Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Clin Microbiol ; 60(6): e0242221, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35510500

RESUMEN

Achromobacter spp. are nonfermenting Gram-negative bacilli mainly studied among cystic fibrosis (CF) patients. The identification of the 19 species within the genus is time-consuming (nrdA-sequencing), thus data concerning the distribution of the species are limited to specific studies. Recently, we built a database using MALDI-TOF mass spectrometry (MS) (Bruker) that allows rapid and accurate species identification and detection of the multiresistant epidemic clones: A. xylosoxidans ST137 spreading among CF patients in various French and Belgium centers, and A. ruhlandii DES in Denmark. Here, we first assessed whether species identification could be achieved with our database solely by analysis of MS spectra without availability of isolates. Then, we conducted a multicentric study describing the distribution of Achromobacter species and of the clone ST137 among French CF centers. We collected and analyzed with our local database the spectra of Achromobacter isolates from 193 patients (528 samples) from 12 centers during 2020. In total, our approach enabled to conclude for 502/528 samples (95.1%), corresponding to 181 patients. Eleven species were detected, only five being involved in chronic colonization, A. xylosoxidans (86.4%), A. insuavis (9.1%), A. mucicolens (2.3%), A. marplatensis (1.1%) and A. genogroup 3 (1.1%). This study confirmed the high prevalence of A. xylosoxidans in chronic colonizations and the circulation of the clone A. xylosoxidans ST137 in France: four patients in two centers. The present study is the first to report the distribution of Achromobacter species from CF patients samples using retrospective MALDI-TOF/MS data. This easy approach could enable future large-scale epidemiological studies.


Asunto(s)
Achromobacter , Fibrosis Quística , Infecciones por Bacterias Gramnegativas , Achromobacter/genética , Fibrosis Quística/epidemiología , Infecciones por Bacterias Gramnegativas/diagnóstico , Infecciones por Bacterias Gramnegativas/epidemiología , Humanos , Estudios Retrospectivos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Análisis Espectral
2.
J Antimicrob Chemother ; 77(4): 926-929, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35029278

RESUMEN

BACKGROUND: Acquired antimicrobial resistance among Achromobacter isolates from cystic fibrosis (CF) patients is frequent. Data concerning the mechanisms involved are scarce. The role of the AxyXY-OprZ and AxyEF-OprN Resistance Nodulation Division (RND) efflux systems has been demonstrated, but not that of AxyABM. OBJECTIVES: To explore the role of efflux systems in the acquired multiresistance observed in a one-step mutant selected after ofloxacin exposure. METHODS: The in vitro resistant mutant NCF-39-Bo2 and its parental strain NCF-39 (MICs of meropenem of 8 and 0.19 mg/L, of ceftazidime of 12 and 3 mg/L, of cefiderocol of 0.094 and 0.032 mg/L and of ciprofloxacin of 8 and 1.5 mg/L, respectively) were investigated by RNA-seq and WGS. Gene inactivation and reverse transcription quantitative PCR (RT-qPCR) were used to explore the role of the efflux systems of interest. RESULTS: RNA-seq showed that the AxyABM efflux system was overproduced (about 40-fold) in the in vitro mutant NCF-39-Bo2 versus its parental strain NCF-39. A substitution in AxyR, the putative regulator of AxyABM, was detected in NCF-39-Bo2. Gene inactivation of axyB (encoding the transporter component) in NCF-39-Bo2 led to a decrease in MICs of ciprofloxacin (5-fold), meropenem (64-fold), ceftazidime (12-fold) and cefiderocol (24-fold). Inactivation of axyB in the clinical isolate AXX-H2 harbouring a phenotype of resistance close to that of NCF-39-Bo2 enhanced the activity of the same molecules, especially meropenem. CONCLUSIONS: AxyABM overproduction is involved in acquired resistance of Achromobacter to ciprofloxacin, meropenem and ceftazidime, antibiotics widely used in CF patients, and increases the MIC of the new promising antibiotic cefiderocol.


Asunto(s)
Achromobacter denitrificans , Achromobacter , Infecciones por Bacterias Gramnegativas , Achromobacter/genética , Achromobacter denitrificans/genética , Antibacterianos/farmacología , Humanos , Pruebas de Sensibilidad Microbiana
3.
BMC Microbiol ; 22(1): 192, 2022 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-35933363

RESUMEN

BACKGROUND: Bacterial biofilm can occur on all medical implanted devices and lead to infection and/or dysfunction of the device. In this study, artificial biofilm was formed on four different medical implants (silicone, piccline, peripheral venous catheter and endotracheal tube) of interest for our daily clinical and/or research practice. We investigated the best conventional technic to dislodge the biofilm on the implants and quantified the number of bacteria. Staphylococcus epidermidis previously isolated from a breast implant capsular contracture on a patient in the university hospital of Dijon was selected for its ability to produce biofilm on the implants. Different technics (sonication, Digest-EUR®, mechanized bead mill, combination of sonication plus Digest-EUR®) were tested and compared to detach the biofilm before quantifying viable bacteria by colony counting. RESULTS: For all treatments, the optical and scanning electron microscope images showed substantial less biofilm biomass remaining on the silicone implant compared to non-treated implant. This study demonstrated that the US procedure was statistically superior to the other physical treatment: beads, Digest-EUR® alone and Digest-EUR® + US (p < 0.001) for the flexible materials (picc-line, PIV, and silicone). The number of bacteria released by the US is significantly higher with a difference of 1 log on each material. The result for a rigid endotracheal tube were different with superiority for the chemical treatment dithiothreitol: Digest-EUR®. Surprisingly the combination of the US plus Digest-EUR® treatment was consistently inferior for the four materials. CONCLUSIONS: Depending on the materials used, the biofilm dislodging technique must be adapted. The US procedure was the best technic to dislodge S. epidermidis biofilm on silicone, piccline, peripheral venous catheter but not endotracheal tube. This suggested that scientists should compare themselves different methods before designing a protocol of biofilm study on a given material.


Asunto(s)
Biopelículas , Staphylococcus epidermidis , Humanos , Siliconas , Sonicación
4.
J Clin Microbiol ; 59(10): e0094621, 2021 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-34346714

RESUMEN

Achromobacter spp. are increasingly reported among cystic fibrosis patients. Genotyping requires time-consuming methods such as multilocus sequence typing or pulsed-field gel electrophoresis. Therefore, data on the prevalence of multiresistant epidemic clones, especially A. xylosoxidans ST137 (AxST137) and the Danish epidemic strain A. ruhlandii (DES), are lacking. We recently developed and published a database for Achromobacter species identification by matrix-assisted laser desorption-ionization-time of flight mass spectrometry (MALDI-TOF MS; Bruker Daltonics). The aim of this study was to evaluate the ability of the MALDI-TOF MS to distinguish these multiresistant epidemic clones within Achromobacter species. All the spectra of A. xylosoxidans (n = 1,571) and A. ruhlandii (n = 174) used to build the local database were analyzed by ClinProTools, MALDI Biotyper PCA, MALDI Biotyper dendrogram, and flexAnalysis software for biomarker peak detection. Two hundred two isolates (including 48 isolates of AxST137 and 7 of DES) were tested. Specific biomarker peaks were identified: absent peak at m/z 6,651 for AxST137 isolates and present peak at m/z 9,438 for DES isolates. All tested isolates were well typed by our local database and clustered within distinct groups (ST137 or non-ST137 and DES or non-DES) no matter the MALDI-TOF software or only by simple visual inspection of the spectra by any user. The use of MALDI-TOF MS allowed us to identify isolates of A. xylosoxidans belonging to the AxST137 clone that spread in France and Belgium (the Belgian epidemic clone) and of A. ruhlandii belonging to the DES clone. This tool will help the implementation of segregation measures to avoid interpatient transmission of these resistant clones.


Asunto(s)
Achromobacter denitrificans , Achromobacter , Fibrosis Quística , Epidemias , Achromobacter denitrificans/genética , Células Clonales , Fibrosis Quística/complicaciones , Fibrosis Quística/epidemiología , Humanos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
5.
J Antimicrob Chemother ; 76(2): 297-304, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33156919

RESUMEN

BACKGROUND: Achromobacter are emerging pathogens in cystic fibrosis patients. Mechanisms of resistance to fluoroquinolones are unknown in clinical isolates. Among non-fermenting Gram-negative bacilli, fluoroquinolone resistance is mostly due to amino acid substitutions in localized regions of the targets (GyrA, GyrB, ParC and ParE) named QRDRs, but also to efflux. OBJECTIVES: To explore quinolone resistance mechanisms in Achromobacter. METHODS: The putative QRDRs of GyrA, GyrB, ParC and ParE were sequenced in 62 clinical isolates, and in vitro one-step mutants obtained after exposure to fluoroquinolones. An in vitro mutant and its parental isolate were investigated by RNASeq and WGS. RT-qPCR and gene inactivation were used to explore the role of efflux systems overexpression. RESULTS: We detected seven substitutions in QRDRs (Q83L/S84P/D87N/D87G for GyrA, Q480P for GyrB, T395A/K525Q for ParE), all in nine of the 27 clinical isolates with ciprofloxacin MIC ≥16 mg/L, whereas none among the in vitro mutants. The RND efflux system AxyEF-OprN was overproduced (about 150-fold) in the in vitro mutant NCF-39-Bl6 versus its parental strain NCF-39 (ciprofloxacin MICs 64 and 1.5 mg/L, respectively). A substitution in AxyT (putative regulator of AxyEF-OprN) was detected in NCF-39-Bl6. Ciprofloxacin MIC in NCF-39-Bl6 dropped from 64 to 1.5 mg/L following gene inactivation of either axyT or axyF. Substitutions in AxyT associated with overexpression of AxyEF-OprN were also detected in seven clinical strains with ciprofloxacin MIC ≥16 mg/L. CONCLUSIONS: Target alteration is not the primary mechanism involved in fluoroquinolone resistance in Achromobacter. The role of AxyEF-OprN overproduction was demonstrated in one in vitro mutant.


Asunto(s)
Achromobacter , Fluoroquinolonas , Antibacterianos/farmacología , Girasa de ADN/genética , Topoisomerasa de ADN IV/genética , Farmacorresistencia Bacteriana , Fluoroquinolonas/farmacología , Humanos , Pruebas de Sensibilidad Microbiana , Mutación
6.
J Clin Microbiol ; 57(5)2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30814269

RESUMEN

Cystic fibrosis (CF) patients are commonly colonized by bacterial pathogens, which can induce persistent lung inflammation and may contribute to clinical deterioration. Colonization of CF patients and cross-transmission by Corynebacterium diphtheriae have not been reported so far. The aim of this article was to investigate the possibility of a cross-transmission of C. diphtheriae biovar Belfanti between four patients of a CF center. C. diphtheriae biovar Belfanti (now formally called C. belfantii) isolates were collected from four patients in a single CF care center over a period of 6 years and analyzed by microbiological methods and whole-genome sequencing. Epidemiological links among patients were investigated. Ten isolates were collected from 4 patients. Whole-genome sequencing of one isolate from each patient showed that a single strain was shared among them. In addition, one patient was found to have the same strain in two consecutive samplings performed 9 months apart. The strain was nontoxigenic and was susceptible to most antimicrobial agents. Ciprofloxacin resistance was observed in one patient. The idea of transmission of the strain among patients was supported by the occurrence of same-day visits to the CF center. This study demonstrated colonization of CF patients by C. diphtheriae biovar Belfanti (C. belfantii), and the data suggest persistence and transmission of a unique strain during at least 6 years in a single CF patient care center.


Asunto(s)
Infecciones Asintomáticas , Corynebacterium diphtheriae/aislamiento & purificación , Fibrosis Quística/microbiología , Difteria/transmisión , Adulto , Antibacterianos/farmacología , Corynebacterium diphtheriae/efectos de los fármacos , Corynebacterium diphtheriae/genética , Fibrosis Quística/complicaciones , Fibrosis Quística/epidemiología , Difteria/epidemiología , Difteria/microbiología , Femenino , Francia , Humanos , Masculino , Secuenciación Completa del Genoma
7.
J Antimicrob Chemother ; 73(7): 1804-1807, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29659873

RESUMEN

Objectives: To characterize the structure of Salmonella genomic islands 1 (SGI1s) from two clinical Proteus mirabilis isolates: one producing an ESBL and the other a penicillinase. Methods: WGS completed by PCR and Sanger sequencing was performed to determine sequences of SGI1s from Pm2CHAMA and Pm37THOMI strains. Results: Two new variants of SGI1 named SGI1-Pm2CHAMA (53.6 kb) and SGI1-K7 (55.1 kb) were identified. The backbone of SGI1-Pm2CHAMA shared 99.9% identity with that of SGI1. Its MDR region (26.3 kb) harboured two class 1 integrons (an In2-type integron and an In4-type integron) containing in particular a qacH cassette (encoding a quaternary ammonium compound efflux pump). These two integrons framed a complex region (harbouring among others blaCARB-4) resulting from transposon insertions mediated by IS26 and successive transposition events of ISs (ISAba14 isoform and the new ISPmi2). The second variant (SGI1-K7) had the same backbone as SGI1-K. Its MDR region (29.7 kb) was derived from that of SGI1-K and was generated by three events. The two main events were mediated by IS26: inversion of a large portion of the MDR region of SGI1-K and insertion of a structure previously reported on plasmids carried by prevalent and successful MDR clones of Enterobacteriaceae. This last event led to the insertion of the blaCTX-M-15 gene into SGI1-K7. Conclusions: This study confirmed the great plasticity of the MDR region of SGI1 and its potential key role for the dissemination of clinically significant antibiotic resistance among Enterobacteriaceae.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Islas Genómicas , Proteus mirabilis/efectos de los fármacos , Proteus mirabilis/genética , Salmonella/genética , beta-Lactamasas/genética , ADN Bacteriano/genética , Francia , Genes Bacterianos , Variación Genética , Hospitalización , Humanos , Integrones/genética , Plásmidos , Reacción en Cadena de la Polimerasa , Infecciones por Proteus/microbiología , Proteus mirabilis/enzimología , Salmonella/enzimología , Secuenciación Completa del Genoma
8.
Emerg Infect Dis ; 23(2): 304-307, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28098548

RESUMEN

We isolated IMP-19-producing Pseudomonas aeruginosa from 7 patients with nosocomial infections linked to contaminated sinks in France. We showed that blaIMP-19 was located on various class 1 integrons among 8 species of gram-negative bacilli detected in sinks: P. aeruginosa, Achromobacter xylosoxidans, A. aegrifaciens, P. putida, Stenotrophomonas maltophilia, P. mendocina, Comamonas testosteroni, and Sphingomonas sp.


Asunto(s)
Infección Hospitalaria , Infecciones por Pseudomonas/epidemiología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/genética , beta-Lactamasas/genética , Farmacorresistencia Bacteriana , Francia/epidemiología , Humanos , Pruebas de Sensibilidad Microbiana , Infecciones por Pseudomonas/transmisión , Pseudomonas aeruginosa/clasificación , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/aislamiento & purificación , Microbiología del Agua , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/biosíntesis
9.
Artículo en Inglés | MEDLINE | ID: mdl-28584156

RESUMEN

AxyXY-OprZ is an RND-type efflux system that confers innate aminoglycoside resistance to Achromobacter spp. We investigated here a putative TetR family transcriptional regulator encoded by the axyZ gene located upstream of axyXY-oprZ An in-frame axyZ gene deletion assay led to increased MICs of antibiotic substrates of the efflux system, including aminoglycosides, cefepime, fluoroquinolones, tetracyclines, and erythromycin, indicating that the product of axyZ negatively regulates expression of axyXY-oprZ Moreover, we identified an amino acid substitution at position 29 of AxyZ (V29G) in a clinical Achromobacter strain that occurred during the course of chronic respiratory tract colonization in a cystic fibrosis (CF) patient. This substitution, also detected in three other strains exposed in vitro to tobramycin, led to an increase in the axyY transcription level (5- to 17-fold) together with an increase in antibiotic resistance level. This overproduction of AxyXY-OprZ is the first description of antibiotic resistance acquisition due to modification of a chromosomally encoded mechanism in Achromobacter and might have an impact on the management of infected CF patients. Indeed, tobramycin is widely used for aerosol therapy within this population, and we have demonstrated that it easily selects mutants with increased MICs of not only aminoglycosides but also fluoroquinolones, cefepime, and tetracyclines.


Asunto(s)
Achromobacter/genética , Antibacterianos/farmacología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de Transporte de Membrana/genética , Tobramicina/farmacología , Transactivadores/genética , Achromobacter/efectos de los fármacos , Achromobacter/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana Múltiple/genética , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica/genética , Humanos , Pruebas de Sensibilidad Microbiana , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética , Transactivadores/biosíntesis
10.
Infection ; 45(5): 697-702, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28283947

RESUMEN

BACKGROUND: Streptococcus pyogenes can colonize genitourinary tract, but it is a rare cause of salpingitis. CASE REPORT: We report a case of bilateral salpingitis due to Streptococcus pyogenes in a 34-year-old woman using an intra-uterine device and which occurred following a family history of recurrent S. pyogenes infections. We review 12 other cases reported in the literature, and discuss the pathophysiological mechanisms of this potentially life-threatening disease. CONCLUSION: It is important to take into account consider Streptococcus pyogenes as a cause of acute salpingitis in the context of recent intra-familial Streptococcus pyogenes infections.


Asunto(s)
Salpingitis/diagnóstico , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/diagnóstico , Streptococcus pyogenes/aislamiento & purificación , Adolescente , Adulto , Femenino , Humanos , Dispositivos Intrauterinos , Recurrencia , Salpingitis/tratamiento farmacológico , Salpingitis/microbiología , Infecciones Estreptocócicas/tratamiento farmacológico , Adulto Joven
11.
BMC Infect Dis ; 17(1): 741, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29195496

RESUMEN

BACKGROUND: In the eighties, a multidrug resistant clone of Salmonella Typhimurium DT104 emerged in UK and disseminated worldwide. This clone harbored a Salmonella genomic island 1 (SGI1) that consists of a backbone and a multidrug resistant region encoding for penta-resistance (ampicillin, chloramphenicol/florfenicol, streptomycin/spectinomycin, sulphonamides and tetracycline (ACSSuT)). Several authors suggested that SGI1 might have a potential role in enhancement of virulence properties of Salmonella enterica. The aim of this study was to investigate whether nontyphoidal S. enterica isolates carrying SGI1 cause more severe illness than SGI1 free ones in humans. METHODS: From 2011 to 2016, all patients infected with nontyphoidal S. enterica in our hospital were retrospectively included. All nontyphoidal S. enterica isolates preserved in our University Hospital (Dijon, France) were screened for the presence of SGI1. Clinical and biological data of patients were retrospectively collected to evaluate illness severity. Statistical analysis of data was performed by Kruskal-Wallis test or Fisher's exact test for univariate analysis, and by logistic regression for multivariate analysis. RESULTS: A total of 100 isolates of S. enterica (22 serovars) were collected. Twelve isolates (12%) belonging to 4 serovars harbored SGI1: S. Typhimurium, S. Infantis, S. Kentucky, S. St Paul. The severity of the disease was age-related (for invasive infection, sepsis and inflammatory response) and was associated with immunosuppression (for invasive infection, sepsis and bacteremia) but not with the presence of SGI1 or with antimicrobial resistance. CONCLUSION: A rather high proportion (12%) of human clinical isolates belonging to various serovars (for the first time serovar St Paul) and harboring various antimicrobial resistance profile carried SGI1. Diseases due to SGI1-positive S. enterica or to antimicrobial resistant isolates were not more severe than the others. This first clinical observation should be confirmed by a multicenter and prospective study.


Asunto(s)
Islas Genómicas/genética , Infecciones por Salmonella/etiología , Salmonella enterica/genética , Salmonella enterica/patogenicidad , Adolescente , Adulto , Factores de Edad , Antibacterianos/farmacología , Niño , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Francia , Humanos , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Estudios Retrospectivos , Infecciones por Salmonella/microbiología , Salmonella enterica/efectos de los fármacos , Salmonella enterica/aislamiento & purificación
12.
J Antimicrob Chemother ; 71(8): 2167-70, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27150396

RESUMEN

OBJECTIVES: The objective of this study was to transfer the Salmonella genomic islands (GIs) SGI1 and SGI1-V and the Proteus GI PGI1-PmESC to clinical isolates of Enterobacteriaceae harbouring an A/C2 plasmid. METHODS: The entire genetic structures of SGI1 and PGI1-PmESC from Salmonella Typhimurium and Proteus mirabilis, respectively, were characterized by PCR and DNA sequencing. Ten enterobacterial isolates from different species carrying blaTEM-24 on an A/C2 plasmid were used for the mobilization of SGI1: Escherichia coli, Enterobacter cloacae, Klebsiella pneumoniae, Proteus mirabilis, Enterobacter aerogenes, Citrobacter freundii, Klebsiella oxytoca, Proteus vulgaris, Providencia stuartii and Serratia marcescens. SGI1-V and PGI1-PmESC were transferred to E. aerogenes. Conjugation attempts were also performed using the wild strain E. aerogenes BOL and E. coli K-12 with or without pA/C2. Detection and location of the GI in the transconjugants were assessed by PCR targeting their junctions. RESULTS: The multidrug resistance region of PGI1-PmESC contained a class 1 integron (aadB and aadA2) and regions deriving from transposon Tn501 and a hybrid Tn502/Tn5053 transposon, whereas SGI1 harboured the known determinants responsible for the pentaresistance. The transfer of SGI1 occurred from Salmonella Typhimurium to the 10 enterobacterial isolates, and transfer of SGI1-V and PGI1-PmESC occurred from P. mirabilis to E. aerogenes. In all transconjugants the GI was located at the 3'-end of trmE. SGI1 was also transferred to E. aerogenes BOL (pA/C2) and E. coli K-12 (pA/C2), but not to E. aerogenes BOL and E. coli K-12. CONCLUSIONS: This is the first known description of SGI1 mobilization into a broad range of enterobacterial species harbouring an A/C2 plasmid and the first demonstration of PGI1 movement. The A/C2 plasmid is responsible for the GI mobilization.


Asunto(s)
Enterobacteriaceae/genética , Transferencia de Gen Horizontal , Islas Genómicas , Plásmidos , Conjugación Genética , ADN Bacteriano/química , ADN Bacteriano/genética , Enterobacteriaceae/aislamiento & purificación , Infecciones por Enterobacteriaceae/microbiología , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN , beta-Lactamasas/genética
13.
Mycoses ; 59(6): 383-90, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26931315

RESUMEN

Unlike bacterial infections, the value of procalcitonin (PCT) in detecting fungal infections in leukaemia patients is not clear. To determine whether the monitoring of PCT coupled with C-reactive protein (CRP) and fibrinogen (Fib) could be helpful in the management of pulmonary aspergillosis (IPA) or mucormycosis (PM), we retrospectively analysed the evolution of PCT, CRP and Fib levels in 94 leukaemia patients with proven/probable IPA (n = 77) or PM (n = 17) from D-12 to D12 relative to IFI onset defined as D0. Overall, 2140 assays were performed. From D-12 to D0, 12%, 5% and 1.4% of patients had PCT >0.5, 1 and 1.5 µg l(-1) , respectively, while CRP was >50, 75 and 100 mg l(-1) in 84%, 70% and 57% and Fib was >4, 5 and 6 g l(-1) in 96%, 80% and 61% of cases respectively (P < 10(-7) ). The same trends were observed from D1 to D12. Overall, between D-12 and D12, only 6.4% of patients had PCT >1.5 µg l(-1) , while CRP >100 mg l(-1) and Fib >6 g l(-1) were observed in 80% and 75% of cases respectively (P < 10(-7) ). In leukaemia patients, IPA or PM was accompanied by a significant increase in CRP and Fib while PCT remained low.


Asunto(s)
Proteína C-Reactiva/análisis , Calcitonina/sangre , Fibrinógeno/análisis , Aspergilosis Pulmonar Invasiva/diagnóstico , Leucemia/complicaciones , Mucormicosis/diagnóstico , Neutropenia/complicaciones , Adulto , Anciano , Anciano de 80 o más Años , Aspergillus/aislamiento & purificación , Biomarcadores/sangre , Femenino , Humanos , Inflamación/sangre , Inflamación/microbiología , Aspergilosis Pulmonar Invasiva/sangre , Aspergilosis Pulmonar Invasiva/complicaciones , Aspergilosis Pulmonar Invasiva/microbiología , Leucemia/sangre , Leucemia/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Mucormicosis/sangre , Mucormicosis/microbiología , Neutropenia/sangre , Neutropenia/tratamiento farmacológico , Estudios Retrospectivos , Adulto Joven
16.
Open Forum Infect Dis ; 11(6): ofae295, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38835495

RESUMEN

Among 15 strains of Listeria monocytogenes tested, a synergy between amoxicillin and ceftriaxone was observed in 14 (93%) according to minimal inhibitory concentration strips and 12 (80%) per the checkerboard methods, as well as for 2 of the 3 strains tested by the time-killing curve. This association may be an alternative treatment for listeriosis in the future.

17.
Antimicrob Agents Chemother ; 57(1): 603-5, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23089757

RESUMEN

Achromobacter xylosoxidans is an innately multidrug-resistant pathogen which is emerging in cystic fibrosis (CF) patients. We characterized a new resistance-nodulation-cell division (RND)-type multidrug efflux pump, AxyXY-OprZ. This system is responsible for the intrinsic high-level resistance of A. xylosoxidans to aminoglycosides (tobramycin, amikacin, and gentamicin). Furthermore, it can extrude cefepime, carbapenems, some fluoroquinolones, tetracyclines, and erythromycin. Some of the AxyXY-OprZ substrates are major components widely used to treat pulmonary infections in CF patients.


Asunto(s)
Achromobacter denitrificans/genética , Aminoglicósidos/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Genes MDR , Genoma Bacteriano , Proteínas de Transporte de Membrana/genética , Achromobacter denitrificans/efectos de los fármacos , Achromobacter denitrificans/aislamiento & purificación , Aminoglicósidos/uso terapéutico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Carbapenémicos/farmacología , Carbapenémicos/uso terapéutico , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/microbiología , Fluoroquinolonas/farmacología , Fluoroquinolonas/uso terapéutico , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Infecciones por Bacterias Gramnegativas/microbiología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Pruebas de Sensibilidad Microbiana , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/microbiología , Análisis de Secuencia de ADN
18.
Appl Environ Microbiol ; 79(23): 7142-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24038696

RESUMEN

Achromobacter xylosoxidans is an aerobic nonfermentative Gram-negative rod considered an important emerging pathogen among cystic fibrosis (CF) patients worldwide and among immunocompromised patients. This increased prevalence remains unexplained, and to date no environmental reservoir has been identified. The aim of this study was to identify potential reservoirs of A. xylosoxidans in hospital, domestic, and outdoor environments and to compare the isolates with clinical ones. From 2011 to 2012, 339 samples were collected in Dijon's university hospital, in healthy volunteers' homes in the Dijon area, and in the outdoor environment in Burgundy (soil, water, mud, and plants). We designed a protocol to detect A. xylosoxidans in environmental samples based on a selective medium: MCXVAA (MacConkey agar supplemented with xylose, vancomycin, aztreonam, and amphotericin B). Susceptibility testing, genotypic analysis by pulsed-field gel electrophoresis, and blaOXA-114 sequencing were performed on the isolates. A total of 50 strains of A. xylosoxidans were detected in hospital (33 isolates), domestic (9 isolates), and outdoor (8 isolates) samples, mainly in hand washing sinks, showers, and water. Most of them were resistant to ciprofloxacin (49 strains). Genotypic analysis and blaOXA-114 sequencing revealed a wide diversity among the isolates, with 35 pulsotypes and 18 variants of oxacillinases. Interestingly, 10 isolates from hospital environment were clonally related to clinical isolates previously recovered from hospitalized patients, and one domestic isolate was identical to one recovered from a CF patient. These results indicate that A. xylosoxidans is commonly distributed in various environments and therefore that CF patients or immunocompromised patients are surrounded by these reservoirs.


Asunto(s)
Achromobacter denitrificans/clasificación , Achromobacter denitrificans/aislamiento & purificación , Microbiología Ambiental , Infecciones por Bacterias Gramnegativas/microbiología , Achromobacter denitrificans/efectos de los fármacos , Achromobacter denitrificans/genética , ADN Bacteriano/química , ADN Bacteriano/genética , Electroforesis en Gel de Campo Pulsado , Francia , Variación Genética , Hospitales , Humanos , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Tipificación Molecular , Características de la Residencia , Análisis de Secuencia de ADN , beta-Lactamasas/genética
19.
J Infect ; 85(5): 492-498, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35933039

RESUMEN

OBJECTIVES: To describe the microorganisms responsible for prosthetic joint infections (PJIs) and their antimicrobial susceptibilities, and to propose appropriate empirical antimicrobial treatments (EATs) according to time of occurrence METHODS: This 10-year retrospective study presents the bacterial etiology of 282 consecutive PJIs in a French hospital according to time of occurrence (adapted from Zimmerli's classification: early, <3 months; delayed, 3-12 months; late acute, >12 months with hematogenous seeding or contiguous spread; late chronic, >12 months without hematogenous seeding). The expected efficacy of various EATs was analyzed for each PJI. RESULTS: Staphylococci were the most commonly found bacteria (S. aureus (44.3%), coagulase-negative staphylococci (25.2%) with 15.2% and 49.3% methicillin resistance, respectively), followed by Gram-negative bacilli (GNB) (17.7%) and streptococci (14.9%). The distribution of species varied between categories, but antibiotics targeting GNBs were required in all categories. Imipenem-vancomycin was the most effective combination (99.3%) but should be reserved for patients with suspected resistant GNB. Cefotaxime-vancomycin was less effective in early/delayed and late PJIs (91.1% and 86.1%, respectively), due to resistant GNB and polymicrobial infections. Piperacillin/tazobactam-vancomycin appeared to be appropriate in all situations (>96% efficacy). CONCLUSION: Proposing universal recommendations remains challenging, but a good understanding of the local epidemiology is important for optimizing EATs.


Asunto(s)
Artritis Infecciosa , Infecciones Relacionadas con Prótesis , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Artritis Infecciosa/tratamiento farmacológico , Cefotaxima , Coagulasa , Farmacorresistencia Microbiana , Bacterias Gramnegativas , Humanos , Imipenem , Pruebas de Sensibilidad Microbiana , Piperacilina , Infecciones Relacionadas con Prótesis/tratamiento farmacológico , Infecciones Relacionadas con Prótesis/epidemiología , Infecciones Relacionadas con Prótesis/microbiología , Estudios Retrospectivos , Staphylococcus aureus , Tazobactam , Vancomicina
20.
Open Forum Infect Dis ; 9(7): ofac271, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35854995

RESUMEN

Background: Streptomyces are environmental gram-positive bacilli that can cause ubiquitous mycetoma and, more rarely, invasive infections. We describe the clinical relevance of Streptomyces spp. identified in human samples and characteristics of patients with invasive Streptomyces infections. Methods: We conducted a retrospective (2006-2017) study of Streptomyces isolates identified in clinical samples in French microbiology laboratories. Streptomyces genus was confirmed by a specific 16S rRNA polymerase chain reaction, and antibiotic susceptibility testing was performed by disk diffusion and trimethoprim-sulfamethoxazole minimum inhibitory concentration (E-test) if resistance was suspected. Patient characteristics, treatments, and outcomes were collected. Invasive infection was defined as a positive culture from a sterile site with signs of infection but without cutaneous inoculation. Results: Of 137 Streptomyces isolates, all were susceptible to amikacin (113/113) and linezolid (112/112), and 92.9% to imipenem (105/113). Using disk diffusion, 50.9% (57/112) of isolates were susceptible to trimethoprim-sulfamethoxazole, but most of the apparently resistant isolates (25/36, 69.4%) tested by E-test were ultimately classified as susceptible. Clinical data were obtained for 63/137 (45.9%) isolates: 30 (47.6%) invasive infections, 8 (12.7%) primary cutaneous infections, 22 (34.9%) contaminations, 3 (4.7%) respiratory colonization. Patients with invasive infection were more frequently receiving corticosteroids than patients without invasive infection (11/30, 36.7%, vs 2/25, 8.0%; P = .03), and at 6-month follow-up, 14 of them were cured, 3 had relapsed, 4 were dead, and 9 were lost to follow-up. Conclusions: Half of the clinical samples that grew Streptomyces were from patients with invasive infection. In that case, antimicrobial therapy should include 1 or 2 antibiotics among linezolid, amikacin, or imipenem.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA