Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Evol Lett ; 7(6): 389-400, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38045720

RESUMEN

Pathogenic bacteria respond to antibiotic pressure with the evolution of resistance but survival can also depend on their ability to tolerate antibiotic treatment, known as tolerance. While a variety of resistance mechanisms and underlying genetics are well characterized in vitro and in vivo, an understanding of the evolution of tolerance, and how it interacts with resistance in situ is lacking. We assayed for tolerance and resistance in isolates of Pseudomonas aeruginosa from chronic cystic fibrosis lung infections spanning up to 40 years of evolution, with 3 clinically relevant antibiotics: meropenem, ciprofloxacin, and tobramycin. We present evidence that tolerance is under positive selection in the lung and that it can act as an evolutionary stepping stone to resistance. However, by examining evolutionary patterns across multiple patients in different clone types, a key result is that the potential for an association between the evolution of resistance and tolerance is not inevitable, and difficult to predict.

2.
Clin Microbiol Infect ; 28(7): 990-995, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35124256

RESUMEN

OBJECTIVES: It is unclear whether recurrent sputum culture with Pseudomonas aeruginosa from patients with chronic obstructive pulmonary disease (COPD) is caused by intermittent airway carriage by different P. aeruginosa lineages or persistent carriage by the same lineage, and whether lineages genetically adapt during carriage. METHODS: Whole-genome sequencing was performed for P. aeruginosa isolates sampled longitudinally from sputum cultures in patients with COPD who were enrolled in an ongoing randomized controlled trial (clinicaltrials.gov: NCT03262142). RESULTS: A total of 153 P. aeruginosa isolates were sequenced for 23 patients during 365 days of follow-up. Recurrent presence of P. aeruginosa was seen in 19 patients (83%) and was caused by persistence of the same clonal lineage in all but one patient. We identified 38 genes mutated in parallel in two or more lineages, suggesting positive selection for adaptive mutations. Mutational enrichment analysis revealed genes important in antibiotic resistance and chronic infections to be more frequently mutated. DISCUSSION: Recurrent P. aeruginosa was common and carried for a prolonged time after initial detection in the airways of patients with COPD. Recurrence was caused by persistence of the same clonal lineage and was associated with genetic adaptation. Trial data on possible clinical benefits of attempting antibiotic eradication of P. aeruginosa in COPD are warranted.


Asunto(s)
Infecciones por Pseudomonas , Enfermedad Pulmonar Obstructiva Crónica , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Humanos , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/genética , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/microbiología , Sistema Respiratorio/microbiología
3.
Chron Respir Dis ; 18: 14799731211061600, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34854775

RESUMEN

Objectives: Primary ciliary dyskinesia (PCD) is a rare congenital disease with defective mucociliary clearance causing frequent and often persistent pulmonary infections. Achromobacter species are opportunistic pathogens renowned for the difficulty of effective treatments and deteriorating effects on lung function. We aimed to describe the occurrence, treatment, and rate of successful eradication of Achromobacter species in patients with PCD. Methods: We retrospectively reviewed 18 years of historical microbiological samples and 10 years of electronic health records for PCD patients in Denmark. Results: We included 136 patients. Twenty-six patients had isolates of Achromobacter species. On average, 5% of the cohort had at least one annual isolate. Infections became persistent in 38% with a median length of 6.6 years leading to a significant number of antibiotic treatments. Resistance toward tobramycin and ciprofloxacin was prevalent. Overall, successful eradication was achieved in 62% of patients. We found the course of lung function significantly worse during persistent Achromobacter species infection than during the two preceding years, but not different to the course in unaffected age-matched controls. Conclusion The prevalence of Achromobacter species in patients with PCD is in line with what has been reported in cystic fibrosis and can occur transiently, intermittently, or develop into a serious persistent lung infection associated with long-term antibiotic treatment.


Asunto(s)
Achromobacter , Trastornos de la Motilidad Ciliar , Fibrosis Quística , Antibacterianos/uso terapéutico , Trastornos de la Motilidad Ciliar/tratamiento farmacológico , Fibrosis Quística/complicaciones , Fibrosis Quística/tratamiento farmacológico , Humanos , Estudios Retrospectivos
4.
Microb Genom ; 7(7)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34232117

RESUMEN

Achromobacter spp. are emerging pathogens in patients with cystic fibrosis (CF) and Achromobacter spp. caused infections are associated with more severe disease outcomes and high intrinsic antibiotic resistance. While conventional CF pathogens are studied extensively, little is known about the genetic determinants leading to antibiotic resistance and the genetic adaptation in Achromobacter spp. infections. Here, we analysed 101 Achromobacter spp. genomes from 51 patients with CF isolated during the course of up to 20 years of infection to identify within-host adaptation, mutational signatures and genetic variation associated with increased antibiotic resistance. We found that the same regulatory and inorganic ion transport genes were frequently mutated in persisting clone types within and between Achromobacter species, indicating convergent genetic adaptation. Genome-wide association study of six antibiotic resistance phenotypes revealed the enrichment of associated genes involved in inorganic ion transport, transcription gene enrichment in ß-lactams, and energy production and translation gene enrichment in the trimethoprim/sulfonamide group. Overall, we provide insights into the pathogenomics of Achromobacter spp. infections in patients with CF airways. Since emerging pathogens are increasingly recognized as an important healthcare issue, our findings on evolution of antibiotic resistance and genetic adaptation can facilitate better understanding of disease progression and how mutational changes have implications for patients with CF.


Asunto(s)
Achromobacter/genética , Adaptación Fisiológica/genética , Fibrosis Quística/microbiología , Farmacorresistencia Bacteriana Múltiple/genética , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones del Sistema Respiratorio/microbiología , Achromobacter/efectos de los fármacos , Achromobacter/aislamiento & purificación , Dinamarca , Progresión de la Enfermedad , Metabolismo Energético/genética , Genoma Bacteriano/genética , Estudio de Asociación del Genoma Completo , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Interacciones Huésped-Patógeno , Humanos , Infecciones del Sistema Respiratorio/tratamiento farmacológico
5.
J Clin Microbiol ; 59(4)2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33472899

RESUMEN

Achromobacter species are increasingly being detected in patients with cystic fibrosis (CF), and this emerging pathogen is associated with antibiotic resistance and more-severe disease outcomes. Nonetheless, little is known about the extent of transmission and antibiotic resistance development in Achromobacter infections. We sequenced the genomes of 101 Achromobacter clinical isolates (identified as Achromobacter xylosoxidans based on matrix-assister laser desorption ionization-time of flight [MALDI-TOF] or API N20 typing) collected from 51 patients with CF-the largest longitudinal data set to date. We performed phylogenetic analysis on the genomes and combined this with epidemiological and antibiotic resistance data to identify patient-to-patient transmission and the development of antibiotic resistance. We confirmed that the MALDI-TOF or API N20 method was not sufficient for Achromobacter species-level typing and that the population of Achromobacter isolates was composed of five different species, among which A. xylosoxidans accounted for 52% of infections. Most patients were infected by unique Achromobacter clone types; nonetheless, suspected patient-to-patient transmission cases identified by shared clone types were observed in 35% (n = 18) of patients. In 15 of 16 cases, the suspected transmissions were further supported by genome- or clinic visit-based epidemiological analysis. Finally, we found that resistance developed over time. We show that whole-genome sequencing (WGS) is essential for Achromobacter species typing and identification of patient-to-patient transmission, which was revealed for Achromobacter ruhlandii, A. xylosoxidans, and, for the first time, Achromobacter insuavis Furthermore, we show that the development of antibiotic resistance is associated with chronic Achromobacter infections. Our findings emphasize that transmission and antibiotic resistance should be considered in future treatment strategies.


Asunto(s)
Achromobacter , Fibrosis Quística , Infecciones por Bacterias Gramnegativas , Achromobacter/genética , Fibrosis Quística/complicaciones , Farmacorresistencia Microbiana , Infecciones por Bacterias Gramnegativas/epidemiología , Humanos , Filogenia
6.
mBio ; 11(5)2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33109761

RESUMEN

Genome analyses have documented that there are differences in gene repertoire between evolutionary distant lineages of the same bacterial species; however, less is known about microevolutionary dynamics of gene loss and acquisition within bacterial lineages as they evolve over years. Here, we analyzed the genomes of 45 Pseudomonas aeruginosa lineages evolving in the lungs of cystic fibrosis (CF) patients to identify genes that are lost or acquired during the first years of infection. On average, lineage genome content changed by 88 genes (range, 0 to 473). Genes were more often lost than acquired, and prophage genes were more variable than bacterial genes. We identified convergent loss or acquisition of the same genes across lineages, suggesting selection for loss and acquisition of certain genes in the host environment. We found that a notable proportion of such genes are associated with virulence; a trait previously shown to be important for adaptation. Furthermore, we also compared the genomes across lineages to show that the within-lineage variable genes (i.e., genes that had been lost or acquired during the infection) often belonged to genomic content not shared across all lineages. In sum, our analysis adds to the knowledge on the pace and drivers of gene loss and acquisition in bacteria evolving over years in a human host environment and provides a basis to further understand how gene loss and acquisition play roles in lineage differentiation and host adaptation.IMPORTANCE Bacterial airway infections, predominantly caused by P. aeruginosa, are a major cause of mortality and morbidity of CF patients. While short insertions and deletions as well as point mutations occurring during infection are well studied, there is a lack of understanding of how gene loss and acquisition play roles in bacterial adaptation to the human airways. Here, we investigated P. aeruginosa within-host evolution with regard to gene loss and acquisition. We show that during long-term infection P. aeruginosa genomes tend to lose genes, in particular, genes related to virulence. This adaptive strategy allows reduction of the genome size and evasion of the host's immune response. This knowledge is crucial to understand the basic mutational steps that, on the timescale of years, diversify lineages and adds to the identification of bacterial genetic determinants that have implications for CF disease.


Asunto(s)
Adaptación Fisiológica/genética , Fibrosis Quística/complicaciones , Evolución Molecular , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/genética , Fibrosis Quística/microbiología , Genes Bacterianos , Genoma Bacteriano , Genómica , Humanos , Pulmón/microbiología , Pseudomonas aeruginosa/patogenicidad , Análisis de Secuencia de ADN , Virulencia
7.
Microb Genom ; 6(5)2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32375975

RESUMEN

Antibiotic resistance has become a serious threat to human health (WHO Antibacterial Agents in Clinical Development: an Analysis of the Antibacterial Clinical Development Pipeline, Including Tuberculosis. Geneva: World Health Organization; 2017), and the ability to predict antibiotic resistance from genome sequencing has become a focal point for the medical community. With this genocentric prediction in mind, we were intrigued about two particular findings for a collection of clinical Pseudomonas aeruginosa isolates (Marvig et al. Nature Genetics 2015;47:57-64; Frimodt-Møller et al. Scientific Reports 2018;8:12512; Bartell et al. Nature Communications 2019;10:629): (i) 15 out of 52 genes found to be frequently targeted by adaptive mutations during the initial infection stage of cystic fibrosis airways ('candidate pathoadaptive genes') (Marvig et al. Nature Genetics 2015;47:57-64) were associated with antibiotic resistance (López-Causapé et al. Fronters in Microbiology 2018;9:685; López-Causapé et al. Antimicrobal Agents and Chemotherapy 2018;62:e02583-17); (ii) there was a parallel lack of resistance development and linkage to the genetic changes in these antibiotic-resistance-associated genes (Frimodt-Møller et al. Scientific Reports 2018;8:12512; Bartell et al. Nature Communications 2019;10:629). In this review, we highlight alternative selective forces that potentially enhance the infection success of P. aeruginosa and focus on the linkage to the 15 pathoadaptive antibiotic-resistance-associated genes, thereby showing the problems we may face when using only genomic information to predict and inform about relevant antibiotic treatment.


Asunto(s)
Fibrosis Quística/microbiología , Farmacorresistencia Bacteriana Múltiple , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/genética , Adaptación Fisiológica , Genoma Bacteriano , Humanos , Mutación , Pseudomonas aeruginosa/efectos de los fármacos , Análisis de Secuencia de ADN
9.
Front Microbiol ; 9: 238, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29497413

RESUMEN

The small RNA ErsA of Pseudomonas aeruginosa was previously suggested to be involved in biofilm formation via negative post-transcriptional regulation of the algC gene that encodes the virulence-associated enzyme AlgC, which provides sugar precursors for the synthesis of several polysaccharides. In this study, we show that a knock-out ersA mutant strain forms a flat and uniform biofilm, not characterized by mushroom-multicellular structures typical of a mature biofilm. Conversely, the knock-out mutant strain showed enhanced swarming and twitching motilities. To assess the influence of ErsA on the P. aeruginosa transcriptome, we performed RNA-seq experiments comparing the knock-out mutant with the wild-type. More than 160 genes were found differentially expressed in the knock-out mutant. Parts of these genes, important for biofilm formation and motility regulation, are known to belong also to the AmrZ transcriptional regulator regulon. Here, we show that ErsA binds in vitro and positively regulates amrZ mRNA at post-transcriptional level in vivo suggesting an interesting contribution of the ErsA-amrZ mRNA interaction in biofilm development at several regulatory levels.

10.
Front Microbiol ; 8: 1180, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28690609

RESUMEN

Type II toxin-antitoxin (TA) systems are most commonly composed of two genes encoding a stable toxin, which harms the cell, and an unstable antitoxin that can inactivate it. TA systems were initially characterized as selfish elements, but have recently gained attention for regulating general stress responses responsible for pathogen virulence, formation of drug-tolerant persister cells and biofilms-all implicated in causing recalcitrant chronic infections. We use a bioinformatics approach to explore the distribution and evolution of type II TA loci of the opportunistic pathogen, Pseudomonas aeruginosa, across longitudinally sampled isolates from cystic fibrosis lungs. We identify their location in the genome, mutations, and gain/loss during infection to elucidate their function(s) in stabilizing selfish elements and pathogenesis. We found (1) 26 distinct TA systems, where all isolates harbor four in their core genome and a variable number of the remaining 22 on genomic islands; (2) limited mutations in core genome TA loci, suggesting they are not under negative selection; (3) no evidence for horizontal transmission of elements with TA systems between clone types within patients, despite their ability to mobilize; (4) no gain and limited loss of TA-bearing genomic islands, and of those elements partially lost, the remnant regions carry the TA systems supporting their role in genomic stabilization; (5) no significant correlation between frequency of TA systems and strain ability to establish as chronic infection, but those with a particular TA, are more successful in establishing a chronic infection.

11.
Acta Otolaryngol ; 137(7): 762-764, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28125321

RESUMEN

CONCLUSION: The present study indicates that parotid saliva is sterile on entry to the oral cavity. OBJECTIVES: The objective was to investigate if parotid saliva is sterile on entry to the oral cavity and, thus, prior to contamination by oral bacteria. METHOD: Forty healthy volunteers were included in sterile parotid saliva collection. Parotid saliva was collected using a sterile Lashley cup, placed over the papilla of the Stensen´s duct, as well as sterile tubes and syringes for collection. All collections were followed by collection of a positive control sample where some of the sterile obtained parotid saliva had been exposed to the contralateral mucosal membranes. All samples parotid saliva, as well as the positive controls, were cultivated, and 10 randomly selected parotid saliva samples underwent polymerase chain reaction (PCR) analyses. RESULTS: In 33 of 40 parotid saliva samples there were no cultivable bacteria, whereas bacteria were cultivated in all positive control samples. In eight of 10 PCR samples no bacterial DNA was detected. The most frequent bacteria in the remaining non-sterile parotid saliva samples and positive control samples were non-haemolytical streptococci and the coagulase negative staphylococci.


Asunto(s)
Glándula Parótida/metabolismo , Saliva/microbiología , Adulto , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
12.
BMC Genomics ; 17: 589, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27506816

RESUMEN

BACKGROUND: The primary cause of morbidity and mortality in cystic fibrosis (CF) patients is lung infection by Pseudomonas aeruginosa. Therefore much work has been done to understand the adaptation and evolution of P. aeruginosa in the CF lung. However, many of these studies have focused on longitudinally collected single isolates, and only few have included cross-sectional analyses of entire P. aeruginosa populations in sputum samples. To date only few studies have used the approach of metagenomic analysis for the purpose of investigating P. aeruginosa populations in CF airways. RESULTS: We analysed five metagenomes together with longitudinally collected single isolates from four recently chronically infected CF patients. With this approach we were able to link the clone type and the majority of SNP profiles of the single isolates to that of the metagenome(s) for each individual patient. CONCLUSION: Based on our analysis we find that when having access to comprehensive collections of longitudinal single isolates it is possible to rediscover the genotypes of the single isolates in the metagenomic samples. This suggests that information gained from genome sequencing of comprehensive collections of single isolates is satisfactory for many investigations of adaptation and evolution of P. aeruginosa to the CF airways.


Asunto(s)
Fibrosis Quística/complicaciones , Genotipo , Infecciones por Pseudomonas/etiología , Pseudomonas aeruginosa/genética , Sistema Respiratorio/microbiología , Adolescente , Adulto , Estudios Transversales , Humanos , Metagenoma , Metagenómica/métodos , Filogenia , Polimorfismo de Nucleótido Simple , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/aislamiento & purificación , Sistema Respiratorio/patología , Esputo/microbiología , Adulto Joven
13.
PLoS One ; 8(12): e82869, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24376599

RESUMEN

Regular moderate exercise has been suggested to exert anti-inflammatory effects and improve immune effector functions, resulting in reduced disease incidence and viral infection susceptibility. Whether regular exercise also affects bacterial infection susceptibility is unknown. The aim of this study was to investigate whether regular voluntary exercise wheel running prior to a pulmonary infection with bacteria (P. aeruginosa) affects lung bacteriology, sickness severity and phagocyte immune function in mice. Balb/c mice were randomly placed in a cage with or without a running wheel. After 28 days, mice were intranasally infected with P. aeruginosa. Our study showed that regular exercise resulted in a higher sickness severity score and bacterial (P. aeruginosa) loads in the lungs. The phagocytic capacity of monocytes and neutrophils from spleen and lungs was not affected. Although regular moderate exercise has many health benefits, healthy mice showed increased bacterial (P. aeruginosa) load and symptoms, after regular voluntary exercise, with perseverance of the phagocytic capacity of monocytes and neutrophils. Whether patients, suffering from bacterial infectious diseases, should be encouraged to engage in exercise and physical activities with caution requires further research.


Asunto(s)
Monocitos/inmunología , Neutrófilos/inmunología , Condicionamiento Físico Animal/efectos adversos , Infecciones por Pseudomonas/inmunología , Infecciones del Sistema Respiratorio/inmunología , Bazo/inmunología , Animales , Carga Bacteriana , Susceptibilidad a Enfermedades , Femenino , Ratones , Ratones Endogámicos C57BL , Monocitos/microbiología , Monocitos/patología , Neutrófilos/microbiología , Neutrófilos/patología , Fagocitosis , Infecciones por Pseudomonas/etiología , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/patogenicidad , Pseudomonas aeruginosa/fisiología , Infecciones del Sistema Respiratorio/etiología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/patología , Índice de Severidad de la Enfermedad , Bazo/microbiología , Bazo/patología
14.
Antimicrob Agents Chemother ; 57(1): 177-82, 2013 01.
Artículo en Inglés | MEDLINE | ID: mdl-23089746

RESUMEN

FKS mutant isolates associated with breakthrough or failure cases are emerging in clinical settings. Discrimination of these from wild-type (wt) isolates in a routine laboratory setting is complicated. We evaluated the ability of caspofungin MIC determination using the new Vitek 2 AST-Y06 yeast susceptibility card to correctly identify the fks mutants from wt isolates and compared the performance to those of the CLSI and EUCAST reference methods. A collection of 98 Candida isolates, including 31 fks hot spot mutants, were included. Performance was evaluated using the FKS genotype as the "gold standard" and compared to those of the CLSI and EUCAST methodologies. The categorical agreement for Vitek 2 was 93.9%, compared to 88.4% for the CLSI method and 98.7% for the EUCAST method. Vitek 2 misclassified 19.4% (6/31) of the fks mutant isolates as susceptible, in contrast to <4% for each of the reference methods. The overall essential agreement between the CLSI method and Vitek 2 MICs was 92.6% (88/95) but was substantially lower for fks mutant isolates (78.6% [22/28]). Correct discrimination between susceptible and intermediate Candida glabrata isolates was not possible, as the revised species-specific susceptibility breakpoint was not included in the Vitek 2 detection range (MIC of ≤0.250 to ≥4 mg/liter). In conclusion, the Vitek 2 allowed correct categorization of all wt isolates as susceptible. However, despite an acceptable categorical agreement, it failed to reliably classify isolates harboring fks hot spot mutations as intermediate or resistant, which was in part due to the fact that the detection range did not span the susceptibility breakpoint for C. glabrata.


Asunto(s)
Antifúngicos/farmacología , Candida glabrata/genética , Candida/genética , Candidiasis/tratamiento farmacológico , Equinocandinas/farmacología , Proteínas Fúngicas/genética , Glucosiltransferasas/genética , Candida/clasificación , Candida/efectos de los fármacos , Candida/aislamiento & purificación , Candida glabrata/clasificación , Candida glabrata/efectos de los fármacos , Candida glabrata/aislamiento & purificación , Candidiasis/microbiología , Caspofungina , Farmacorresistencia Fúngica/efectos de los fármacos , Farmacorresistencia Fúngica/genética , Humanos , Lipopéptidos , Pruebas de Sensibilidad Microbiana , Mutación , Tiras Reactivas/normas , Sensibilidad y Especificidad
15.
J Infect Dis ; 206(6): 981-5, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22782947

RESUMEN

Azole resistance in Aspergillus terreus isolates was explored. Twenty related (MB) and 6 unrelated A. terreus isolates were included. CYP51A sequencing and RAPD genotyping was performed. Five MB isolates were itraconazole susceptible, whereas the minimum inhibitory concentrations (MICs) for 15 MB isolates were elevated (1-2 mg/L). Voriconazole and posaconazole MICs were 0.5-4 and 0.06-0.5 mg/L, respectively, for MB isolates but 0.25-0.5 and <0.03-0.06 mg/L, respectively, for controls. Sequencing identified a Cyp51Ap M217I alteration in all 15 isolates with elevated itraconazole MICs. Genotyping showed that 18 of 20 MB isolates were identical and unique, suggesting endogenous origin. In conclusion, itraconazole resistance in A. terreus was linked to an M217I Cyp51A alteration.


Asunto(s)
Antifúngicos/farmacología , Aspergillus/clasificación , Aspergillus/efectos de los fármacos , Azoles/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Farmacorresistencia Fúngica Múltiple/genética , Proteínas Fúngicas/metabolismo , Sistema Enzimático del Citocromo P-450/genética , ADN de Hongos/genética , Proteínas Fúngicas/genética , Regulación Fúngica de la Expresión Génica , Genotipo , Humanos , Itraconazol/farmacología , Pruebas de Sensibilidad Microbiana , Mutación
16.
Antimicrob Agents Chemother ; 56(2): 1019-30, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22106224

RESUMEN

Pseudomonas aeruginosa can develop resistance to polymyxin and other cationic antimicrobial peptides. Previous work has shown that mutations in the PmrAB and PhoPQ regulatory systems can confer low to moderate levels of colistin (polymyxin E) resistance in laboratory strains and clinical isolates of this organism (MICs of 8 to 64 mg/liter). To explore the role of PmrAB in high-level clinical polymyxin resistance, P. aeruginosa isolates from chronically colistin-treated cystic fibrosis patients, most with colistin MICs of >512 mg/liter, were analyzed. These cystic fibrosis isolates contained probable gain-of-function pmrB alleles that conferred polymyxin resistance to strains with a wild-type or pmrAB deletion background. Double mutant pmrB alleles that contained mutations in both the periplasmic and dimerization-phosphotransferase domains markedly augmented polymyxin resistance. Expression of mutant pmrB alleles induced transcription from the promoter of the arnB operon and stimulated addition of 4-amino-l-arabinose to lipid A, consistent with the known role of this lipid A modification in polymyxin resistance. For some highly polymyxin-resistant clinical isolates, repeated passage without antibiotic selection pressure resulted in loss of resistance, suggesting that secondary suppressors occur at a relatively high frequency and account for the instability of this phenotype. These results indicate that pmrB gain-of-function mutations can contribute to high-level polymyxin resistance in clinical strains of P. aeruginosa.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/genética , Fibrosis Quística/microbiología , Farmacorresistencia Bacteriana/genética , Mutación , Polimixinas/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Factores de Transcripción/genética , Proteínas Bacterianas/metabolismo , Colistina/farmacología , Colistina/uso terapéutico , Fibrosis Quística/tratamiento farmacológico , Regulación Bacteriana de la Expresión Génica , Humanos , Pruebas de Sensibilidad Microbiana , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/aislamiento & purificación
17.
Eur J Echocardiogr ; 12(6): 414-20, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21685200

RESUMEN

AIMS: Staphylococcus aureus infective endocarditis (IE) is a critical medical condition associated with a high morbidity and mortality. In the present study, we prospectively evaluated the importance of screening with echocardiography in an unselected S. aureus bacteraemia (SAB) population. METHODS AND RESULTS: From 1 January 2009 to 31 August 2010, a total of 244 patients with SAB at six Danish hospitals underwent screening echocardiography. The inclusion rate was 73% of all eligible patients (n= 336), and 53 of the 244 included patients (22%; 95% CI: 17-27%) were diagnosed with definite IE. In patients with native heart valves the prevalence was 19% (95% CI: 14-25%) compared with 38% (95% CI: 20-55%) in patients with prosthetic heart valves and/or cardiac rhythm management devices (P= 0.02). No difference was found between Main Regional Hospitals and Tertiary Cardiac Hospitals, 20 vs. 23%, respectively (NS). The prevalence of IE in high-risk patients with one or more predisposing condition or clinical evidence of IE were significantly higher compared with low-risk patients with no additional risk factors (38 vs. 5%; P < 0.001). IE was associated with a higher 6 months mortality, 14(26%) vs. 28(15%) in SAB patients without IE, respectively (P < 0.05). CONCLUSION: SAB patients carry a high risk for development of IE, which is associated with a worse prognosis compared with uncomplicated SAB. The presenting symptoms and clinical findings associated with IE are often non-specific and echocardiography should always be considered as part of the initial evaluation of SAB patients.


Asunto(s)
Ecocardiografía/instrumentación , Endocarditis Bacteriana/epidemiología , Infecciones Estafilocócicas/epidemiología , Staphylococcus aureus/aislamiento & purificación , Distribución de Chi-Cuadrado , Intervalos de Confianza , Dinamarca/epidemiología , Endocarditis Bacteriana/diagnóstico por imagen , Humanos , Modelos Logísticos , Tamizaje Masivo , Oportunidad Relativa , Prevalencia , Pronóstico , Estudios Prospectivos , Factores de Riesgo , Infecciones Estafilocócicas/diagnóstico por imagen , Estadística como Asunto , Estadísticas no Paramétricas
19.
BMC Med Res Methodol ; 6: 40, 2006 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-16907965

RESUMEN

BACKGROUND: Disease specific mortality is often used as outcome rather than total mortality in clinical trials. This approach assumes that the classification of cause of death is unbiased. We explored whether use of fungal infection-related mortality as outcome rather than total mortality leads to bias in trials of antifungal agents in cancer patients. METHODS: As an estimate of bias we used relative risk of death in those patients the authors considered had not died from fungal infection. Our sample consisted of 69 trials included in four systematic reviews of prophylactic or empirical antifungal treatment in patients with cancer and neutropenia we have published previously. RESULTS: Thirty trials met the inclusion criteria. The trials comprised 6130 patients and 869 deaths, 220 (25%) of which were ascribed to fungal infection. The relative risk of death was 0.85 (95% CI 0.75-0.96) for total mortality, 0.57 (95% CI 0.44-0.74) for fungal mortality, and 0.95 (95% CI 0.82-1.09) for mortality among those who did not die from fungal infection. CONCLUSION: We could not support the hypothesis that use of disease specific mortality introduces bias in antifungal trials on cancer patients as our estimate of the relative risk for mortality in those who survived the fungal infection was not increased. We conclude that it seems to be reliable to use fungal mortality as the primary outcome in trials of antifungal agents. Data on total mortality should be reported as well, however, to guard against the possible introduction of harmful treatments.


Asunto(s)
Antifúngicos/uso terapéutico , Causas de Muerte , Huésped Inmunocomprometido , Micosis/tratamiento farmacológico , Neoplasias/mortalidad , Neutropenia/mortalidad , Infecciones Oportunistas/mortalidad , Ensayos Clínicos Controlados Aleatorios como Asunto , Antifúngicos/farmacología , Sesgo , Enfermedad Crítica/mortalidad , Humanos , Incidencia , Micosis/complicaciones , Micosis/mortalidad , Neoplasias/complicaciones , Neoplasias/inmunología , Neutropenia/complicaciones , Neutropenia/inmunología , Infecciones Oportunistas/complicaciones , Riesgo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...