RESUMEN
Nanoplastics pose significant environmental problems due to their high mobility and increased toxicity. These particles can cause infertility and inflammation in aquatic organisms, disrupt microbial signaling and act as pollutants carrier. Despite extensive studies on their harmful impact on living organisms, the microbial degradation of nanoplastics is still under research. This study investigated the degradation of nanoplastics by isolating bacteria from the gut microbiome of Tenebrio molitor larvae fed various plastic diets. Five bacterial strains capable of degrading polystyrene were identified, with Achromobacter xylosoxidans M9 showing significant nanoplastic degradation abilities. Within 6 days, this strain reduced nanoplastic particle size by 92.3%, as confirmed by SEM and TEM analyses, and altered the chemical composition of the nanoplastics, indicating a potential for enhanced bioremediation strategies. The strain also caused a 7% weight loss in polystyrene film over 30 days, demonstrating its efficiency in degrading nanoplastics faster than polystyrene film. These findings might enhance plastic bioremediation strategies.
Asunto(s)
Achromobacter denitrificans , Biodegradación Ambiental , Microbioma Gastrointestinal , Poliestirenos , Animales , Poliestirenos/metabolismo , Achromobacter denitrificans/metabolismo , Plásticos/metabolismo , Plásticos/química , Larva/microbiología , Microplásticos/metabolismoRESUMEN
PURPOSE: Cefiderocol susceptibility testing (AST) represents an open challenge for clinical microbiology. Herein, we evaluated the performance of the UMIC® Cefiderocol broth microdilution (BMD) test and disc diffusion on Gram-negative species. METHODS: UMIC® Cefiderocol BMD test, disc diffusion and reference BMD were in parallel performed on a collection of 256 clinical isolates. Categorical agreement (CA), essential agreement (EA), bias, major errors (MEs) and very major errors (VMEs) were calculated for both AST methods. RESULTS: The UMIC® Cefiderocol BMD strip exhibited an EA < 90% (85.5%), a CA higher than 90% (93.7%) and a low number of VMEs (n = 4, 4.2%) and MEs (n = 12, 7.4%). UMIC® Cefiderocol identified 96.2% of the resistant isolates [Enterobacterales, (39/40); P. aeruginosa (19/19); A. xylosoxidans (5/6); S. maltophilia (5/6); Burkholderia spp. (8/8)]. Disc diffusion showed a high CA (from 94.9 to 100%) regardless of disc manufacturer in Enterobacterales, P. aeuroginosa, A. baumannii and S. maltophilia. However, high rates of results falling in the area of technical uncertainty (ATU) were observed in Enterobacterales (34/90, 37.8%) and P. aeruginosa (16/40, 40%). Disc diffusion showed a poor performance in A. xylosoxidans and Burkholderia spp. if PK/PD breakpoint was used (overall, 5/9 VMEs; in contrast, the use of P. aeruginosa-specific breakpoints resulted in 100% of CA with 24.6% of results in the ATU). CONCLUSION: In conclusion, disc diffusion and UMIC® Cefiderocol are valid methods for the determination of cefiderocol susceptibility. Given the high number of results in the ATU by disc diffusion, a combined use of both AST methods may represent a solution to overcome the challenge of cefiderocol susceptibility testing in routine microbiology laboratories.
Asunto(s)
Achromobacter denitrificans , Acinetobacter baumannii , Stenotrophomonas maltophilia , Humanos , Cefiderocol , Antibacterianos/farmacología , Pseudomonas aeruginosa , Pruebas de Sensibilidad MicrobianaRESUMEN
Achromobacter xylosoxidans is one of the nitrogen-fixing bacteria associated with cowpea rhizosphere across Africa. Although its role in improving soil fertility and inducing systemic resistance in plants against pathogens has been documented, there is limited information on its complete genomic characteristics from cowpea roots. Here, we report the complete genome sequence of A. xylosoxidans strain DDA01 isolated from the topsoil of a field where cowpea plants tolerant to cucumber mosaic virus (CMV) were grown in Ibadan, Nigeria. The genome of DDA01 was sequenced via Illumina MiSeq and contained 6,930,067 nucleotides with 67.55% G + C content, 73 RNAs, 59 tRNAs, and 6421 protein-coding genes, including those associated with nitrogen fixation, phosphate solubilization, Indole3-acetic acid production, and siderophore activity. Eleven genetic clusters for secondary metabolites, including alcaligin, were identified. The potential of DDA01 as a plant growth-promoting bacteria with genetic capabilities to enhance soil fertility for resilience against CMV infection in cowpea is discussed. To our knowledge, this is the first complete genome of diazotrophic bacteria obtained from cowpea rhizosphere in sub-Saharan Africa, with potential implications for improved soil fertility, plant disease resistance, and food security.
Asunto(s)
Achromobacter denitrificans , Cucumovirus , Genoma Bacteriano , Rizosfera , Microbiología del Suelo , Vigna , Vigna/virología , Vigna/microbiología , Cucumovirus/genética , Achromobacter denitrificans/genética , Filogenia , Enfermedades de las Plantas/microbiología , Enfermedades de las Plantas/virología , Fijación del Nitrógeno , Composición de Base , Raíces de Plantas/microbiología , Raíces de Plantas/virología , Nigeria , Bacterias Fijadoras de Nitrógeno/genéticaRESUMEN
PURPOSE: Achromobacter xylosoxidans is an emerging pathogen mainly associated with resistant nosocomial infections. This bacteria had been isolated in the ear together with other pathogens in cultures from patients with chronic otitis media, but it had never been reported as a cause of osteomyelitis of the external auditory canal. CASE PRESENTATION: We present a unique case of a healthy 81-year-old woman who presented with left chronic otorrhea refractory to topical and oral antibiotic treatment. Otomicroscopy revealed an erythematous and exudative external auditory canal (EAC) with scant otorrhea. The tympanic membrane was intact, but an area of bone remodeling with a small cavity anterior and inferior to the bony tympanic frame was observed. Otic culture isolated multi-drug-resistant A. xylosoxidans, only sensitive to meropenem and cotrimoxazole. Temporal bone computed tomography showed an excavation of the floor of the EAC compatible with osteomyelitis. Targeted antibiotherapy for 12 weeks was conducted, with subsequent resolution of symptoms and no progression of the bone erosion. CONCLUSIONS: Atypical pathogens such as A. xylosoxidans can be the cause of chronic otitis externa. Early diagnosis and specific antibiotherapy can prevent the development of further complications, such as osteomyelitis. In these cases, otic cultures play an essential role to identify the causal germ. This is the first case of EAC osteomyelitis due to A. xylosoxidans reported to date.
Asunto(s)
Achromobacter denitrificans , Enfermedades del Oído , Osteomielitis , Otitis Externa , Femenino , Humanos , Anciano de 80 o más Años , Conducto Auditivo Externo/diagnóstico por imagen , Otitis Externa/diagnóstico , Otitis Externa/tratamiento farmacológico , Osteomielitis/diagnóstico , Osteomielitis/tratamiento farmacológico , Osteomielitis/complicacionesRESUMEN
Achromobacter xylosoxidans (Ax) is an opportunistic pathogen and causative agent of numerous infections particularly in immunocompromised individuals with increasing prevalence in cystic fibrosis (CF). To date, investigations have focused on the clinical epidemiology and genomic comparisons of Ax isolates, yet little is known about disease pathology or the role that specific virulence factors play in tissue invasion or damage. Here, we model an acute Ax lung infection in immunocompetent C57BL/6 mice and immunocompromised CF mice, revealing a link between in vitro cytotoxicity and disease in an intact host. Mice were intratracheally challenged with sublethal doses of a cytotoxic (GN050) or invasive (GN008) strain of Ax. Bacterial burden, immune cell populations, and inflammatory markers in bronchoalveolar lavage fluid and lung homogenates were measured at different time points to assess disease severity. CF mice had a similar but delayed immune response toward both Ax strains compared to C57BL/6J mice. GN050 caused more severe disease and higher mortality which correlated with greater bacterial burden and increased proinflammatory responses in both mouse models. In agreement with the cytotoxicity of GN050 toward macrophages in vitro, mice challenged with GN050 had fewer macrophages. Mutants with transposon insertions in predicted virulence factors of GN050 showed that disease severity depended on the type III secretion system, Vi capsule, antisigma-E factor, and partially on the ArtA adhesin. The development of an acute infection model provides an essential tool to better understand the infectivity of diverse Ax isolates and enable improved identification of virulence factors important to bacterial persistence and disease.
Asunto(s)
Achromobacter denitrificans , Fibrosis Quística , Infecciones por Bacterias Gramnegativas , Animales , Ratones , Achromobacter denitrificans/genética , Factores de Virulencia/genética , Modelos Animales de Enfermedad , Infecciones por Bacterias Gramnegativas/microbiología , Ratones Endogámicos C57BL , Fibrosis Quística/microbiologíaRESUMEN
Cystic fibrosis (CF) is a genetic disease affecting epithelial ion transport, resulting in thickened mucus and impaired mucociliary clearance. Persons with CF (pwCF) experience life-long infections of the respiratory mucosa caused by a diverse array of opportunists, which are leading causes of morbidity and mortality. In recent years, there has been increased appreciation for the range and diversity of microbes causing CF-related respiratory infections. The introduction of new therapeutics and improved detection methodology has revealed CF-related opportunists such as Achromobacter xylosoxidans (Ax). Ax is a Gram-negative bacterial species which is widely distributed in environmental sources and has been increasingly observed in sputa and other samples from pwCF, typically in patients in later stages of CF disease. In this study, we characterized CF clinical isolates of Ax and tested colonization and persistence of Ax in respiratory infection using immortalized human CF respiratory epithelial cells and BALB/c mice. Genomic analyses of clinical Ax isolates showed homologs for factors including flagellar synthesis, antibiotic resistance, and toxin secretion systems. Ax isolates adhered to polarized cultures of CFBE41o- human immortalized CF bronchial epithelial cells and caused significant cytotoxicity and depolarization of cell layers. Ax colonized and persisted in mouse lungs for up to 72 h post infection, with inflammatory consequences that include increased neutrophil influx in the lung, lung damage, cytokine production, and mortality. We also identified genes that are differentially expressed in synthetic CF sputum media. Based on these results, we conclude that Ax is an opportunistic pathogen of significance in CF.
Asunto(s)
Achromobacter denitrificans , Fibrosis Quística , Infecciones por Bacterias Gramnegativas , Infecciones del Sistema Respiratorio , Animales , Ratones , Humanos , Achromobacter denitrificans/genética , Fibrosis Quística/complicaciones , Fibrosis Quística/microbiología , Esputo/microbiología , Infecciones por Bacterias Gramnegativas/microbiología , Perfilación de la Expresión GénicaRESUMEN
BACKGROUND: Cavendish (AAA) banana plant (Musa spp.) worldwide cultivated crop harbors many endophytic bacteria. Endophytic bacteria are those that live inside plant tissues without producing any visible symptoms of infection. RESULTS: Endophytic bacterium (MRH 11), isolated from root tissue of Musa spp.was identified as Achromobacter xylosoxidans (ON955872) which showed positive effects in IAA production, phosphate solubilization, catalase production. A. xylosoxidans also showed in vitro antagonism against Curvularia lunata causing leaf spot disease of Cavendish (AAA) banana (G-9 variety). The GC-MS analysis of culture filtrate of A. xylosoxidans (ON955872) confirmed this finding. GC-MS analysis was carried by using two solvent etheyl acetate and chloroform and it showed several antifungal compounds. The identification of these bioactive secondary metabolites compounds was based on the peak area, retention time, molecular weight, molecular formula and antimicrobial actions. GC-MS analysis result revealed the presence of major components including Cyclododecane, 1-Octanol, Cetene, Diethyl phthalate. In vivo test to banana plants was carried out in separate field as well as in potted conditions. Appearance of leaf spots after foliar spray of spore of pathogen and reduction in leaf spots after application of bacterial suspension was found. CONCLUSION: The present study has highlighted the role of endophytic bacterium as antagonist to the pathogen Curvularia lunata.
Asunto(s)
Achromobacter denitrificans , Musa , Achromobacter denitrificans/metabolismoRESUMEN
Polycyclic aromatic hydrocarbons (PAHs) are a group of organic pollutants ubiquitous and persistent in soil. In order to provide a viable solution for bioremediation of PAHs-contaminated soil, a strain of Achromobacter xylosoxidans BP1 with superior PAHs degradation ability was isolated from contaminated soil at a coal chemical site in northern China. The degradation of phenanthrene (PHE) and benzo[a]pyrene (BaP) by strain BP1 was investigated in three different liquid phase cultures, and the removal rates of PHE and BaP by strain BP1 were 98.47% and 29.86% after 7 days under the conditions of PHE and BaP as the only carbon source, respectively. In the medium with the coexistence of PHE and BaP, the removal rates of BP1 were 89.44% and 9.42% after 7 days, respectively. Then, strain BP1 was investigated for its feasibility in remediating PAH-contaminated soil. Among the four PAHs-contaminated soils treated differently, the treatment inoculated with BP1 exhibited higher removal rates of PHE and BaP (p < 0.05), especially the CS-BP1 treatment (inoculation of BP1 into unsterilized PAHs-contaminated soil) showed 67.72%, 13.48% removal of PHE and BaP, respectively, over 49 days of incubation. Bioaugmentation also significantly increased the activity of dehydrogenase and catalase in the soil (pï¼0.05). Furthermore, the effect of bioaugmentation on the removal of PAHs was investigated by measuring the activity of dehydrogenase (DH) and catalase (CAT) during incubation. Among them, the DH and CAT activities of CS-BP1 and SCS-BP1 (inoculation of BP1 into sterilized PAHs-contaminated soil) treatments inoculated with strain BP1 were significantly higher than those of treatments without BP1 addition during incubation (p < 0.01). The structure of the microbial community varied among treatments, but the Proteobacteria phylum showed the highest relative abundance in all treatments of the bioremediation process, and most of the bacteria with higher relative abundance at the genus level also belonged to the Proteobacteria phylum. Prediction of microbial functions in soil by FAPROTAX analysis showed that bioaugmentation enhanced microbial functions associated with the degradation of PAHs. These results demonstrate the effectiveness of Achromobacter xylosoxidans BP1 as a PAH-contaminated soil degrader for the risk control of PAHs contamination.
Asunto(s)
Achromobacter denitrificans , Microbiota , Hidrocarburos Policíclicos Aromáticos , Contaminantes del Suelo , Achromobacter denitrificans/metabolismo , Catalasa , Microbiología del Suelo , Contaminantes del Suelo/análisis , Biodegradación Ambiental , Suelo/químicaRESUMEN
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 MicrobianaRESUMEN
OBJECTIVE AND DESIGN: Our research aimed to investigate the role of CD14 in pulmonary infection by Achromobacter xylosoxidans in an experimental murine model. METHODS: C57Bl/6 or CD14-deficient mice were infected intratracheally with non-lethal inoculum of A. xylosoxidans. At times 1, 3 and 7 days after infection, lungs, bronchoalveolar lavage and blood were collected. CD14 gene expression was determined by RT-PCR. The bacterial load in the lungs was assessed by counting colony forming units (CFU). Cytokines, chemokines, lipocalin-2 and sCD14 were quantified by the ELISA method. Inflammatory infiltrate was observed on histological sections stained with HE, and leukocyte subtypes were assessed by flow cytometry. In another set of experiments, C57Bl/6 or CD14-deficient mice were inoculated with lethal inoculum and the survival rate determined. RESULTS: CD14-deficient mice are protected from A. xylosoxidans-induced death, which is unrelated to bacterial load. The lungs of CD14-deficient mice presented a smaller area of tissue damage, less neutrophil and macrophage infiltration, less pulmonary edema, and a lower concentration of IL-6, TNF-α, CXCL1, CCL2 and CCL3 when compared with lungs of C57Bl/6 mice. We also observed that A. xylosoxidans infection increases the number of leukocytes expressing mCD14 and the levels of sCD14 in BALF and serum of C57Bl/6-infected mice. CONCLUSIONS: In summary, our data show that in A. xylosoxidans infection, the activation of CD14 induces intense pulmonary inflammatory response resulting in mice death.
Asunto(s)
Achromobacter denitrificans , Infecciones por Bacterias Gramnegativas , Receptores de Lipopolisacáridos , Neumonía , Animales , Ratones , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/metabolismo , Pulmón/metabolismo , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo , Infecciones por Bacterias Gramnegativas/metabolismoRESUMEN
Lungs of cystic fibrosis patients are often colonized or infected with organisms, such as Pseudomonas aeruginosa and other emerging pathogenic bacteria such as Achromobacter xylosoxidans. Further, it is well established that infections of the cystic fibrosis lung airways are caused by polymicrobial infections, although its composition and diversity may change throughout the patient's life. In the present study, we investigated the effects of N-acetylcysteine (NAC) and amikacin, aztreonam, ciprofloxacin, and tobramycin alone and in combination against single- and dual-species biofilms of P. aeruginosa and A. xylosoxidans, in vitro and in the Caenorhabditis elegans infection model. Results showed that tobramycin and ciprofloxacin were the most effective antibiotics, while aztreonam was the least effective antibiotic against both single- and dual-species biofilms of P. aeruginosa and A. xylosoxidans. However, NAC showed little effect on both single- and dual-species, even with a combination of antibiotics. Increased survival was observed in C. elegans when treated with NAC in combination with tobramycin or ciprofloxacin, compared to no treatment or NAC alone. Tobramycin and ciprofloxacin were found effective in biofilms, but more research is needed to better understand the effects of NAC and antibiotics against single- and dual-species biofilms.
Asunto(s)
Achromobacter denitrificans , Fibrosis Quística , Animales , Humanos , Pseudomonas aeruginosa , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Acetilcisteína/farmacología , Aztreonam/farmacología , Fibrosis Quística/microbiología , Caenorhabditis elegans , Biopelículas , Tobramicina/farmacología , Ciprofloxacina/farmacologíaRESUMEN
The indoor culture method was carried out to study the toxic effect of silver nanoparticles (AgNPs) on Achromobacter denitrificans. Specifically, the effects of AgNPs concentration, temperature and coexisting anions were analyzed. The results showed that AgNPs exerted significant inhibition on the bacteria, which was closely correlated with its concentration and temperature. Both the ammonia oxidation and generation capacity of Achromobacter denitrificans decreased significantly with an increase in AgNPs concentration. Compared with the inhibition performance at 30 °C, NH4+-N generation rates decreased by 45.31% at 20 °C and 17.58% at 40 °C, respectively, revealing that too low or too high temperature induced to reduce the nitrogen conversion ability of Achromobacter denitrificans. While compared with temperature, the effect of coexisting ions (Cl- and SO42-) was not significant (P > 0.05). Electron microscopy observations found that AgNPs non-specifically bound to the cells (content ranging from 0.04% to 0.10%) and acted on the cell surface structure, causing wrinkles, depressions, and ruptures on the surface of cell membranes, and leakage of substances in the membranes. AgNPs increased the rate of cell apoptosis and decreased the cell body volume mainly with short-term acute effects.
Asunto(s)
Achromobacter denitrificans , Nanopartículas del Metal , Temperatura , Plata/toxicidad , Nanopartículas del Metal/toxicidad , AnionesRESUMEN
Treatment options for Achromobacter xylosoxidans are limited. Eight cystic fibrosis patients with A. xylosoxidans were treated with 12 cefiderocol courses. Pretreatment in vitro resistance was seen in 3 of 8 cases. Clinical response occurred after 11 of 12 treatment courses. However, microbiologic relapse was observed after 11 of 12 treatment courses, notably without emergence of resistance.
Asunto(s)
Achromobacter denitrificans , Fibrosis Quística , Infecciones por Bacterias Gramnegativas , Adulto , Antibacterianos/uso terapéutico , Cefalosporinas , Niño , Fibrosis Quística/complicaciones , Fibrosis Quística/tratamiento farmacológico , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Humanos , CefiderocolRESUMEN
Polycyclic aromatic hydrocarbons (PAHs) are widespread and harmful contaminants and are more persistent under anaerobic conditions. The bioremediation of PAHs in anaerobic zones has been enhanced by treating the contamination with nitrate, which is thermodynamically favourable, cost-effective, and highly soluble. However, anaerobic PAHs biotransformation processes that employ nitrate as an electron acceptor have not been fully explored. In this study, we investigated the anaerobic biotransformation of PAHs by strain PheN1, a newly isolated phenanthrene-degrading denitrifier, using phenanthrene as a model compound. PheN1 is phylogenetically closely related to Achromobacter denitrificans and reduces nitrate to nitrite (not N2 ) during the anaerobic phenanthrene degradation process. Phenanthrene biotransformation processes were detected using gas chromatography-mass spectrometry and were further examined by reverse transcription-quantitative PCR and genome analyses. Carboxylation and methylation were both found to be the initial steps in the phenanthrene degradation process. Downstream biotransformation processed benzene compounds and cyclohexane derivatives. This study describes the isolation of an anaerobic phenanthrene-degrading bacterium along with the pure-culture evidence of phenanthrene biotransformation processes with nitrate as an electron acceptor. The findings in this study can improve our understanding of anaerobic PAHs biodegradation processes and guide PAHs bioremediation by adding nitrate to anaerobic environments.
Asunto(s)
Achromobacter denitrificans/metabolismo , Genoma Bacteriano , Nitratos/metabolismo , Fenantrenos/metabolismo , Achromobacter denitrificans/química , Achromobacter denitrificans/genética , Anaerobiosis , Biodegradación Ambiental , Biotransformación , Cromatografía de Gases y Espectrometría de Masas , Nitratos/química , Fenantrenos/química , Hidrocarburos Policíclicos Aromáticos/química , Hidrocarburos Policíclicos Aromáticos/metabolismoRESUMEN
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 DesorciónRESUMEN
BACKGROUND: Cystic fibrosis (CF) affects >70,000 people worldwide, yet the microbiologic trigger for pulmonary exacerbations (PExs) remains unknown. The objective of this study was to identify changes in bacterial metabolic pathways associated with clinical status. METHODS: Respiratory samples were collected at hospital admission for PEx, end of intravenous (IV) antibiotic treatment, and follow-up from 27 hospitalized children with CF. Bacterial DNA was extracted and shotgun DNA sequencing was performed. MetaPhlAn2 and HUMAnN2 were used to evaluate bacterial taxonomic and pathway relative abundance, while DESeq2 was used to evaluate differential abundance based on clinical status. RESULTS: The mean age of study participants was 10 years; 85% received combination IV antibiotic therapy (beta-lactam plus a second agent). Long-chain fatty acid (LCFA) biosynthesis pathways were upregulated in follow-up samples compared to end of treatment: gondoate (p = 0.012), oleate (p = 0.048), palmitoleate (p = 0.043), and pathways of fatty acid elongation (p = 0.012). Achromobacter xylosoxidans and Escherichia sp. were also more prevalent in follow-up compared to PEx (p < 0.001). CONCLUSIONS: LCFAs may be associated with persistent infection of opportunistic pathogens. Future studies should more closely investigate the role of LCFA production by lung bacteria in the transition from baseline wellness to PEx in persons with CF. IMPACT: Increased levels of LCFAs are found after IV antibiotic treatment in persons with CF. LCFAs have previously been associated with increased lung inflammation in asthma. This is the first report of LCFAs in the airway of persons with CF. This research provides support that bacterial production of LCFAs may be a contributor to inflammation in persons with CF. Future studies should evaluate LCFAs as predictors of future PExs.
Asunto(s)
Achromobacter denitrificans/metabolismo , Fibrosis Quística/complicaciones , Escherichia coli/metabolismo , Inflamación/complicaciones , Adolescente , Niño , Preescolar , Fibrosis Quística/metabolismo , Fibrosis Quística/microbiología , Farmacorresistencia Bacteriana , Femenino , Humanos , Lactante , Inflamación/metabolismo , Inflamación/microbiología , Masculino , Estudios ProspectivosRESUMEN
Achromobacter xylosoxidans (A. xylosoxidans) is an aerobic gram-negative bacillus and often isolated from aquatic environments. It is supposed to cause infections in patients with malignancy or immunodeficiency. It causes various healthcare-associated infections, but cellulitis is rare. Herein, we report the first case of sever cellulitis by A. xylosoxidans after allogeneic hematopoietic stem cell transplantation (HSCT). A 49-year-old man underwent allogeneic HSCT from 8/8 HLA-matched unrelated donor with myeloablative conditioning for relapsed acute myeloid leukemia. He developed skin chronic graft versus host disease 11 months after HSCT. During the prolonged treatment with prednisolone and cyclosporine, he developed cellulitis on his left leg and admitted to our hospital. Blood and exudate culture revealed A. xylosoxidans. Although empirical therapy with cefepime was ineffective, his symptoms were dramatically improved after administration of meropenem. To our knowledge, this is the first case of A. xylosoxidans cellulitis after allogeneic HSCT. A. xylosoxidans should be considered as a possible cause of cellulitis in post-allogeneic HSCT patients on prolonged immunosuppressive therapy.
Asunto(s)
Achromobacter denitrificans , Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Celulitis (Flemón)/tratamiento farmacológico , Celulitis (Flemón)/etiología , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Masculino , Persona de Mediana Edad , Trasplante Homólogo/efectos adversosRESUMEN
(1) Background: Environmental contamination with antibiotics is particularly serious because the usual methods used in wastewater treatment plants turn out to be insufficient or ineffective. An interesting idea is to support natural biodegradation processes with physicochemical methods as well as with bioaugmentation with efficient microbial degraders. Hence, the aim of our study is evaluation of the effectiveness of different methods of nitrofurazone (NFZ) degradation: photolysis and photodegradation in the presence of two photocatalysts, the commercial TiO2-P25 and a self-obtained Fe3O4@SiO2/TiO2 magnetic photocatalyst. (2) Methods: The chemical nature of the photocatalysis products was investigated using a spectrometric method, and then, they were subjected to biodegradation using the strain Achromobacter xylosoxidans NFZ2. Additionally, the effects of the photodegradation products on bacterial cell surface properties and membranes were studied. (3) Results: Photocatalysis with TiO2-P25 allowed reduction of NFZ by over 90%, demonstrating that this method is twice as effective as photolysis alone. Moreover, the bacterial strain used proved to be effective in the removal of NFZ, as well as its intermediates. (4) Conclusions: The results indicated that photocatalysis alone or coupled with biodegradation with the strain A. xylosoxidans NFZ2 leads to efficient degradation and almost complete mineralization of NFZ.
Asunto(s)
Achromobacter denitrificans/metabolismo , Nitrofurazona/aislamiento & purificación , Contaminantes Químicos del Agua/aislamiento & purificación , Achromobacter denitrificans/efectos de los fármacos , Antibacterianos/aislamiento & purificación , Biodegradación Ambiental , Catálisis , Nitrofurazona/química , Nitrofurazona/toxicidad , Procesos Fotoquímicos , Fotólisis , Dióxido de Silicio/química , Espectrofotometría Infrarroja , Titanio/química , Contaminantes Químicos del Agua/química , Contaminantes Químicos del Agua/toxicidadRESUMEN
Achromobacter xylosoxidans is increasingly recognized as a colonizer of cystic fibrosis (CF) patients, but the role that A. xylosoxidans plays in pathology remains unknown. This knowledge gap is largely due to the lack of model systems available to study the toxic potential of this bacterium. Recently, a phospholipase A2 (PLA2) encoded by a majority of A. xylosoxidans genomes, termed AxoU, was identified. Here, we show that AxoU is a type III secretion system (T3SS) substrate that induces cytotoxicity to mammalian cells. A tissue culture model was developed showing that a subset of A. xylosoxidans isolates from CF patients induce cytotoxicity in macrophages, suggestive of a pathogenic or inflammatory role in the CF lung. In a toxic strain, cytotoxicity is correlated with transcriptional activation of axoU and T3SS genes, demonstrating that this model can be used as a tool to identify and track expression of virulence determinants produced by this poorly understood bacterium.
Asunto(s)
Achromobacter denitrificans/fisiología , Infecciones por Bacterias Gramnegativas/microbiología , Sistemas de Secreción Tipo III , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biomarcadores , Línea Celular Tumoral , Fibrosis Quística/complicaciones , Citocinas/metabolismo , Citotoxicidad Inmunológica , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Fagocitosis/inmunología , Factores de VirulenciaRESUMEN
Achromobacter spp. are recognized as emerging pathogens in patients with cystic fibrosis (CF). Though recent works have established species-level identification using nrdA sequencing, there is a dearth in knowledge relating to species-level antimicrobial susceptibility patterns and antimicrobial combinations, which hampers the use of optimal antimicrobial combinations for the treatment of chronic infections. The aims of this study were to (i) identify at species-level referred Achromobacter isolates, (ii) describe species-level antimicrobial susceptibility profiles, and (iii) determine the most promising antimicrobial combination for chronic Achromobacter infections. A total of 112 multidrug-resistant (MDR) Achromobacter species isolates from 39 patients were identified using nrdA sequencing. Antimicrobial susceptibility and combination testing were carried out using the Etest method. We detected six species of Achromobacter and found that Achromobacter xylosoxidans was the most prevalent species. Interestingly, sequence analysis showed it was responsible for persistent infection (18/28 patients), followed by Achromobacter ruhlandii (2/3 patients). Piperacillin-tazobactam (70.27%) and co-trimoxazole (69.72%) were the most active antimicrobials. Differences were observed in species-level susceptibility to ceftazidime, carbapenems, ticarcillin-clavulanate, and tetracycline. Antimicrobial combinations with co-trimoxazole or tobramycin demonstrate the best synergy, while co-trimoxazole gave the best susceptibility breakpoint index values. This study enriches the understanding of MDR Achromobacter spp. epidemiology and confirms prevalence and chronic colonization of A. xylosoxidans in CF lungs. It presents in vitro data to support the efficacy of new combinations for use in the treatment of chronic Achromobacter infections.