339 research outputs found

    Proposing Kluyvera georgiana as the Origin of the Plasmid-Mediated Resistance Gene fosA4

    Get PDF
    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.Fil: Rodriguez, Maria Margarita. Universidad de Buenos Aires. Facultad de Farmacia y BioquĂ­mica. Departamento de MicrobiologĂ­a, InmunologĂ­a y BiotecnologĂ­a. CĂĄtedra de MicrobiologĂ­a; Argentina. Consejo Nacional de Investigaciones CientĂ­ficas y TĂ©cnicas; ArgentinaFil: Ghiglione, Barbara. Consejo Nacional de Investigaciones CientĂ­ficas y TĂ©cnicas; Argentina. Universidad de Buenos Aires. Facultad de Farmacia y BioquĂ­mica. Departamento de MicrobiologĂ­a, InmunologĂ­a y BiotecnologĂ­a. CĂĄtedra de MicrobiologĂ­a; ArgentinaFil: Power, Pablo. Consejo Nacional de Investigaciones CientĂ­ficas y TĂ©cnicas; Argentina. Universidad de Buenos Aires. Facultad de Farmacia y BioquĂ­mica. Departamento de MicrobiologĂ­a, InmunologĂ­a y BiotecnologĂ­a. CĂĄtedra de MicrobiologĂ­a; ArgentinaFil: Naas, Thierry. HĂŽpital de BicĂȘtre. Service de BactĂ©riologie HygiĂšne; FranciaFil: Gutkind, Gabriel Osvaldo. Universidad de Buenos Aires. Facultad de Farmacia y BioquĂ­mica. Departamento de MicrobiologĂ­a, InmunologĂ­a y BiotecnologĂ­a. CĂĄtedra de MicrobiologĂ­a; Argentina. Consejo Nacional de Investigaciones CientĂ­ficas y TĂ©cnicas; Argentin

    Complete Sequence of a F33:A-:B- Conjugative Plasmid Carrying the oqxAB, fosA3, and blaCTX-M-55 Elements from a Foodborne Escherichia coli Strain

    Get PDF
    This study reports the complete sequence of pE80, a conjugative IncFII plasmid recovered from an Escherichia coli strain isolated from chicken meat. This plasmid harbors multiple resistance determinants including oqxAB, fosA3, blaCTX-M-55, and blaTEM-1, and is a close variant of the recently reported p42-2 element, which was recovered from E. coli of veterinary source. Recovery of pE80 constitutes evidence that evolution or genetic re-arrangement of IncFII type plasmids residing in animal-borne organisms is an active event, which involves acquisition and integration of foreign resistance elements into the plasmid backbone. Dissemination of these plasmids may further compromise the effectiveness of current antimicrobial strategies.Department of Applied Biology and Chemical Technolog

    Plasmid-mediated fosfomycin resistance in Escherichia coli isolated from pig.

    Get PDF
    Previous studies have reported plasmid-mediated fosA3 among Escherichia coli originating from human and companion animals. In this study, the plasmid, designated pHK23a originating from a multidrug-resistant E. coli isolate recovered from a slaughter pig in December 2008 in Hong Kong, China was sequenced. In conjugation, the plasmid readily transferred to E. coli J53 at high frequencies. It belongs to the narrow host range IncFII incompatibility group and is 73,607bp in length. Sequence alignment showed that pHK23a has a 59.1kb backbone which shares high homology with the prototype R100 plasmid and a 14.5kb variable region. The variable region includes three genes mediating antimicrobial resistance (fosA3, ΔblaTEM-1, blaCTX-M-3), ten mobile genetic elements (four copies of IS26, insA, ΔinsB, ΔTn2, IS1, ΔISEcp1, Δintl1), the tir transfer inhibition protein, the pemI/pemK addiction system and eight ORFs of unknown functions (orf1, orf2, Δorf3, orf20, orf23, orf24, ycdA and ycdB). The three resistance genes were organized in a novel IS26-composite transposon-like structure. In conclusion, this is the first report of fosA3 containing plasmid in an isolate of pig origin. Since IncFII plasmids spread efficiently in Enterobacteriaceae, the detection of fosA3 with blaCTX-M is worrisome and might become a public health concern. © 2012 Elsevier B.V..postprin

    Characterization of a multidrug-resistant porcine Klebsiella pneumoniae sequence type 11 strain coharboring blaKPC-2 and fosA3 on two novel hybrid plasmids

    Get PDF
    The occurrence of carbapenemase-producing Enterobacteriaceae (CPE) poses a considerable risk for public health. The gene for Klebsiella pneumoniae carbapenemase-2 (KPC-2) has been reported in many countries worldwide, and KPC-2-producing strains are mainly of human origin. In this study, we identified two novel hybrid plasmids that carry either blaKPC-2 or the fosfomycin resistance gene fosA3 in the multiresistant K. pneumoniae isolate K15 of swine origin in China. The blaKPC-2-bearing plasmid pK15-KPC was a fusion derivative of an IncF33:A−:B− incompatibility group (Inc) plasmid and chromosomal sequences of K. pneumoniae (CSKP). A 5-bp direct target sequence duplication (GACTA) was identified at the boundaries of the CSKP, suggesting that the integration might have been due to a transposition event. The blaKPC-2 gene on pK15-KPC was in a derivative of ΔTn6296-1. The multireplicon fosA3-carrying IncN-IncR plasmid pK15-FOS also showed a mosaic structure, possibly originating from a recombination between an epidemic fosA3-carrying pHN7A8-like plasmid and a pKPC-LK30-like IncR plasmid. Stability tests demonstrated that both novel hybrid plasmids were stably maintained in the original host without antibiotic selection but were lost from the transformants after approximately 200 generations. This is apparently the first description of a porcine sequence type 11 (ST11) K. pneumoniae isolate coproducing KPC-2 and FosA3 via pK15-KPC and pK15-FOS, respectively. The multidrug resistance (MDR) phenotype of this high-risk K. pneumoniae isolate may contribute to its spread and its persistence

    High Incidence and Endemic Spread of NDM-1-Positive Enterobacteriaceae in Henan Province, China

    Get PDF
    The emergence and spread of New Delhi metallo-ÎČ-lactamase 1 (NDM-1)-producing carbapenem-resistant Enterobacteriaceae (CRE) present an urgent threat to human health. In China, the blaNDM-1 gene has been reported mostly in Acinetobacter spp. but is rarely found in Enterobacteriaceae. Here, we report a high incidence and endemic spread of NDM-1-producing CRE in Henan Province in China. Sixteen (33.3%) of the 48 CRE isolates obtained from patients during June 2011 to July 2012 were positive for blaNDM-1, and the gene was found to be carried on plasmids of various sizes (∌55 to ∌360 kb). These plasmids were readily transferrable to recipient Escherichia coli by conjugation, conferred resistance to multiple antibiotics, and belonged to multiple replicon types. The blaNDM-1-positive CRE isolates were genetically diverse, and six new multilocus sequence typing (MLST) sequence types were linked to the carriage of NDM-1. Five of the isolates were classified as extensively drug-resistant (XDR) isolates, four of which also carried the fosA3 gene conferring resistance to fosfomycin, an alternative drug for treating infections by CRE. In each blaNDM-1-positive CRE isolate, the blaNDM-1 gene was downstream of an intact ISAba125 element and upstream of the bleMBL gene. Furthermore, gene environment analysis suggested the possible transmission of blaNDM-1-containing sequences from Acinetobacter spp. to Klebsiella pneumoniae and Klebsiella oxytoca. These findings reveal the emergence and active transmission of NDM-1-positive CRE in China and underscore the need for heightened measures to control their further spread

    Complete sequence of the FII plasmid p42-2, carrying blaCTX-M-55, oqxAB, fosA3, and floR from Escherichia coli

    Get PDF
    We sequenced a novel conjugative multidrug resistance IncF plasmid, p42-2, isolated from Escherichia coli strain 42-2, previously identified in China. p42-2 is 106,886 bp long, composed of a typical IncFII-type backbone (∌54 kb) and one distinct acquired DNA region spanning ∌53 kb, harboring 12 antibiotic resistance genes [blaCTX-M-55, oqxA, oqxB, fosA3, floR, tetA(A), tetA(R), strA, strB, sul2, aph(3â€Č)-II, and ΔblaTEM-1]. The spread of these multidrug resistance determinants on the same plasmid is of great concern and, because of coresistance to antibiotics from different classes, is therapeutically challenging

    Resistance to carbapenems in non-typhoidal Salmonella enterica serovars from humans, animals and food

    Get PDF
    Non-typhoidal serovars of Salmonella enterica (NTS) are a leading cause of food-borne disease in animals and humans worldwide. Like other zoonotic bacteria, NTS have the potential to act as reservoirs and vehicles for the transmission of antimicrobial drug resistance in different settings. Of particular concern is the resistance to critical “last resort” antimicrobials, such as carbapenems. In contrast to other Enterobacteriaceae (e.g., Klebsiella pneumoniae, Escherichia coli, and Enterobacter, which are major nosocomial pathogens affecting debilitated and immunocompromised patients), carbapenem resistance is still very rare in NTS. Nevertheless, it has already been detected in isolates recovered from humans, companion animals, livestock, wild animals, and food. Five carbapenemases with major clinical importance—namely KPC (Klebsiella pneumoniae carbapenemase) (class A), IMP (imipenemase), NDM (New Delhi metallo-ÎČ-lactamase), VIM (Verona integron-encoded metallo-ÎČ-lactamase) (class B), and OXA-48 (oxacillinase, class D)—have been reported in NTS. Carbapenem resistance due to the production of extended spectrum- or AmpC ÎČ-lactamases combined with porin loss has also been detected in NTS. Horizontal gene transfer of carbapenemase-encoding genes (which are frequently located on self-transferable plasmids), together with co- and cross-selective adaptations, could have been involved in the development of carbapenem resistance by NTS. Once acquired by a zoonotic bacterium, resistance can be transmitted from humans to animals and from animals to humans through the food chain. Continuous surveillance of resistance to these “last resort” antibiotics is required to establish possible links between reservoirs and to limit the bidirectional transfer of the encoding genes between S. enterica and other commensal or pathogenic bacteria

    Antimicrobial susceptibility and emerging resistance determinants (blaCTX-M, rmtB, fosA3) in clinical isolates from urinary tract infections in the Bolivian Chaco

    Get PDF
    Summary Background Bolivia is among the lowest-resourced South American countries, with very few data available on antibiotic resistance in bacterial pathogens. The phenotypic and molecular characterization of bacterial isolates responsible for urinary tract infections (UTIs) in the Bolivian Chaco are reported here. Methods All clinical isolates from UTIs collected in the Hospital Basico Villa Montes between June 2010 and January 2014 were analyzed ( N =213). Characterization included susceptibility testing, extended-spectrum beta-lactamase (ESBL) detection, identification of relevant resistance determinants (e.g., CTX-M-type ESBLs, 16S rRNA methyltransferases, glutathione S-transferases), and genotyping of CTX-M producers. Results Very high resistance rates were observed. Overall, the lowest susceptibility was observed for trimethoprim–sulphamethoxazole, tetracycline, nalidixic acid, amoxicillin–clavulanic acid, ciprofloxacin, and gentamicin. Of E. coli and K. pneumoniae , 11.6% were ESBL producers. Resistance to nitrofurantoin, amikacin, and fosfomycin remained low, and susceptibility to carbapenems was fully preserved. CTX-M-15 was the dominant CTX-M variant. Four E. coli ST131 (two being H30-Rx) were identified. Of note, isolates harbouring rmtB and fosA3 were detected. Conclusions Bolivia is not an exception to the very high resistance burden affecting many South American countries. Optimization of alternative approaches to monitor local antibiotic resistance trends in resource-limited settings is strongly encouraged to support the implementation of effective empiric treatment guidelines

    Molecular Mechanisms and Clinical Impact of Acquired and Intrinsic Fosfomycin Resistance

    Get PDF
    Bacterial infections caused by antibiotic-resistant isolates have become a major health problem in recent years, since they are very difficult to treat, leading to an increase in morbidity and mortality. Fosfomycin is a broad-spectrum bactericidal antibiotic that inhibits cell wall biosynthesis in both Gram-negative and Gram-positive bacteria. This antibiotic has a unique mechanism of action and inhibits the initial step in peptidoglycan biosynthesis by blocking the enzyme, MurA. Fosfomycin has been used successfully for the treatment of urinary tract infections for a long time, but the increased emergence of antibiotic resistance has made fosfomycin a suitable candidate for the treatment of infections caused by multidrug-resistant pathogens, especially in combination with other therapeutic partners. The acquisition of fosfomycin resistance could threaten the reintroduction of this antibiotic for the treatment of bacterial infection. Here, we analyse the mechanism of action and molecular mechanisms for the development of fosfomycin resistance, including the modification of the antibiotic target, reduced antibiotic uptake and antibiotic inactivation. In addition, we describe the role of each pathway in clinical isolates.This work was supported by the Ministerio de Economía y Competitividad, Instituto de Salud Carlos III, co-financed by the European Development Regional Fund “A way to achieve Europe” ERDF, Spanish Network for the Research in Infectious Diseases (REIPI RD12/0015 and FIS PI10/00105) and the PAR Project (Ref 241476) of the EU 7th Framework Programme

    Acinetobacter baumannii producing ESBLs and carbapenemases in the Intensive Care Units developing fosfomycin and colistin resistance

    Get PDF
    Acinetobacter baumannii is responsible for causing difficult-to-treat healthcare-associated infections globally, owing to its resistance to antibiotics. The intensive care unit (ICU) settings mediate spread of multidrug resistance (MDR) strains. This research aimed to evaluate non-susceptible colistin and fosfomycin A. baumannii, harboring extended-spectrum beta-lactamases (ESBLs) and carbapenemases in ICU setting. During the period of 2019-2021, this study obtained 200 A. baumanni isolates out of 1410 burns samples from an ICU setting. The antibiotic sensitivity, ESBLs and carbapenemase production were determined using clinical and laboratory standards institute (CLSI) 2020. The colistin (mcr-1 and mcr-2) and fosfomycin (fosA3) resistance genes was amplified. The highest resistance was to ceftazidime (98%), cefepime (86%), tetracycline (84%), levofloxacin (78%) and piperacillin-tazobactam (76%), while the highest sensitivity was to meropenem (63%) and tigecycline (62%). ESBL production was determined in 94% and carbapenemases were observed in 54% of A. baumannii. Four isolates (2%) were found to carry the mcr-1 gene, and three isolates (1.5%) were found to carry the mcr-2 gene. Moreover, the fosA3 was not detected in the isolates. This study showed that MDR A. baumannii was high in ICU settings. The spread of antibiotics considered the last line of defense against infections is a concern that necessitates surveillance and control measures
    • 

    corecore