Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 198
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Environ Sci Technol ; 58(24): 10806-10816, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38829301

RESUMEN

Temporal and spatial variability of per- and polyfluoroalkyl substances (PFASs) in herring, cod, eelpout, and guillemot covering four decades and more than 1000 km in the Baltic Sea was investigated to evaluate the effect of PFAS regulations and residence times of PFASs. Overall, PFAS concentrations responded rapidly to recent regulations but with some notable basin- and homologue-specific variability. The well-ventilated Kattegat and Bothnian Bay showed a faster log-linear decrease for most PFASs than the Baltic Proper, which lacks a significant loss mechanism. PFOS and FOSA, for example, have decreased with 0-7% y-1 in the Baltic Proper and 6-16% y-1 in other basins. PFNA and partly PFOA are exceptions and continue to show stagnant or increasing concentrations. Further, we found that Bothnian Bay herring contained the highest concentrations of >C12 perfluoroalkyl carboxylic acids (PFCAs), likely from rivers with high loads of dissolved organic carbon. In the Kattegat, low PFAS concentrations, but a high FOSA fraction, could be due to influence from the North Sea inflow below the halocline and possibly a local source of FOSA and/or isomer-specific biotransformation. This study represents the most comprehensive spatial and temporal investigation of PFASs in Baltic wildlife while providing new insights into cycling of PFASs within the Baltic Sea ecosystem.


Asunto(s)
Monitoreo del Ambiente , Fluorocarburos , Contaminantes Químicos del Agua , Contaminantes Químicos del Agua/análisis , Fluorocarburos/análisis , Océanos y Mares , Animales
2.
Euro Surveill ; 29(21)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38785090

RESUMEN

Fosfomycin-resistant FosA8-producing Enterobacterales are uncommon strains with extremely low incidence in Europe, based on only three reports in the literature. We detected FosA8-producing Escherichia coli ST131 in clinical isolates from two patients admitted in February 2023 to a rehabilitation unit in Italy. The occurrence of rare fosA-like genes in the high-risk clone ST131 is of clinical relevance. The dissemination of FosA-producing E. coli, although still at low levels, should be continuously monitored.


Asunto(s)
Antibacterianos , Infecciones por Escherichia coli , Escherichia coli , Humanos , Italia/epidemiología , Escherichia coli/aislamiento & purificación , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/epidemiología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Pruebas de Sensibilidad Microbiana , Fosfomicina/farmacología , Fosfomicina/uso terapéutico , Masculino , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Femenino , Farmacorresistencia Bacteriana , Tipificación de Secuencias Multilocus
3.
Chem Biodivers ; 20(12): e202301567, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37956152

RESUMEN

The presence of multidrug-resistant pathogenic microorganisms makes it challenging to cure bacterial illnesses. Syzygium aromaticum has been used for medicinal purposes since ancient times. The objective of this study was to investigate the potential synergistic effect of the combination of Eugenol and Fosfomycin against clinically Uropathogenic Escherichia coli (UPEC) and their possible co-treatment as well as their contribution to plasmid-mediated Fosfomycin resistance (fosA3 and fosA4) genes using molecular assays. Eugenol was extracted from clove (Syzygium aromaticum) plants using steam distillation by Clevenger and analyzed by high-performance liquid chromatography (HPLC). UPEC accounted for 63.6 % of all isolates. Specifically, 99.3 % of the UPEC isolates exhibited resistance to multiple types of antibiotics [multidrug-resistant (MDR)]. The MIC for Eugenol was 1.25-5 µg/mL, and Fosfomycin was 512-1024 µg/mL, while the MBC for Eugenol was 5-10 µg/mL and Fosfomycin was 2048 µg/mL. The synergistic effects were considerable, with 1/4 MIC of Eugenol resulting in 1/8 MIC Fosfomycin. Eugenol inhibited most of the UPEC isolates at 4-8 hours, Fosfomycin at 8-12 hours, and co-treatment at 4-8 hours. The fosA3 and fosA4 genes were detected in 5.7 % and 2.9 % of the isolates, respectively. The results showed variable gene expression changes in response to the different treatments.


Asunto(s)
Infecciones por Escherichia coli , Fosfomicina , Humanos , Fosfomicina/farmacología , Escherichia coli/genética , Eugenol/farmacología , Farmacorresistencia Bacteriana , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , beta-Lactamasas/farmacología , Plásmidos , Antibacterianos/farmacología , Pruebas de Sensibilidad Microbiana , Infecciones por Escherichia coli/microbiología
4.
Plasmid ; 123-124: 102651, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36191658

RESUMEN

To characterize IncI1 and IncF18:A-:B1 multidrug-resistance plasmids from an avian Escherichia coli isolate, antibiotic susceptibility testing, conjugation assays, transformation assays, S1-PFGE, and WGS analysis were performed. The 119,457-bp plasmid pEC014-1 with a multidrug-resistance region (MRR) containing four different segments interspersed with six IS26 elements, belonged to incompatibility group I1 and sequence type 71. The 154,516-bp plasmid pEC014-2 with two replicons, typed as FII-18 and FIB-1, carried 14 resistance determinants including blaTEM-1b, blaOXA-1, oqxAB, dfrA17, aac(6')-Ib-cr, sul1, sul2, tet(A), floR, catB3, hph(aph(4)-Ia), aacC4(aac(3)-IV), aadA5, arr-3, and a merEDACPTR loci in MRR, and additionally encoded three virulence loci: iroNEDCB, sitABCD, and iucABCD-iutA. Plasmid stability assays showed that pEC014-1 and pEC014-2 were stable in recipient E. coli C600 for at least 15 days of passage. Competition assays were carried out to evaluate the fitness impact of pEC014-2 carriage in vitro, revealing a decrease in host fitness. Growth kinetics showed that the growth rate for pEC014-1 or/and pEC014-2 bearing cells was significantly slower than that of the E. coli C600 host strain in the exponential stage (p < 0.01), with only cells carrying pEC014-1 sustaining rapid growth after 6 h of exponential growth. Our findings highlight the mosaic structures of epidemic plasmid IncI1/ST71 and F18:A-:B1 lineages and contribute to a better understanding of the evolution and dissemination of these multidrug resistance and virulence plasmids.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli , Humanos , Plásmidos/genética , beta-Lactamasas/genética , Pruebas de Sensibilidad Microbiana , Infecciones por Escherichia coli/epidemiología , Resistencia a Múltiples Medicamentos , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/genética
5.
Int J Mol Sci ; 23(16)2022 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-36012441

RESUMEN

Antimicrobial resistance is a public health burden with worldwide impacts and was recently identified as one of the major causes of death in 2019. Fosfomycin is an antibiotic commonly used to treat urinary tract infections, and resistance to it in Enterobacteriaceae is mainly due to the metalloenzyme FosA3 encoded by the fosA3 gene. In this work, we adapted a CRISPR-Cas9 system named pRE-FOSA3 to restore the sensitivity of a fosA3+ Escherichia coli strain. The fosA3+ E. coli strain was generated by transforming synthetic fosA3 into a nonpathogenic E. coli TOP10. To mediate the fosA3 disruption, two guide RNAs (gRNAs) were selected that used conserved regions within the fosA3 sequence of more than 700 fosA3+ E. coli isolates, and the resensitization plasmid pRE-FOSA3 was assembled by cloning the gRNA into pCas9. gRNA_195 exhibited 100% efficiency in resensitizing the bacteria to fosfomycin. Additionally, the edited strain lost the ampicillin resistance encoded in the same plasmid containing the synthetic fosA3 gene, despite not being the CRISPR-Cas9 target, indicating plasmid clearance. The in vitro analysis presented here points to a path that can be explored to assist the development of effective alternative methods of treatment against fosA3+ bacteria.


Asunto(s)
Infecciones por Escherichia coli , Fosfomicina , Antibacterianos/farmacología , Farmacorresistencia Bacteriana/genética , Escherichia coli , Infecciones por Escherichia coli/microbiología , Fosfomicina/farmacología , Humanos , Pruebas de Sensibilidad Microbiana , Plásmidos/genética , ARN Guía de Kinetoplastida , beta-Lactamasas/genética
6.
Rev Argent Microbiol ; 54(2): 120-124, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34154873

RESUMEN

Fosfomycin tromethamol (FT) was reintroduced as an option for the treatment of low urinary tract infection (UTI) in children. In this study, we described the antibiotic sensitivity and mechanisms of resistance to fosfomycin in isolates from children older than 6 years with UTI. Urine culture and antibiotic susceptibility study were performed. In fosfomycin resistant strains, PCR for fos, blaCTX-M was performed followed by classification by phylogenetic group and sequencetyping. Escherichia coli was the most frequent etiological agent (89.2%). The susceptibility percentages were: fosfomycin 97.9%; amoxicillin-clavulanate 92.7%; cefuroxime and ceftriaxone 99%; nitrofurantoin 94.4%. An E. coli strain (ST69, phylogenetic group D) was resistant to fosfomycin (MIC 256mg/l) and carried the blaCTX-M-14 and fosA3 genes in a 45kb IncN-type plasmid. This is the first report of E. coli ST69 with blaCTX-M-14/fosA3 of human origin.


Asunto(s)
Infecciones por Escherichia coli , Fosfomicina , Infecciones Urinarias , Antibacterianos/farmacología , Niño , Farmacorresistencia Bacteriana , Escherichia coli/genética , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/epidemiología , Fosfomicina/farmacología , Fosfomicina/uso terapéutico , Humanos , Pruebas de Sensibilidad Microbiana , Filogenia , Infecciones Urinarias/tratamiento farmacológico , beta-Lactamasas/genética
7.
Artículo en Inglés | MEDLINE | ID: mdl-33820767

RESUMEN

The plasmid-mediated tet(X7) conferring high-level tigecycline resistance was identified in five mcr-1.1-positive Escherichia coli strains (ST10 [n = 3] and ST155 [n = 2]) isolated from chickens in Egypt. Two fosfomycin-resistant fosA4-carrying IncFII plasmids (∼79 kb in size) were detected. Transposase ISCR3 (IS91 family) is syntenic with tet(X7) in all isolates, suggesting its role in the mobilization of tet(X7). To our knowledge, this is the first global report of ST4-IncHI2 plasmids cocarrying tet(X7) and mcr-1.1 from chickens.


Asunto(s)
Infecciones por Escherichia coli , Proteínas de Escherichia coli , Fosfomicina , Animales , Antibacterianos/farmacología , Pollos , Colistina/farmacología , Farmacorresistencia Bacteriana/genética , Egipto , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Fosfomicina/farmacología , Plásmidos/genética , Tigeciclina
8.
BMC Microbiol ; 21(1): 109, 2021 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-33838639

RESUMEN

BACKGROUND: Fosfomycin has been proven to be a vital choice to treat infection caused by multidrug resistance bacteria, especially carbapenem-resistant Klebsiella pneumoniae (CRKP). However, fosfomycin resistant cases has been reported gradually. In this study, we reported the fosfomycin-resistant rate in CRKP strains and further revealed the molecular mechanisms in resistance gene dissemination. RESULTS: A total of 294 non-duplicated CRKP strains were collected. And 55 fosfomyin-resistant strains were detected, 94.5% of which were clustered to sequence type (ST) 11 by PCR followed up sequencing. PFGE further revealed two major groups and four singletons. The positive rates of genes responsible to fosfomycin and carbapenem resistance were 81.8% (fosA3), 12.7% (fosA5) and 94.5% (blaKPC-2), respectively. Genomic analysis confirmed insertion sequence (IS) 26 was the predominant structure surrounding fosA3. The fosA3 genes in six isolates were located on plasmids which were able to transfer to E. coli J53 recipient cells by means of conjugation. CONCLUSIONS: Although the resistant rate of CRKP to fosfomycin is relatively low in our area, considering its gene is located on transferrable plasmid and inserted in IS structure, continuous monitoring is still needed.


Asunto(s)
Enterobacteriaceae Resistentes a los Carbapenémicos/genética , Farmacorresistencia Bacteriana/genética , Fosfomicina/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Epidemiología Molecular , China , Humanos , Infecciones por Klebsiella/microbiología , Centros de Atención Terciaria
9.
Plasmid ; 114: 102555, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33472047

RESUMEN

To analyze characteristics and underlying evolutionary processes of IncC and IncI1 plasmids in a multidrug-resistant avian E. coli strain, antibiotic susceptibility testing, PCR, conjugation assays, and next-generation sequencing were performed. The type 1 IncC plasmid pEC009.1 harbored three antimicrobial resistance regions including ISEcp1-blaCMY-2-blc-sugE, ARI-B resistance island, and ARI-A island that was a mosaic multidrug resistance region (MRR) comprised of a class 1 integron with cassette array |aac(6')-II(aacA7)|qacE∆1|sul1|, IS26-mphR(A)-mrx-mph(A)-IS26, IS26-fosA3-IS26, and mercury resistance cluster merRTPABDE. It is the first report of three different size circular forms derived from IS26-mphR(A)-mrx-mph(A)-IS26-fosA3-IS26 in ARI-A of type 1 IncC plasmid. In IncI1/ST136 pEC009.2, the truncated transposon Tn1722 carrying blaTEM-1b, rmtB, aac(3)-IId(aacC2d), and a class 1 integron with cassette array |dfrA12|orfF|aadA2|, inserted into the plasmid backbone generating 5-bp direct repeats (DRs, TATAA) at the boundaries of the region, which was highly similar to that of other IncI1 plasmids, and differed by the arrangements of resistance determinants. Comparison among two epidemic plasmid lineages showed complex MRRs respectively located in the specific position in type 1 IncC and IncI1/ST136 plasmids with conserved backbones, and these have evolved via multiple events involved in mobile elements-mediated loss and gain of resistance genes and accessory genes. Strains harboring these plasmids may serve as a reservoir for antibiotic resistance genes, thereby contributing to the rapid spread of resistance genes and posing a public health threat.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Antibacterianos/farmacología , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Metiltransferasas , Plásmidos/genética , beta-Lactamasas/genética
10.
Artículo en Inglés | MEDLINE | ID: mdl-33077665

RESUMEN

Here, we characterize the fosA genes from three Escherichia coli clinical isolates recovered from Canadian patients. Each fosA sequence was individually overexpressed in E. coli BW25113, and antimicrobial susceptibility testing was performed to assess their role in fosfomycin resistance. The findings from this study identify and functionally characterize FosA3, FosA8, and novel FosA7 members and highlight the importance of phenotypic characterization of fosA genes.


Asunto(s)
Infecciones por Escherichia coli , Fosfomicina , Antibacterianos/farmacología , Canadá , Farmacorresistencia Bacteriana/genética , Escherichia coli/genética , Infecciones por Escherichia coli/tratamiento farmacológico , Fosfomicina/farmacología , Hospitales , Humanos , Pruebas de Sensibilidad Microbiana , Plásmidos , beta-Lactamasas/genética
11.
Artículo en Inglés | MEDLINE | ID: mdl-32122889

RESUMEN

A fosfomycin-resistant and carbapenemase (OXA-48)-producing Klebsiella pneumoniae isolate was recovered, and whole-genome sequencing revealed ISEcp1-blaCTX-M-14b tandemly inserted upstream of the chromosomally encoded lysR-fosA locus. Quantitative evaluation of the expression of lysR and fosA genes showed that this insertion brought a strong hybrid promoter leading to overexpression of the fosA gene, resulting in fosfomycin resistance. This work showed the concomitant acquisition of resistance to broad-spectrum cephalosporins and fosfomycin due to a single genetic event.


Asunto(s)
Antibacterianos/farmacología , Cefalosporinas/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Fosfomicina/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Proteínas Bacterianas/genética , Elementos Transponibles de ADN/genética , Genoma Bacteriano/genética , Humanos , Klebsiella pneumoniae/aislamiento & purificación , Pruebas de Sensibilidad Microbiana , Mutagénesis Insercional/genética , Factores de Transcripción/genética , Secuenciación Completa del Genoma , beta-Lactamasas/genética
12.
Artículo en Inglés | MEDLINE | ID: mdl-31481445

RESUMEN

A plasmid-located fosfomycin resistance gene, fosA8, was identified from a CTX-M-15-producing Escherichia coli isolate recovered from urine. Identification of this gene was obtained by whole-genome sequencing. It encoded FosA8, which shares 79% and 78% amino acid identity with the most closely related FosA2 and FosA1 enzymes, respectively. The fosA8 gene was located on a transferable 50-kb plasmid of IncN type encoding high-level resistance to fosfomycin. In silico analysis and cloning experiments identified fosA8 analogues (99% identity) in the genome of Leclercia decarboxylata, which is an enterobacterial species with natural resistance to fosfomycin. This finding adds L. decarboxylata to the list of enterobacterial species that are a reservoir of fosA-like genes which have been captured from the chromosome of a progenitor and are then acquired by E. coli.


Asunto(s)
Antibacterianos/farmacología , Enterobacteriaceae/genética , Proteínas de Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Fosfomicina/farmacología , Genes Bacterianos/genética , Plásmidos/genética , Clonación Molecular , Farmacorresistencia Bacteriana/genética , Enterobacteriaceae/efectos de los fármacos , Escherichia coli/genética , Filogenia , Secuenciación Completa del Genoma
13.
Artículo en Inglés | MEDLINE | ID: mdl-30642934

RESUMEN

The spread of multidrug or extensively drug-resistant Gram-negative bacteria is a serious public health issue. There are too few new antibiotics in development to combat the threat of multidrug-resistant infections, and consequently the rate of increasing antibiotic resistance is outpacing the drug development process. This fundamentally threatens our ability to treat common infectious diseases. Fosfomycin (FOM) has an established track record of safety in humans and is highly active against Escherichia coli, including multidrug-resistant strains. However, many other Gram-negative pathogens, including the "priority pathogens" Klebsiella pneumoniae and Pseudomonas aeruginosa, are inherently resistant to FOM due to the chromosomal fosA gene, which directs expression of a metal-dependent glutathione S-transferase (FosA) that metabolizes FOM. In this study, we describe the discovery and biochemical and structural characterization of ANY1 (3-bromo-6-[3-(3-bromo-2-oxo-1H-pyrazolo[1,5-a]pyrimidin-6-yl)-4-nitro-1H-pyrazol-5-yl]-1H-pyrazolo[1,5-a]pyrimidin-2-one), a small-molecule active-site inhibitor of FosA. Importantly, ANY1 potentiates FOM activity in representative Gram-negative pathogens. Collectively, our study outlines a new strategy to expand FOM activity to a broader spectrum of Gram-negative pathogens, including multidrug-resistant strains.


Asunto(s)
Antibacterianos/farmacología , Proteínas de Escherichia coli/antagonistas & inhibidores , Escherichia coli/efectos de los fármacos , Fosfomicina/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Farmacorresistencia Bacteriana/fisiología , Farmacorresistencia Bacteriana Múltiple , Humanos , Pruebas de Sensibilidad Microbiana
14.
J Clin Microbiol ; 57(10)2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31340992

RESUMEN

With multidrug-resistant (MDR) Enterobacterales on the rise, a nontoxic antimicrobial agent with a unique mechanism of action such as fosfomycin seems attractive. However, establishing accurate fosfomycin susceptibility testing for non-Escherichia coli isolates in a clinical microbiology laboratory remains problematic. We evaluated fosfomycin susceptibility by multiple methods with 96 KPC-producing clinical isolates of multiple strains and species collected at a single center between 2008 and 2016. In addition, we assessed the presence of fosfomycin resistance genes from whole-genome sequencing (WGS) data using NCBI's AMRFinder and custom HMM search. Susceptibility testing was performed using a glucose-6-phosphate-supplemented fosfomycin Etest and Kirby-Bauer disk diffusion (DD) assays, and the results were compared to those obtained by agar dilution. Clinical Laboratory and Standards Institute (CLSI) breakpoints for E. coli were applied for interpretation. Overall, 63% (60/96) of isolates were susceptible by Etest, 70% (67/96) by DD, and 88% (84/96) by agar dilution. fosA was detected in 80% (70/88) of previously sequenced isolates, with species-specific associations and alleles, and fosA-positive isolates were associated with higher MIC distributions. Disk potentiation testing was performed using sodium phosphonoformate to inhibit fosA and showed significant increases in the zone diameter of DD testing for isolates that were fosA positive compared to those that were fosA negative. The addition of sodium phosphonoformate (PPF) corrected 10/14 (71%) major errors in categorical agreement with agar dilution. Our results indicate that fosA influences the inaccuracy of susceptibility testing by methods readily available in a clinical laboratory compared to agar dilution. Further research is needed to determine the impact of fosA on clinical outcomes.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana , Fosfomicina/farmacología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , beta-Lactamasas/genética , Proteínas Bacterianas/biosíntesis , Genoma Bacteriano , Humanos , Klebsiella pneumoniae/aislamiento & purificación , Pruebas de Sensibilidad Microbiana , Secuenciación Completa del Genoma , beta-Lactamasas/biosíntesis
15.
Artículo en Inglés | MEDLINE | ID: mdl-29866858

RESUMEN

A putative fosA gene in Kluyvera georgiana 14751 showed 99% nucleotide identity with plasmid-encoded fosA4 Due to a single-nucleotide insertion translating to a truncated protein, K. georgiana 14751 fosA does not confer fosfomycin resistance. However, analysis of another genome deposit (Kluyvera ascorbata WCH1410) that could be recategorized as K. georgiana after phylogenetic analysis revealed a fosA gene 100% identical to the plasmid-borne fosA4 gene. We suggest that Kluyvera georgiana represents the most probable origin of fosA4.


Asunto(s)
Antibacterianos/farmacología , Kluyvera/efectos de los fármacos , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana/genética , Fosfomicina/farmacología , Kluyvera/genética , Pruebas de Sensibilidad Microbiana , Filogenia , Plásmidos/genética
16.
Artículo en Inglés | MEDLINE | ID: mdl-29967018

RESUMEN

A novel 139,487-bp SXT/R391 integrative and conjugative element, ICEPmiChnBCP11, was characterized in Proteus mirabilis of swine origin in China. ICEPmiChnBCP11 harbors 20 different antimicrobial resistance genes, including the clinically important rRNA methyltransferase gene cfr, the extended-spectrum ß-lactamase gene blaCTX-M-65, fosfomycin resistance gene fosA3, and fluoroquinolone resistance gene aac(6')-Ib-cr An ISPpu12-mediated composite transposon containing various resistance genes and 10 copies of IS26 is inserted in hot spot 4. ICEPmiChnBCP11 was successfully transferred to Escherichia coli.


Asunto(s)
Conjugación Genética/genética , Elementos Transponibles de ADN/genética , Farmacorresistencia Bacteriana Múltiple/genética , Transferencia de Gen Horizontal/genética , Proteus mirabilis/efectos de los fármacos , Proteus mirabilis/genética , Animales , China , ADN Bacteriano/genética , Escherichia coli/genética , Fluoroquinolonas/farmacología , Fosfomicina/farmacología , Metiltransferasas/genética , Pruebas de Sensibilidad Microbiana , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteus mirabilis/aislamiento & purificación , Porcinos , beta-Lactamasas/genética
17.
Ecotoxicol Environ Saf ; 161: 669-675, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29935431

RESUMEN

Perfluorooctane sulfonamide (FOSA) is an important perfluorooctane sulfonate (PFOS) precursor used for commercial applications. In order to investigate the transformation and responses of selected antioxidant and degradation enzymes of FOSA in the plants, in vivo exposure of soybean (Glycine max L. Merrill) and pumpkin (Cucurbita maxima L.) were conducted in the solution-plant microcosms. FOSA was readily taken up by soybean and pumpkin roots and translocated to shoots, and metabolized to PFOS, perfluorohexane sulfonate (PFHxS) and perfluorobutane sulfonate (PFBS). Although morphological and biomass effects were not visible, significant changes in oxidative stress response were observed except for thiobarbituric acid reactive substances (TBARS). Superoxide dismutase (SOD) and peroxidase (POD) activities were significantly increased by 19.2-30.8% and 19.2-20.7% in soybean (8-12 d) respectively, and increased by 39.2-92.8% and 21.1-37.6% in pumpkin (3-12 d) respectively, suggesting an activation of the antioxidant defense system in the plants exposed to FOSA. Glutathione-S-transferase (GST) activities were decreased in soybean (2-12 d) with 9.0-36.1% inhibition and increased in pumpkin (3-12 d) with 22.5-47.3% activation respectively; cytochrome P450 (CYP450) activities were increased markedly in soybean and pumpkin with 13.2-53.6% and 26.7-50.2% activation respectively, giving indirect evidences on the involvement of CYP450 and GST in degradation of FOSA in plants.


Asunto(s)
Cucurbita/metabolismo , Contaminantes Ambientales/farmacocinética , Fluorocarburos/farmacocinética , Glycine max/metabolismo , Sulfonamidas/farmacocinética , Ácidos Alcanesulfónicos/metabolismo , Antioxidantes , Biotransformación , Cucurbita/enzimología , Fluorocarburos/metabolismo , Hidroponía , Estrés Oxidativo , Glycine max/enzimología , Ácidos Sulfónicos/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
18.
J Cell Biochem ; 118(11): 4088-4094, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28409871

RESUMEN

Klebsiella pneumoniae, BA6753 was cultured from a patient in the Clinical Microbiology Laboratory of Christian Medical College. K. pneumoniae, BA6753 has a multidrug resistance plasmid encoding novel FosA variant-7, fosfomycin resistance enzyme. Minimal side effects and a wide range of bactericidal activity of fosfomycin have resulted in its expanded clinical use that prompts the rise of fosfomycin-resistant strains. At present, there are no effective inhibitors available to conflict the FosA-medicated fosfomycin resistance. To develop effective FosA inhibitors, it is crucial to understand the structural and dynamic properties of resistance enzymes. Hence, the present study focuses on the identification of potent inhibitors that can effectively bind to the fosfomycin resistance enzyme, thus predispose the target to inactivate by the second antibiotic. Initially, a series of active compounds were screened against the resistant enzyme, and the binding affinities were confirmed using docking simulation analysis. For efficient activity, the binding affinity of the resistance enzyme ought to be high with the inhibitor than the fosfomycin drug. Consequently, the enzyme-ligand complex which showed higher binding affinity than the fosfomycin was employed for subsequent analysis. The stability of the top scoring enzyme-ligand complex was further validated using molecular dynamics simulation studies. On the whole, we presume that the compound 19583672 demonstrates a higher binding affinity for the resistance enzyme comparing to other compounds and fosfomycin. We believe that further enhancement of the lead compound can serve as a potential inhibitor against resistance enzyme in drug discovery process. J. Cell. Biochem. 118: 4088-4094, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Proteínas Bacterianas/química , Farmacorresistencia Bacteriana , Fosfomicina , Klebsiella pneumoniae/enzimología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Humanos , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/aislamiento & purificación
19.
Emerg Infect Dis ; 23(9): 1564-1567, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28820368
20.
Artículo en Inglés | MEDLINE | ID: mdl-28874374

RESUMEN

Fosfomycin exhibits broad-spectrum antibacterial activity and is being reevaluated for the treatment of extensively drug-resistant pathogens. Its activity in Gram-negative organisms, however, can be compromised by expression of FosA, a metal-dependent transferase that catalyzes the conjugation of glutathione to fosfomycin, rendering the antibiotic inactive. In this study, we solved the crystal structures of two of the most clinically relevant FosA enzymes: plasmid-encoded FosA3 from Escherichia coli and chromosomally encoded FosA from Klebsiella pneumoniae (FosAKP). The structure, molecular dynamics, catalytic activity, and fosfomycin resistance of FosA3 and FosAKP were also compared to those of FosA from Pseudomonas aeruginosa (FosAPA), for which prior crystal structures exist. E. coli TOP10 transformants expressing FosA3 and FosAKP conferred significantly greater fosfomycin resistance (MIC, >1,024 µg/ml) than those expressing FosAPA (MIC, 16 µg/ml), which could be explained in part by the higher catalytic efficiencies of the FosA3 and FosAKP enzymes. Interestingly, these differences in enzyme activity could not be attributed to structural differences at their active sites. Instead, molecular dynamics simulations and hydrogen-deuterium exchange experiments with FosAKP revealed dynamic interconnectivity between its active sites and a loop structure that extends from the active site of each monomer and traverses the dimer interface. This dimer interface loop is longer and more extended in FosAKP and FosA3 than in FosAPA, and kinetic analyses of FosAKP and FosAPA loop-swapped chimeric enzymes highlighted its importance in FosA activity. Collectively, these data yield novel insights into fosfomycin resistance that could be leveraged to develop new strategies to inhibit FosA and potentiate fosfomycin activity.


Asunto(s)
Farmacorresistencia Bacteriana/fisiología , Proteínas de Escherichia coli/química , Escherichia coli/efectos de los fármacos , Fosfomicina/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Dominio Catalítico , Cristalografía por Rayos X , Medición de Intercambio de Deuterio , Farmacorresistencia Bacteriana/efectos de los fármacos , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Klebsiella pneumoniae/genética , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Potasio/metabolismo , Multimerización de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA