases. The present review summarizes the molecular approaches that are used for L. pneumophila genotyping with a major focus on the contribution of whole genome sequencing (WGS) to the investigation of local L. pneumophila outbreaks and global epidemiology studies. We report the newest knowledge regarding the phylogeny and the evolution of Legionella and then focus on virulence evolution of those Legionella species that are known to have the capacity to infect humans. Finally, we discuss the evolutionary forces and adaptation mechanisms acting on the Dot/Icm system itself as well as the role of mobile genetic elements (MGE) encoding T4ASSs and of gene duplications in the evolution of Legionella and its adaptation to different hosts and lifestyles.
- Published
- 2016
- Full Text
- View/download PDF
47. Multiple major disease-associated clones of Legionella pneumophila have emerged recently and independently
- Author
-
Timothy G. Harrison, Laurence Ma, Christophe Ginevra, Carmen Buchrieser, John A. Lees, Anthony Underwood, Christiane Bouchier, Julian Parkhill, Simon R. Harris, Christophe Rusniok, Pierre Lechat, Sophia David, Philippe Glaser, Sophie Jarraud, M. Mentasti, Laura Gomez-Valero, The Wellcome Trust Sanger Institute [Cambridge], Public Health England [London], Biologie des Bactéries intracellulaires - Biology of Intracellular Bacteria, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Hub Bioinformatique et Biostatistique - Bioinformatics and Biostatistics HUB, Centre National de Référence des Légionelles [Lyon], Institut des Agents Infectieux [Lyon] (IAI), Hospices Civils de Lyon (HCL)-Hospices Civils de Lyon (HCL), Ecologie et Evolution de la Résistance aux Antibiotiques / Ecology and Evolution of Antibiotics Resistance (EERA), Université Paris-Sud - Paris 11 (UP11)-Institut Pasteur [Paris] (IP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Centre National de la Recherche Scientifique (CNRS), Génomique (Plate-Forme) - Genomics Platform, Institut Pasteur [Paris] (IP), Work in the C.B. laboratory is financed by the Institut Pasteur, the Institut Carnot-Pasteur MI, the French Region Ile de France (DIM Malinf), the Agence Nationale de la Recherche (ANR) Grant No. ANR-10-LABX-62-IBEID, the ANR-10-PATH-004 project, in the frame of ERA-Net PathoGenoMics, and the Fondation pour la Recherche Médicale (FRM) Grant No. DEQ20120323697., We thank M. Tichit and M. Hunt for their technical support. The Plate-forme Génomique is a member of 'France Génomique' consortium (ANR10-INBS-09-08). Work at the Sanger Institute is funded by The Wellcome Trust Grant No. 098051., ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), Parkhill, Julian [0000-0002-7069-5958], Apollo - University of Cambridge Repository, Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS), Institut Pasteur [Paris]-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Sud Orsay-Centre National de la Recherche Scientifique (CNRS), and Institut Pasteur [Paris]
- Subjects
0301 basic medicine ,MESH: Mutation ,MESH: Selection, Genetic ,Legionella pneumophila ,Genome ,Polymorphism, Single Nucleotide ,Evolution, Molecular ,03 medical and health sciences ,Phylogenetics ,Convergent evolution ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,MESH: Evolution, Molecular ,MESH: Virulence / genetics ,Genetics ,medicine ,MESH: Legionella pneumophila / isolation & purification ,Humans ,Allele ,Selection, Genetic ,MESH: Phylogeny ,Genetics (clinical) ,Phylogeny ,MESH: Humans ,biology ,Phylogenetic tree ,Virulence ,MESH: Polymorphism, Single Nucleotide ,Research ,MESH: Legionella pneumophila / classification ,MESH: Legionella pneumophila / pathogenicity ,biology.organism_classification ,medicine.disease ,MESH: Legionella pneumophila / genetics ,030104 developmental biology ,[SDV.MP]Life Sciences [q-bio]/Microbiology and Parasitology ,Mutation ,Biological dispersal ,Legionnaires' disease ,MESH: Legionnaires' Disease / microbiology ,Legionnaires' Disease - Abstract
Legionella pneumophila is an environmental bacterium and the leading cause of Legionnaires’ disease. Just five sequence types (ST), from more than 2000 currently described, cause nearly half of disease cases in northwest Europe. Here, we report the sequence and analyses of 364 L. pneumophila genomes, including 337 from the five disease-associated STs and 27 representative of the species diversity. Phylogenetic analyses revealed that the five STs have independent origins within a highly diverse species. The number of de novo mutations is extremely low with maximum pairwise single-nucleotide polymorphisms (SNPs) ranging from 19 (ST47) to 127 (ST1), which suggests emergences within the last century. Isolates sampled geographically far apart differ by only a few SNPs, demonstrating rapid dissemination. These five STs have been recombining recently, leading to a shared pool of allelic variants potentially contributing to their increased disease propensity. The oldest clone, ST1, has spread globally; between 1940 and 2000, four new clones have emerged in Europe, which show long-distance, rapid dispersal. That a large proportion of clinical cases is caused by recently emerged and internationally dispersed clones, linked by convergent evolution, is surprising for an environmental bacterium traditionally considered to be an opportunistic pathogen. To simultaneously explain recent emergence, rapid spread and increased disease association, we hypothesize that these STs have adapted to new man-made environmental niches, which may be linked by human infection and transmission.
- Published
- 2016
- Full Text
- View/download PDF
48. Legionella pneumophila Sequence Type 1/Paris Pulsotype Subtyping by Spoligotyping
- Author
-
Laure Diancourt, Christophe Ginevra, Jerome Etienne, Hélène Meugnier, Ghislaine Descours, Gerard Lina, Ghislaine Guigon, Nathalie Jacotin, François Vandenesch, Romain Arquilliere, Sophie Jarraud, Valérie Caro, Immunité infection vaccination (I2V), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-IFR128-Institut National de la Santé et de la Recherche Médicale (INSERM), Génotypage des Pathogènes et Santé Publique (Plate-forme) (PF8), Institut Pasteur [Paris] (IP), Centre National de Référence des Staphylocoques, Hospices Civils de Lyon (HCL), Institut National de la Santé et de la Recherche Médicale (INSERM)-IFR128-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon, and Institut Pasteur [Paris]
- Subjects
Microbiology (medical) ,Genotype ,[SDV]Life Sciences [q-bio] ,MESH: Legionnaires' Disease ,Legionella pneumophila ,MESH: Genotype ,03 medical and health sciences ,MESH: DNA Fingerprinting ,Pulsed-field gel electrophoresis ,medicine ,Cluster Analysis ,Humans ,MESH: Molecular Epidemiology ,Genotyping ,030304 developmental biology ,Molecular Epidemiology ,0303 health sciences ,MESH: Humans ,MESH: Molecular Typing ,biology ,Molecular epidemiology ,030306 microbiology ,Bacteriology ,biology.organism_classification ,medicine.disease ,DNA Fingerprinting ,MESH: Cluster Analysis ,MESH: Legionella pneumophila ,Virology ,Subtyping ,Electrophoresis, Gel, Pulsed-Field ,3. Good health ,Molecular Typing ,MESH: France ,MESH: Electrophoresis, Gel, Pulsed-Field ,DNA profiling ,CRISPR Loci ,Legionnaires' disease ,France ,Legionnaires' Disease - 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.
- Published
- 2012
- Full Text
- View/download PDF
49. Insertion Sequences as Highly Resolutive Genomic Markers for Sequence Type 1 Legionella pneumophila Paris
- Author
-
Dominique Schneider, Max Maurin, Sophie Jarraud, Jean Thioulouse, Elisabeth Kay, Mike Vergnes, Christophe Ginevra, Philippe Normand, Laboratoire Adaptation et pathogénie des micro-organismes [Grenoble] (LAPM), Université Joseph Fourier - Grenoble 1 (UJF)-Centre National de la Recherche Scientifique (CNRS), Centre National de Référence des Légionella, Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM), Immunité infection vaccination (I2V), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-IFR128-Institut National de la Santé et de la Recherche Médicale (INSERM), Laboratoire d'Ecologie Microbienne - UMR 5557 (LEM), Institut National de la Recherche Agronomique (INRA)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Ecole Nationale Vétérinaire de Lyon (ENVL)-VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS)-Centre National de la Recherche Scientifique (CNRS), Ecologie quantitative et évolutive des communautés, Département écologie évolutive [LBBE], Laboratoire de Biométrie et Biologie Evolutive - UMR 5558 (LBBE), Université de Lyon-Université de Lyon-Institut National de Recherche en Informatique et en Automatique (Inria)-VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS)-Centre National de la Recherche Scientifique (CNRS)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de Recherche en Informatique et en Automatique (Inria)-VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS)-Centre National de la Recherche Scientifique (CNRS)-Laboratoire de Biométrie et Biologie Evolutive - UMR 5558 (LBBE), Université de Lyon-Université de Lyon-Institut National de Recherche en Informatique et en Automatique (Inria)-VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS)-Centre National de la Recherche Scientifique (CNRS), Laboratoire de Bacteriologie, BRON, Hospices Civils de Lyon (HCL), CHU Grenoble, Université Joseph Fourier - Grenoble 1 (UJF)-CHU Grenoble, French Centre National de la Recherche Scientifique, Bonus Qualité Recherche grant, University Joseph Fourier, Agence Française de Sécurité Sanitaire de l’Environnement et du Travail (AFSSET, grant EST-2007-1), French Ministry of Education and Research, Delorme, Christine, Centre National de la Recherche Scientifique (CNRS)-Université Joseph Fourier - Grenoble 1 (UJF), Institut National de la Santé et de la Recherche Médicale (INSERM)-IFR128-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon, Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS)-Ecole Nationale Vétérinaire de Lyon (ENVL), CNRS UMR 5163, Université Joseph Fourier - Grenoble 1 (UJF), Centre National de la Recherche Scientifique (CNRS)-Ecole Nationale Vétérinaire de Lyon (ENVL)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Recherche Agronomique (INRA)-VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS), Laboratoire Adaptation et pathogénie des micro-organismes [Grenoble] ( LAPM ), Université Joseph Fourier - Grenoble 1 ( UJF ) -Centre National de la Recherche Scientifique ( CNRS ), Université Joseph Fourier - Grenoble 1 ( UJF ), Université de Lyon-Institut National de la Santé et de la Recherche Médicale ( INSERM ), Immunité infection vaccination ( I2V ), Université Claude Bernard Lyon 1 ( UCBL ), Université de Lyon-Université de Lyon-IFR128-Institut National de la Santé et de la Recherche Médicale ( INSERM ), Ecologie microbienne ( EM ), Centre National de la Recherche Scientifique ( CNRS ) -Ecole Nationale Vétérinaire de Lyon ( ENVL ) -Université Claude Bernard Lyon 1 ( UCBL ), Université de Lyon-Université de Lyon-Institut National de la Recherche Agronomique ( INRA ) -VetAgro Sup ( VAS ), Laboratoire de Biométrie et Biologie Evolutive ( LBBE ), Université de Lyon-Université de Lyon-Institut National de Recherche en Informatique et en Automatique ( Inria ) -Centre National de la Recherche Scientifique ( CNRS ), Hospices Civils de Lyon ( HCL ), Université Joseph Fourier - Grenoble 1 ( UJF ) -CHU Grenoble, and Schneider, Dominique
- Subjects
Microbiology (medical) ,Genotype ,Legionella ,colonisation ,infection humaine ,marqueur génomique ,Legionella pneumophila ,Microbiology ,03 medical and health sciences ,eau ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Environmental Microbiology ,medicine ,Humans ,Typing ,Serotyping ,Insertion sequence ,030304 developmental biology ,Genetics ,Molecular Epidemiology ,0303 health sciences ,biology ,030306 microbiology ,Genetic Variation ,Bacteriology ,biology.organism_classification ,medicine.disease ,DNA Fingerprinting ,Bacterial Typing Techniques ,Electrophoresis, Gel, Pulsed-Field ,DNA profiling ,[ SDV.BBM.GTP ] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,DNA Transposable Elements ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Legionnaires' disease ,Legionnaires' Disease ,Restriction fragment length polymorphism ,france ,Polymorphism, Restriction Fragment Length - Abstract
The causative agent of legionellosis, Legionella pneumophila , colonizes all natural and human-made water networks, thus constituting the source of contaminated aerosols responsible for airborne human infections. Efficient control of infections, especially during epidemics, necessitates the fastest and most resolutive identification possible of the bacterial source for subsequent disinfection of reservoirs. We thus compared recognized typing approaches for Legionella with a method based on characterization of insertion sequence (IS) content. A total of 86 clinical or environmental isolates of L. pneumophila , including 84 Paris isolates, sampled from 25 clinical investigations in France between 2001 and 2007, were obtained from the Legionella National Reference Center. All strains were typed by monoclonal antibody subgrouping, sequence-based typing, pulsed-field gel electrophoresis, and restriction fragment length polymorphism based on the presence or absence of IS elements. We identified six different types of IS elements in L. pneumophila Paris and used them as genomic markers in hybridization experiments. One IS type, IS Lpn11 , revealed a high discriminatory power. Simpson's index of discrimination, calculated from the distribution of IS elements, was higher than that obtained with the other typing methods used for L. pneumophila Paris. Moreover, specific IS Lpn11 copies were found only in strains isolated from particular cities. In more than half of the cases, each clinical isolate had an IS Lpn11 profile that was recovered in at least one environmental isolate from the same geographical location, suggesting that our method could identify the infection source. Phylogenetic analysis suggests a clonal expansion for the L. pneumophila Paris strain.
- Published
- 2011
- Full Text
- View/download PDF
50. Evaluation of BMPA, MWY, GVPC and BCYE media for the isolation of Legionella species from respiratory samples
- Author
-
Françoise Forey, S. Jarraud, Christophe Ginevra, Gerard Lina, Ghislaine Descours, P. Cassier, and Jerome Etienne
- Subjects
Microbiology (medical) ,Respiratory samples ,MWY ,Legionella ,Respiratory System ,Biology ,biology.organism_classification ,Isolation (microbiology) ,Microbiology ,Virology ,Culture Media ,Isolation ,BMPA ,Humans ,GVPC ,Legionella species ,Molecular Biology - Abstract
Culture media performance is a critical factor in the isolation of Legionellae from respiratory samples. We showed that BMPA and MWY media yielded significantly higher isolation rates than GVPC and BCYE media in regard to performance with samples that harbored low Legionella inocula and high contamination levels.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.