51
|
Papagiannitsis CC, Tsilipounidaki K, Malli E, Petinaki E. Detection in Greece of a clinical Enterococcus faecium isolate carrying the novel oxazolidinone resistance gene poxtA. J Antimicrob Chemother 2020; 74:2461-2462. [PMID: 31009054 DOI: 10.1093/jac/dkz155] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | | | - Ergina Malli
- Department of Microbiology, University Hospital of Larissa, Larissa, Greece
| | - Efi Petinaki
- Department of Microbiology, University Hospital of Larissa, Larissa, Greece
| |
Collapse
|
52
|
Li P, Yang Y, Ding L, Xu X, Lin D. Molecular Investigations of Linezolid Resistance in Enterococci OptrA Variants from a Hospital in Shanghai. Infect Drug Resist 2020; 13:2711-2716. [PMID: 32801805 PMCID: PMC7414921 DOI: 10.2147/idr.s251490] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 07/05/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose The OptrA protein is a member of ATP-binding cassette (ABC) transporters, a transporter family which can confer resistance to oxazolidinone antibiotics by transferring plasmid. We aim to describe the distribution of optrA-harbored Enterococcus in Huashan hospital in 2017 and to address the effects of optrA mutations on the susceptibility of linezolid antibiotic drug. Methods Linezolid-resistance-related genes were tested for Enterococcus by polymerase chain reaction (PCR) and then sequenced for amino acid substitution site analysis. Broth microdilution and agar dilution test were applied to determine the minimal inhibitory concentration (MIC) of linezolid for Enterococcus containing optrA. Pulsed field gel electrophoresis (PFGE) and multilocus sequence typing (MLST) were used to evaluate the genotypes of optrA-positive isolates. To verify the functions of two main optrA variants, optrA over-expressing strains were constructed. Results Among 20 optrA-positive strains, only two were resistant to linezolid. No amino acid substitution existed in 23S rRNA V domain among Enterococcus faecalis. None had cfr, cfr(B) or cfr(C) genes. F101L and G4D/K/R or T150A were the main substitutions of ribosomal protein L4, L3, respectively. We found one Enterococcus faecium isolate co-contained optrA and poxtA and another E. faecalis isolate co-contained optrA and cfr(D), but they were not resistant to linezolid. Among 20 optrA-positive strains, ST-16 was the main type. Two main optrA variants KD (T112K, Y176D) and RDK (I104R, Y176D, E256K) slightly raised enterococci's MIC of linezolid. Conclusion OptrA exists in linezolid non-resistant enterococci with diverse amino acid substitutions. The variants play different roles in changing the MIC of linezolid.
Collapse
Affiliation(s)
- Pei Li
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yang Yang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Li Ding
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xiaogang Xu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Dongfang Lin
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
53
|
García-Angulo VA, Herve B, Melo J, Sanhueza C, la Fuente SD, Aguirre LL, Baysdorfer C, Ulloa MT. Isolation and first draft genome sequence of a linezolid-dependent Staphylococcus aureus clinical strain. Future Microbiol 2020; 15:1123-1129. [PMID: 32954844 DOI: 10.2217/fmb-2020-0068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: Antibiotic-dependent pathogenic bacteria are sporadically isolated from patients that received prolonged antibiotic treatments. Evolution of antibiotics dependence and its clinical implications are scarcely studied. Materials & methods: A linezolid-dependent Staphylococcus aureus strain was isolated from a cystic fibrosis patient. A draft genome sequence was obtained and searched for known antibiotics resistance determinants and virulence factors. Results: The genome was assembled into 79 contigs for a total of 2.83 Mbp. This strain is a sequence type 5 methicillin-resistant Staphylococcus aureus with a type I SCCmec cassette also conserving the Panton-Valentine leukocidin. The G2576T substitution, conferring linezolid resistance, was harbored by all five copies of the 23S rRNA. Conclusion: The linezolid-dependent strain is related to a strain circulating in Latin America that acquired a mutation conferring linezolid resistance.
Collapse
Affiliation(s)
- Víctor A García-Angulo
- Microbiology & Mycology Program, Biomedical Sciences Institute, School of Medicine, University of Chile, Santiago 8380453, Chile
| | - Beatrice Herve
- Laboratorio Clínico, Clínica Las Condes, RM, Santiago 8340518, Chile
| | - Joel Melo
- Departamento de Medicina Interna, Clínica Las Condes, RM, Santiago 8340518, Chile
| | - Camila Sanhueza
- Microbiology & Mycology Program, Biomedical Sciences Institute, School of Medicine, University of Chile, Santiago 8380453, Chile
| | | | - Luis Lozano Aguirre
- Unidad de Análisis Bioinformáticos, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca 62209, México
| | - Chris Baysdorfer
- Department of Biological Sciences, California State University, East Bay, Hayward, CA 94542, USA
| | - María Teresa Ulloa
- Microbiology & Mycology Program, Biomedical Sciences Institute, School of Medicine, University of Chile, Santiago 8380453, Chile
| |
Collapse
|
54
|
Mechanisms of Linezolid Resistance Among Enterococci of Clinical Origin in Spain-Detection of optrA- and cfr(D)-Carrying E. faecalis. Microorganisms 2020; 8:microorganisms8081155. [PMID: 32751552 PMCID: PMC7464793 DOI: 10.3390/microorganisms8081155] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
The mechanisms of linezolid resistance among 13 E. faecalis and 6 E. faecium isolates, recovered from six Spanish hospitals during 2017–2018, were investigated. The presence of acquired linezolid resistance genes and mutations in 23S rDNA and in genes encoding for ribosomal proteins was analyzed by PCR and amplicon sequencing. Moreover, the susceptibility to 18 antimicrobial agents was investigated, and the respective molecular background was elucidated by PCR-amplicon sequencing and whole genome sequencing. The transferability of the linezolid resistance genes was evaluated by filter-mating experiments. The optrA gene was detected in all 13 E. faecalis isolates; and one optrA-positive isolate also carried the recently described cfr(D) gene. Moreover, one E. faecalis isolate displayed the nucleotide mutation G2576T in the 23S rDNA. This mutation was also present in all six E. faecium isolates. All linezolid-resistant enterococci showed a multiresistance phenotype and harbored several antimicrobial resistance genes, as well as many virulence determinants. The fexA gene was located upstream of the optrA gene in 12 of the E. faecalis isolates. Moreover, an erm(A)-like gene was located downstream of optrA in two isolates recovered from the same hospital. The optrA gene was transferable in all but one E. faecalis isolates, in all cases along with the fexA gene. The cfr(D) gene was not transferable. The presence of optrA and mutations in the 23S rDNA are the main mechanisms of linezolid resistance among E. faecalis and E. faecium, respectively. We report the first description of the cfr(D) gene in E. faecalis. The presence of the optrA and cfr(D) genes in Spanish hospitals is a public health concern.
Collapse
|
55
|
Staphylococcal infections and infertility: mechanisms and management. Mol Cell Biochem 2020; 474:57-72. [PMID: 32691256 DOI: 10.1007/s11010-020-03833-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/11/2020] [Indexed: 12/18/2022]
Abstract
Infertility is a subject of worldwide concern as it affects approximately 15% of couples. Among the prime contributors of infertility, urogenital bacterial infections have lately gained much clinical importance. Staphylococcal species are commensal bacteria and major human pathogens mediating an array of reproductive tract infections. Emerging evidences are 'bit by bit' revealing the mechanisms by which Staphylococci strategically disrupt normal reproductive functions. Staphylococcal species can directly or through hematogenous routes can invade the reproductive tissues. In the testicular cells, epididymis as well as in various compartments of female reproductive tracts, the pathogen recognition receptors, toll-like receptors (TLRs), can recognize the pathogen-associated molecular patterns on the Staphylococci and thereby activate inflammatory signalling pathways. These elicit pro-inflammatory mediators trigger other immune cells to infiltrate and release further inflammatory agents and reactive oxygen species (ROS). Adaptive immune responses may intensify the inflammation-induced reproductive tissue damage, particularly via activation of T-helper (Th) cells, Th1 and Th17 by the innate components or by staphylococcal exotoxins. Staphylococcal surface factors binding with sperm membrane proteins can directly impair sperm functions. Although Staphylococci, being one of the most virulent bacterial species, are major contributors in infection-induced infertility in both males and females, the mechanisms of their operations remain under-discussed. The present review aims to provide a comprehensive perception of the possible mechanisms of staphylococcal infection-induced male and female infertility and aid potential interventions to address the lack of competent therapeutic measures for staphylococcal infection-induced infertility.
Collapse
|
56
|
Hasman H, Clausen PTLC, Kaya H, Hansen F, Knudsen JD, Wang M, Holzknecht BJ, Samulioniené J, Røder BL, Frimodt-Møller N, Lund O, Hammerum AM. LRE-Finder, a Web tool for detection of the 23S rRNA mutations and the optrA, cfr, cfr(B) and poxtA genes encoding linezolid resistance in enterococci from whole-genome sequences. J Antimicrob Chemother 2020; 74:1473-1476. [PMID: 30863844 DOI: 10.1093/jac/dkz092] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/16/2019] [Accepted: 02/11/2019] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES In enterococci, resistance to linezolid is often mediated by mutations in the V domain of the 23S rRNA gene (G2576T or G2505A). Furthermore, four genes [optrA, cfr, cfr(B) and poxtA] encode linezolid resistance in enterococci. We aimed to develop a Web tool for detection of the two mutations and the four genes encoding linezolid resistance in enterococci from whole-genome sequence data. METHODS LRE-Finder (where LRE stands for linezolid-resistant enterococci) detected the fraction of Ts in position 2576 and the fraction of As in position 2505 of the 23S rRNA and the cfr, cfr(B), optrA and poxtA genes by aligning raw sequencing reads (fastq format) with k-mer alignment. For evaluation, fastq files from 21 LRE isolates were submitted to LRE-Finder. As negative controls, fastq files from 1473 non-LRE isolates were submitted to LRE-Finder. The MICs of linezolid were determined for the 21 LRE isolates. As LRE-negative controls, 26 VRE isolates were additionally selected for linezolid MIC determination. RESULTS LRE-Finder was validated and showed 100% concordance with phenotypic susceptibility testing. A cut-off of 10% mutations in position 2576 and/or position 2505 was set in LRE-Finder for predicting a linezolid resistance phenotype. This cut-off allows for detection of a single mutated 23S allele in both Enterococcus faecalis and Enterococcus faecium, while ignoring low-level sequencing noise. CONCLUSIONS A Web tool for detection of the 23S rRNA mutations (G2576T and G2505A) and the optrA, cfr, cfr(B) and poxtA genes from whole-genome sequences from enterococci is now available online.
Collapse
Affiliation(s)
- Henrik Hasman
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Philip T L C Clausen
- Department of Genomic Epidemiology, National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Hülya Kaya
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Frank Hansen
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Jenny Dahl Knudsen
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - Mikala Wang
- Department of Clinical Microbiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Jurgita Samulioniené
- Department of Clinical Microbiology, Aalborg University Hospital, Aalborg, Denmark
| | - Bent L Røder
- Department of Clinical Microbiology, Slagelse Hospital, Slagelse, Denmark
| | | | - Ole Lund
- Department of Genomic Epidemiology, National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anette M Hammerum
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
57
|
Sassi M, Guérin F, Zouari A, Beyrouthy R, Auzou M, Fines-Guyon M, Potrel S, Dejoies L, Collet A, Boukthir S, Auger G, Bonnet R, Cattoir V. Emergence of optrA-mediated linezolid resistance in enterococci from France, 2006-16. J Antimicrob Chemother 2020; 74:1469-1472. [PMID: 30897199 DOI: 10.1093/jac/dkz097] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 02/11/2019] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES To describe the epidemiological trend of linezolid-resistant enterococci (LRE) collected in France from 2006 to 2016 and to extensively characterize LRE isolates. METHODS The National Reference Center for Enterococci (NRC-Enc) received enterococcal isolates suspected to be VRE and/or LRE from all French hospitals between 2006 and 2016. LRE isolates were phenotypically characterized and their genomes were entirely sequenced by Miseq (Illumina). Transfer of linezolid resistance was attempted by filter mating experiments. RESULTS Out of 3974 clinical isolates of enterococci received at the NRC-Enc over the period, 9 (0.2%) were LRE (MICs 8 to >32 mg/L), including 6 Enterococcus faecium and 3 Enterococcus faecalis. This overall prevalence significantly increased over the study period, reaching 0.8% in 2016. The five LRE isolated before 2016 were vanA-positive E. faecium whereas strains isolated in 2016 (one E. faecium and three E. faecalis) were susceptible to vancomycin. None of these isolates was part of an outbreak, while E. faecium strains were assigned to four different STs [17 (1), 80 (3), 412 (1) and 650 (1)] and all three E. faecalis belonged to ST480. Except for the strain isolated in 2010, all LRE were positive for optrA, which was located on plasmids (5/8) or in the chromosome (3/8). Plasmid transfer of optrA was successful in three cases. CONCLUSIONS There has been a significant increase in the prevalence of LRE in France over time; this is due to the spread of optrA among E. faecium and E. faecalis human clinical isolates (VRE or not).
Collapse
Affiliation(s)
- Mohamed Sassi
- Université de Rennes 1, Inserm U1230, Rennes, France
| | - François Guérin
- CHU de Caen, Service de Microbiologie, Caen, France.,Université de Caen Normandie, EA4655, Caen, France
| | - Asma Zouari
- CNR de la Résistance aux Antibiotiques (laboratoire associé «Entérocoques»), Rennes, France
| | - Racha Beyrouthy
- CHU Clermont-Ferrand, Laboratoire de Bactériologie & CNR de la Résistance aux Antibiotiques, Clermont-Ferrand, France.,CNR de la Résistance aux Antibiotiques (laboratoire associé «Entérobactéries: résistance aux C3G et colistine»), Clermont-Ferrand, France.,Université Clermont Auvergne, UMR INSERM 1071 USC INRA2018, Clermont-Ferrand, France
| | - Michel Auzou
- CHU de Caen, Service de Microbiologie, Caen, France
| | | | - Sophie Potrel
- CNR de la Résistance aux Antibiotiques (laboratoire associé «Entérocoques»), Rennes, France.,CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
| | - Loren Dejoies
- Université de Rennes 1, Inserm U1230, Rennes, France.,CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
| | - Anaïs Collet
- CNR de la Résistance aux Antibiotiques (laboratoire associé «Entérocoques»), Rennes, France.,CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
| | - Sarrah Boukthir
- CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
| | - Gabriel Auger
- CNR de la Résistance aux Antibiotiques (laboratoire associé «Entérocoques»), Rennes, France.,CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
| | - Richard Bonnet
- CHU Clermont-Ferrand, Laboratoire de Bactériologie & CNR de la Résistance aux Antibiotiques, Clermont-Ferrand, France.,CNR de la Résistance aux Antibiotiques (laboratoire associé «Entérobactéries: résistance aux C3G et colistine»), Clermont-Ferrand, France.,Université Clermont Auvergne, UMR INSERM 1071 USC INRA2018, Clermont-Ferrand, France
| | - Vincent Cattoir
- Université de Rennes 1, Inserm U1230, Rennes, France.,CNR de la Résistance aux Antibiotiques (laboratoire associé «Entérocoques»), Rennes, France.,CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
| |
Collapse
|
58
|
Comparison of Antimicrobial Resistance and Pan-Genome of Clinical and Non-Clinical Enterococcus cecorum from Poultry Using Whole-Genome Sequencing. Foods 2020; 9:foods9060686. [PMID: 32466367 PMCID: PMC7353540 DOI: 10.3390/foods9060686] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/20/2020] [Accepted: 05/24/2020] [Indexed: 02/07/2023] Open
Abstract
Enterococcus cecorum is an emerging avian pathogen, particularly in chickens, but can be found in both diseased (clinical) and healthy (non-clinical) poultry. To better define differences between E. cecorum from the two groups, whole-genome sequencing (WGS) was used to identify and compare antimicrobial resistance genes as well as the pan-genome among the isolates. Eighteen strains selected from our previous study were subjected to WGS using Illumina MiSeq and comparatively analyzed. Assembled contigs were analyzed for resistance genes using ARG-ANNOT. Resistance to erythromycin was mediated by ermB, ermG, and mefA, in clinical isolates and ermB and mefA, in non-clinical isolates. Lincomycin resistance genes were identified as linB, lnuB, lnuC, and lnuD with lnuD found only in non-clinical E. cecorum; however, lnuB and linB were found in only one clinical isolate. For both groups of isolates, kanamycin resistance was mediated by aph3-III, while tetracycline resistance was conferred by tetM, tetO, and tetL. No mutations or known resistance genes were found for isolates resistant to either linezolid or chloramphenicol, suggesting possible new mechanisms of resistance to these drugs. A comparison of WGS results confirmed that non-clinical isolates contained more resistance genes than clinical isolates. The pan-genome of clinical and non-clinical isolates resulted in 3651 and 4950 gene families, respectively, whereas the core gene sets were comprised of 1559 and 1534 gene families in clinical and non-clinical isolates, respectively. Unique genes were found more frequently in non-clinical isolates than clinical. Phylogenetic analysis of the isolates and all the available complete and draft genomes showed no correlation between healthy and diseased poultry. Additional genomic comparison is required to elucidate genetic factors in E. cecorum that contribute to disease in poultry.
Collapse
|
59
|
Detection of the enterococcal oxazolidinone/phenicol resistance gene optrA in Campylobacter coli. Vet Microbiol 2020; 246:108731. [PMID: 32605743 DOI: 10.1016/j.vetmic.2020.108731] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 11/22/2022]
Abstract
The transferable optrA gene encodes an ABC-F protein which confers resistance to oxazolidinones and phenicols, and has so far been detected exclusively in Gram-positive bacteria, including enterococci, staphylococci and streptococci. Here, we identified for the first time the presence of optrA in naturally occurring Gram-negative bacteria. Seven optrA-positive Campylobacter coli were identified from 563 Campylobacter isolates of animal origin from Guangdong (n = 1, chicken) and Shandong (n = 6, duck) provinces of China in 2017-2018. The detected optrA genes were functionally active and mediated resistance or elevated minimal inhibitory concentrations of linezolid, florfenicol and chloramphenicol in the respective C. coli isolates. The optrA gene, together with other transferable resistance genes, such as fexA, catA9, tet(O), tet(L), erm(A)-like, spc, or aadE, was located in two different chromosome-borne multidrug resistance genomic islands (MDRGIs). In both MDRGIs, complete or truncated copies of the insertion sequence IS1216E were present in the vicinity of optrA. The IS1216E-bracketed genetic environment of optrA was almost identical to the optrA regions on enterococcal plasmids, suggesting that the optrA in Campylobacter probably originated from Enterococcus spp.. Moreover, the formation of an optrA-carrying translocatable unit by recombination of IS1216E indicated that this IS element may play an important role in the horizontal transfer of optrA in Campylobacter. Although optrA was only found in a small number of C. coli isolates, enhanced surveillance is needed to monitor the distribution and the potential emergence of optrA in Campylobacter.
Collapse
|
60
|
Ruiz-Ripa L, Bellés A, García M, Torres C. Detection of a cfr-positive MRSA CC398 strain in a pig farmer in Spain. Enferm Infecc Microbiol Clin 2020; 39:139-141. [PMID: 32386788 DOI: 10.1016/j.eimc.2020.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To characterize one linezolid- and methicillin-resistant Staphylococcus aureus (MRSA) isolate recovered from a nasal sample of a pig farmer patient. METHODS The detection of linezolid resistance mechanisms was performed by PCR and sequencing. The antimicrobial resistance and virulence profile was investigated, and the molecular typing was performed by molecular techniques. The transference of cfr gene was assessed by conjugation experiments and its genetic environment was investigated by specific PCRs. RESULTS The linezolid-resistant MRSA isolate was typed as t011-ST398/CC398-SCCmecV-agrI and carried the cfr gene. The isolate was multidrug-resistant but lacked the virulence genes studied. The cfr gene was co-located with the fexA gene on a Tn558 variant and was successfully transferred by conjugation. CONCLUSION We report the first description of LA-MRSA-CC398 carrying the cfr gene in Spain. This finding highlights the importance of surveillance programmes to determine the presence and spread of the cfr gene in the livestock and clinical settings.
Collapse
Affiliation(s)
- Laura Ruiz-Ripa
- Área de Bioquímica y Biología Molecular, Universidad de La Rioja, Logroño, Spain
| | - Alba Bellés
- Sección de Microbiología, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | - Mercè García
- Sección de Microbiología, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | - Carmen Torres
- Área de Bioquímica y Biología Molecular, Universidad de La Rioja, Logroño, Spain.
| |
Collapse
|
61
|
Kosecka-Strojek M, Sadowy E, Gawryszewska I, Klepacka J, Tomasik T, Michalik M, Hryniewicz W, Miedzobrodzki J. Emergence of linezolid-resistant Staphylococcus epidermidis in the tertiary children's hospital in Cracow, Poland. Eur J Clin Microbiol Infect Dis 2020; 39:1717-1725. [PMID: 32350737 PMCID: PMC7427702 DOI: 10.1007/s10096-020-03893-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 04/01/2020] [Indexed: 11/26/2022]
Abstract
Coagulase-negative staphylococci, ubiquitous commensals of human skin, and mucous membranes represent important pathogens for immunocompromised patients and neonates. The increasing antibiotic resistance among Staphylococcus epidermidis is an emerging problem worldwide. In particular, the linezolid-resistant S. epidermidis (LRSE) strains are observed in Europe since 2014. The aim of our study was to genetically characterize 11 LRSE isolates, recovered mostly from blood in the University Children’s Hospital in Krakow, Poland, between 2015 and 2017. For identification of the isolates at the species level, we used 16S rRNA sequencing and RFLP of the saoC gene. Isolates were characterized phenotypically by determining their antimicrobial resistance patterns and using molecular methods such as PFGE, MLST, SCCmec typing, detection of the ica operon, and analysis of antimicrobial resistance determinants. All isolates were multidrug-resistant, including resistance to methicillin, and exhibited so-called PhLOPSA phenotype. In PFGE, all isolates (excluding one from a catheter) represented identical patterns, were identified as ST2, and harbored the ica operon, responsible for biofilm formation. Linezolid resistance was associated with acquisition of A157R mutation in the ribosomal protein L3 and the presence of cfr gene. All isolates revealed new SCCmec cassette element composition. Recently, pediatric patients with serious staphylococcal infections are often treated with linezolid. The increasing linezolid resistance in bacterial strains becomes a real threat for patients, and monitoring such infections combined with surveillance and infection prevention programs is very important to decrease number of linezolid-resistant staphylococcal strains.
Collapse
Affiliation(s)
- Maja Kosecka-Strojek
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Ewa Sadowy
- Department of Molecular Microbiology, National Medicines Institute, Warsaw, Poland
| | - Iwona Gawryszewska
- Department of Molecular Microbiology, National Medicines Institute, Warsaw, Poland
| | - Joanna Klepacka
- Department of Clinical Microbiology, Children's University Hospital, Jagiellonian University, Krakow, Poland
| | - Tomasz Tomasik
- Department of Clinical Microbiology, Children's University Hospital, Jagiellonian University, Krakow, Poland
| | | | - Waleria Hryniewicz
- Department of Epidemiology and Clinical Microbiology, National Medicines Institute, Warsaw, Poland
| | - Jacek Miedzobrodzki
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.
| |
Collapse
|
62
|
Guerin F, Sassi M, Dejoies L, Zouari A, Schutz S, Potrel S, Auzou M, Collet A, Lecointe D, Auger G, Cattoir V. Molecular and functional analysis of the novel cfr(D) linezolid resistance gene identified in Enterococcus faecium. J Antimicrob Chemother 2020; 75:1699-1703. [DOI: 10.1093/jac/dkaa125] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/08/2020] [Accepted: 03/10/2020] [Indexed: 12/17/2022] Open
Abstract
Abstract
Objectives
To characterize the novel cfr(D) gene identified in an Enterococcus faecium clinical isolate (15-307.1) collected from France.
Methods
The genome of 15-307.1 was entirely sequenced using a hybrid approach combining short-read (MiSeq, Illumina) and long-read (GridION, Oxford Nanopore Technologies) technologies in order to analyse in detail the genetic support and environment of cfr(D). Transfer of linezolid resistance from 15-307.1 to E. faecium BM4107 was attempted by filter-mating experiments. The recombinant plasmid pAT29Ωcfr(D), containing cfr(D) and its own promoter, was transferred to E. faecium HM1070, Enterococcus faecalis JH2-2 and Escherichia coli AG100A.
Results
As previously reported, 15-307.1 belonged to ST17 and was phenotypically resistant to linezolid (MIC, 16 mg/L), vancomycin and teicoplanin. A hybrid sequencing approach confirmed the presence of several resistance genes including vanA, optrA and cfr(D). Located on a 103 kb plasmid, cfr(D) encoded a 357 amino acid protein, which shared 64%, 64%, 48% and 51% amino acid identity with Cfr, Cfr(B), Cfr(C) and Cfr(E), respectively. Both optrA and cfr(D) were successfully co-transferred to E. faecium BM4107. When expressed in E. faecium HM1070 and E. faecalis JH2-2, pAT29Ωcfr(D) did not confer any resistance, whereas it was responsible for an expected PhLOPSA resistance phenotype in E. coli AG100A. Analysis of the genetic environment of cfr(D) showed multiple IS1216 elements, putatively involved in its mobilization.
Conclusions
Cfr(D) is a novel member of the family of 23S rRNA methyltransferases. While only conferring a PhLOPSA resistance phenotype when expressed in E. coli, enterococci could constitute an unknown reservoir of cfr(D).
Collapse
Affiliation(s)
- François Guerin
- CHU de Caen, Service de Microbiologie, Caen, France
- Université de Caen Normandie, EA4655 Caen, France
| | - Mohamed Sassi
- Université de Rennes 1, Inserm U1230, Rennes, France
| | - Loren Dejoies
- Université de Rennes 1, Inserm U1230, Rennes, France
- CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
| | - Asma Zouari
- CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
- CNR de la Résistance aux Antibiotiques (laboratoire associé ‘Entérocoques’), Rennes, France
| | - Sacha Schutz
- CHU de Rennes, Service de Génétique moléculaire et Génomique médicale, Rennes, France
| | - Sophie Potrel
- CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
- CNR de la Résistance aux Antibiotiques (laboratoire associé ‘Entérocoques’), Rennes, France
| | - Michel Auzou
- CHU de Caen, Service de Microbiologie, Caen, France
| | - Anaïs Collet
- CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
- CNR de la Résistance aux Antibiotiques (laboratoire associé ‘Entérocoques’), Rennes, France
| | - Didier Lecointe
- Unité Fonctionnelle d’Hygiène Hospitalière et de Lutte contre les Infections Nosocomiales, Centre Hospitalier Sud Francilien, Corbeil-Essonnes, France
| | - Gabriel Auger
- CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
- CNR de la Résistance aux Antibiotiques (laboratoire associé ‘Entérocoques’), Rennes, France
| | - Vincent Cattoir
- Université de Rennes 1, Inserm U1230, Rennes, France
- CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, Rennes, France
- CNR de la Résistance aux Antibiotiques (laboratoire associé ‘Entérocoques’), Rennes, France
| |
Collapse
|
63
|
In vitro synergy with fosfomycin plus doxycyclin against linezolid and vancomycin-resistant Enterococcus faecium. J Glob Antimicrob Resist 2020; 22:78-83. [PMID: 32007618 DOI: 10.1016/j.jgar.2020.01.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/05/2019] [Accepted: 01/20/2020] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Linezolid and vancomycin-resistant Enterococcus faecium (LRVREF) is globally emerging as a urinary nosocomial pathogen. A decrease in the amount of successful treatment options has created a necessity for the development of novel antimicrobial therapies. Combination therapy may provide an effective alternative for treatment. Fosfomycin and doxycyclin are currently used individually for the treatment of urinary tract infections. Fosfomycin has recently seen increased use because of its persistent activity against a large spectrum of multidrug-resistant organisms. The purpose of this study was to investigate the interaction of fosfomycin plus doxycyclin against LRVREF isolates. METHODS MICs for fosfomycin and doxycyclin were determined for 24 unique clinical LRVREF isolates (4%, fosfomycin-susceptible; 92%, doxycyclin-susceptible). In vitro synergy testing with the combination of fosfomycin and doxycyclin was performed using an Etest method and time-kill assay. RESULTS The combination of fosfomycin and doxycyclin demonstrated synergy with the Etest method in 11/24 (46%) isolates and in 10/24 (42%) isolates by the time-kill assay. Results from the Etest method and time-kill assay were in agreement for 7/24 (29.2%) of isolates. No antagonism was found. CONCLUSIONS The combination of fosfomycin and doxycyclin should be evaluated further with additional LRVREF isolates. In vivo studies should also be performed before use in clinical situations.
Collapse
|
64
|
Morpholine as ubiquitous pharmacophore in medicinal chemistry: Deep insight into the structure-activity relationship (SAR). Bioorg Chem 2020; 96:103578. [PMID: 31978684 DOI: 10.1016/j.bioorg.2020.103578] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/09/2019] [Accepted: 01/09/2020] [Indexed: 12/15/2022]
Abstract
Morpholine is a versatile moiety, a privileged pharmacophore and an outstanding heterocyclic motif with wide ranges of pharmacological activities due to different mechanisms of action. The ability of morpholine to enhance the potency of the molecule through molecular interactions with the target protein (kinases) or to modulate the pharmacokinetic properties propelled medicinal chemists and researchers to synthesize morpholine ring by the efficient ways and to incorporate this moiety to develop various lead compounds with diverse therapeutic activities. The present review primarily focused on discussing the most promising synthetic leads containing morpholine ring along with structure-activity relationship (SAR) to reveal the active pharmacophores accountable for anticancer, anti-inflammatory, antiviral, anticonvulsant, antihyperlipidemic, antioxidant, antimicrobial and antileishmanial activity. This review outlines some of the recent effective chemical synthesis for morpholine ring. The review also highlighted the metabolic liability of some clinical drugs containing this nucleus and various researches on modified morpholine to enhance the metabolic stability of drugs as well. Drugs bearing morpholine ring and those under clinical trials are also mentioned with the role of morpholine and their mechanism of action. This review will provide the necessary knowledge base to the medicinal chemists in making strategic structural changes in designing morpholine derivatives.
Collapse
|
65
|
Molecular characteristics and predictors of mortality among Gram-positive bacteria isolated from bloodstream infections in critically ill patients during a 5-year period (2012-2016). Eur J Clin Microbiol Infect Dis 2020; 39:863-869. [PMID: 31898796 PMCID: PMC7223776 DOI: 10.1007/s10096-019-03803-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
To identify the molecular characteristics of Gram-positive cocci isolated from blood cultures and clinical outcome among critically ill patients. This retrospective study was conducted in the general intensive care unit of the University General Hospital of Patras, Greece, during a 5-year period (2012–2016). All adult patients with a Gram-positive BSI were included. PCR was applied to identify mecA gene (staphylococci); vanA, vanB, and vanC genes (enterococci). Linezolid-resistant S. epidermidis, MRSA, and VRE were further typed by multilocus sequence typing. Mutations in region V of 23S rDNA and ribosomal protein L4were investigated by PCR and sequencing analysis. The presence of the cfr gene was tested by PCR. In total, 141 Gram-positive BSIs were included. Coagulase-negative staphylococci predominated (n = 69; 65 methicillin-resistant, 23 linezolid-resistant carrying both C2534T and T2504A mutations and belonging to the ST22 clone), followed by enterococci (n = 46; 11 vancomycin-resistant carrying vanA gene, classified into four clones), S. aureus (n = 22; 10 methicillin-resistant, classified into three clones) and streptococci (n = 4). The most common type of infection was catheter-related (66; 46.8%), followed by primary BSI (28; 19.9%). Overall 14-day fatality was 24.8%. Multivariate analysis revealed septic shock as independent predictor of fatality, while appropriate empiric antimicrobial treatment and catheter-related BSI were identified as a predictor of good prognosis. Even though most of Gram-positive cocci were multidrug-resistant, fatality rate was low, associated with catheter-related BSIs. Among CNS, LR isolates represented one-third of BSIs due to the dissemination of ST22 S. epidermidis propagated by utilization of linezolid.
Collapse
|
66
|
Zheng JX, Chen Z, Xu ZC, Chen JW, Xu GJ, Sun X, Yu ZJ, Qu D. In vitro evaluation of the antibacterial activities of radezolid and linezolid for Streptococcus agalactiae. Microb Pathog 2019; 139:103866. [PMID: 31715321 DOI: 10.1016/j.micpath.2019.103866] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 11/03/2019] [Accepted: 11/08/2019] [Indexed: 11/25/2022]
Abstract
PURPOSE This study aims to evaluate the antimicrobial activities of linezolid and radezolid against Streptococcus agalactiae in vitro and compared for genetic resistance factors. METHOD Nonduplicate S. agalactiae clinical isolates (n = 136) were collected and the minimal inhibitory concentrations of antimicrobials were determined by agar dilution methodology. The linezolid-resistant mechanism in the clinical linezolid-non-susceptible S. agalactiae isolates and that induced by linezolid pressure in vitro were analyzed by PCR and sequence alignment. Antimicrobial activities and resistance mechanism distinctions between linezolid and radezolid were further investigated in the clinical linezolid-non-susceptible S. agalactiae isolates and that induced by linezolid pressure in vitro. RESULTS Our data indicated that 17 (13%) of the 136 clinical S. agalactiae isolates were not susceptible to linezolid. For individual S. agalactiae isolates, including linezolid-nonsusceptible isolates with 23S rRNA V domain mutations, radezolid MIC90 values were generally one-half to one-quarter of the linezolid MIC90 values. Radezolid MICs remained low relative to linezolid MICs among linezolid-resistant S. agalactiae isolates, but exhibited the synchronous increases with the increasing copy numbers of 23S rRNA V domain mutations. Overall, 13 optrA-carrying clinical S. agalactiae isolates were found in this study and their MICs all remained sensitive to both linezolid and radezolid. Clinical S. agalactiae isolates with high radezolid MICs showed clonality clustering to sequence type (ST)10. CONCLUSION Radezolid exhibits stronger potency against S. agalactiae than linezolid and there is a concerning presence of linezolid-nonsusceptible S. agalactiae in clinical samples.
Collapse
Affiliation(s)
- Jin-Xin Zheng
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, China; Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Zhong Chen
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, China; Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Zhi-Chao Xu
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Jun-Wen Chen
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Guang-Jian Xu
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Xiang Sun
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Zhi-Jian Yu
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China.
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, China.
| |
Collapse
|
67
|
Tan F, She P, Zhou L, Liu Y, Chen L, Luo Z, Wu Y. Bactericidal and Anti-biofilm Activity of the Retinoid Compound CD437 Against Enterococcus faecalis. Front Microbiol 2019; 10:2301. [PMID: 31649642 PMCID: PMC6794434 DOI: 10.3389/fmicb.2019.02301] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 09/20/2019] [Indexed: 01/24/2023] Open
Abstract
Enterococcus faecalis (E. faecalis), a biofilm-forming pathogen, causes nosocomial infections. In recent years, drug resistance by enterococci has become increasingly severe due to widespread antibiotic abuse. Therefore, novel antibacterial agents are urgently needed. In this study, the synthetic retinoid compound CD437 was found to have potent bactericidal effect on E. faecalis. In addition, CD437 exhibited synergistic effects when administered in combination with gentamicin and additive effects when combined with ceftriaxone sodium. CD437 also inhibited biofilm formation by E. faecalis and exerted bactericidal effect on mature biofilm. Moreover, CD437 exhibited antibacterial and anti-biofilm effects against Staphylococcus. No bactericidal action of CD437 was observed against the gram-negative bacillus, but Pseudomonas aeruginosa biofilm extracellular polymeric substances (EPS) matrix formation was reduced. Overall, these findings indicate that CD437 has the potential to be developed as a novel antibacterial drug.
Collapse
Affiliation(s)
- Fang Tan
- Department of Medicine Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Pengfei She
- Department of Medicine Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Linying Zhou
- Department of Medicine Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yiqing Liu
- Department of Medicine Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Lihua Chen
- Department of Medicine Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhen Luo
- Department of Medicine Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yong Wu
- Department of Medicine Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
68
|
Deshpande LM, Castanheira M, Flamm RK, Mendes RE. Evolving oxazolidinone resistance mechanisms in a worldwide collection of enterococcal clinical isolates: results from the SENTRY Antimicrobial Surveillance Program. J Antimicrob Chemother 2019; 73:2314-2322. [PMID: 29878213 DOI: 10.1093/jac/dky188] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/18/2018] [Indexed: 01/28/2023] Open
Abstract
Objectives This study evaluated the oxazolidinone resistance mechanisms among a global collection of enterococcal clinical isolates. The epidemiology of optrA-carrying isolates and the optrA genetic context were determined. Methods Enterococcal isolates (26 648) from the SENTRY Antimicrobial Surveillance Program (2008-16) were identified by MALDI-TOF MS and MICs were determined by broth microdilution. Isolates with linezolid MICs of ≥4 mg/L were screened for resistance mechanisms. Isolates carrying optrA had their genome sequenced for genetic context and epidemiology information. Results Thirty-six Enterococcus faecalis and 66 Enterococcus faecium had linezolid MICs of ≥4 mg/L (0.38% of surveillance enterococci). E. faecalis had a linezolid MIC range of 4-16 mg/L, while E. faecium displayed higher values (4-64 mg/L). Nine E. faecalis had G2576T mutations and optrA was detected in 26 (72.2%) isolates from the Asia-Pacific region, North America, Latin America and Europe; 3 isolates also produced Cfr [Thailand (1)] or Cfr(B) [Panama (2)]. All E. faecium isolates had G2576T alterations, while three isolates from the USA had concomitant presence of cfr(B). The optrA gene was plasmid- and chromosome-located in 22 and 3 E. faecalis, respectively. One isolate signalled hybridization on plasmid and chromosome. The genetic context of optrA varied. E. faecalis belonging to the same clonal complex were detected in distinct geographical regions. Also, genetically distinct isolates from Ireland had an identical optrA context, indicating plasmid dissemination. Conclusions Alterations in 23S rRNA remained the main oxazolidinone resistance mechanism in E. faecium, while optrA prevailed in E. faecalis. These results demonstrate global dissemination of optrA and warrant surveillance for monitoring.
Collapse
Affiliation(s)
| | | | - R K Flamm
- JMI Laboratories, North Liberty, IA, USA
| | - R E Mendes
- JMI Laboratories, North Liberty, IA, USA
| |
Collapse
|
69
|
Mendes RE, Deshpande L, Streit JM, Sader HS, Castanheira M, Hogan PA, Flamm RK. ZAAPS programme results for 2016: an activity and spectrum analysis of linezolid using clinical isolates from medical centres in 42 countries. J Antimicrob Chemother 2019; 73:1880-1887. [PMID: 29659858 DOI: 10.1093/jac/dky099] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/02/2018] [Indexed: 12/11/2022] Open
Abstract
Objectives To report the linezolid activity, resistance mechanisms and epidemiological typing of selected isolates observed during the 2016 Zyvox® Annual Appraisal of Potency and Spectrum (ZAAPS) programme. Methods A total of 8325 organisms were consecutively collected from 76 centres in 42 countries (excluding the USA). Broth microdilution susceptibility testing was performed and isolates displaying linezolid MICs of ≥4 mg/L were molecularly characterized. Results Linezolid inhibited 99.8% of all Gram-positive pathogens at the respective susceptible breakpoints and showed a modal MIC of 1 mg/L, except for CoNS, for which the modal MIC result was 0.5 mg/L. Among isolates displaying linezolid MICs of ≥4 mg/L, one Staphylococcus aureus (linezolid MIC of 4 mg/L) harboured cfr and belonged to ST72, while four CoNS (MICs of 16-32 mg/L; ST2) showed drug target alterations. Two Enterococcus faecium (ST117) from a single site in Rome were linezolid non-susceptible (MICs of 8 mg/L) and had G2576T mutations. Eight linezolid-non-susceptible Enterococcus faecalis (MICs of 4 mg/L; 4 sites in 4 countries; ST256, ST480, ST766 and ST775) carried optrA and isolates carrying optrA from the same medical centre were genetically related. One Streptococcus gallolyticus (MIC of 4 mg/L) and one Streptococcus mitis (MIC of 16 mg/L) carried optrA and G2576T mutations, respectively. Conclusions These results document the continued long-term in vitro potency of linezolid. Alterations in the 23S rRNA and/or L3/L4 proteins remain the main oxazolidinone resistance mechanisms in E. faecium and CoNS, whereas optrA emerged as the sole mechanism in E. faecalis. Surveillance and infection control will be important strategies to detect optrA and prevent it from disseminating.
Collapse
|
70
|
Du F, Lv X, Duan D, Wang L, Huang J. Characterization of a Linezolid- and Vancomycin-Resistant Streptococcus suis Isolate That Harbors optrA and vanG Operons. Front Microbiol 2019; 10:2026. [PMID: 31551963 PMCID: PMC6746840 DOI: 10.3389/fmicb.2019.02026] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022] Open
Abstract
Linezolid and vancomycin are among the last-resort antimicrobial agents in the treatment of multidrug-resistant Gram-positive bacterial infections. Linezolid- and vancomycin-resistant (LVR) Gram-positive bacteria may pose severe threats to public health. In this study, three optrA- and vanG-positive Streptococcus suis strains were isolated from two farms of different cities. There were only 1 and 343 single-nucleotide polymorphisms in coding region (cSNPs) of HCB4 and YSJ7 to YSJ17, respectively. Mobilome analysis revealed the presence of vanG, erm(B), tet(O/W/32/O), and aadE-apt-sat4-aphA3 cluster on an integrative and conjugative element, ICESsuYSJ17, and erm(B), aphA3, aac(6')-aph(2″), catpC194, and optrA on a prophage, ΦSsuYSJ17-3. ICESsuYSJ17 exhibited a mosaic structure and belongs to a highly prevalent and transferable ICESa2603 family of Streptococcus species. ΦSsuYSJ17-3 shared conserved backbone to a transferable prophage Φm46.1. A novel composite transposon, IS1216E-araC-optrA-hp-catpC194-IS1216E, which can be circulated as translocatable unit (TU) by IS1216E, was integrated on ΦSsuYSJ17-3. Vancomycin resistance phenotype and vanG transcription assays revealed that the vanG operon was inducible. The LVR strain YSJ17 exhibited moderate virulence in a zebrafish infection model. To our knowledge, this is the first report of LVR isolate, which is mediated by acquired resistance genes optrA and vanG operons in Gram-positive bacteria. Since S. suis has been recognized as an antimicrobial resistance reservoir in the spread of resistance genes to major streptococcal pathogens, the potential risks of disseminating of optrA and vanG from S. suis to other Streptococcus spp. are worrisome and routine surveillance should be strengthened.
Collapse
Affiliation(s)
- Fanshu Du
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xi Lv
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Duan Duan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Liping Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jinhu Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
71
|
O'Dea M, Sahibzada S, Jordan D, Laird T, Lee T, Hewson K, Pang S, Abraham R, Coombs GW, Harris T, Pavic A, Abraham S. Genomic, Antimicrobial Resistance, and Public Health Insights into Enterococcus spp. from Australian Chickens. J Clin Microbiol 2019; 57:e00319-19. [PMID: 31118269 PMCID: PMC6663891 DOI: 10.1128/jcm.00319-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/16/2019] [Indexed: 12/15/2022] Open
Abstract
Due to Australia's management of antimicrobial use in poultry, particularly the discontinued use of avoparcin for nearly 20 years, it is hypothesized that vancomycin-resistant enterococci associated with human disease are not derived from poultry isolates. This study evaluated antimicrobial resistance (AMR) of five enterococcal species isolated from Australian meat chickens, genomic features of Enterococcus faecium and Enterococcus faecalis, and the phylogenetic relationship of the poultry-derived E. faecium with isolates from human sepsis cases. All enterococcal isolates from chicken ceca were subjected to antimicrobial susceptibility testing. E. faecium and E. faecalis underwent whole-genome sequencing. E. faecium was compared at the core genome level to a collection of human isolates (n = 677) obtained from cases of sepsis over a 2-year period spanning 2015 to 2016. Overall, 205 enterococci were isolated consisting of five different species. E. faecium was the most frequently isolated species (37.6%), followed by E. durans (29.7%), E. faecalis (20%), E. hirae (12.2%), and E. gallinarum (0.5%). All isolates were susceptible to vancomycin and gentamicin, while one isolate was linezolid resistant (MIC 16 mg/liter). Core genome analysis of the E. faecium demonstrated two clades consisting predominantly of human or chicken isolates in each clade, with minimal overlap. Principal component analysis for total gene content revealed three clusters comprised of vanA-positive, vanB-positive, and both vanA- and vanB-negative E. faecium populations. The results of this study provide strong evidence that Australian chicken E. faecium isolates are unlikely to be precursor strains to the currently circulating vancomycin-resistant strains being isolated in Australian hospitals.
Collapse
Affiliation(s)
- Mark O'Dea
- Antimicrobial Resistance and Infectious Diseases Laboratory, Murdoch University, Murdoch, WA, Australia
| | - Shafi Sahibzada
- Antimicrobial Resistance and Infectious Diseases Laboratory, Murdoch University, Murdoch, WA, Australia
| | - David Jordan
- New South Wales Department of Primary Industries, Wollongbar, NSW, Australia
| | - Tanya Laird
- Antimicrobial Resistance and Infectious Diseases Laboratory, Murdoch University, Murdoch, WA, Australia
| | - Terence Lee
- Antimicrobial Resistance and Infectious Diseases Laboratory, Murdoch University, Murdoch, WA, Australia
| | - Kylie Hewson
- Australian Chicken Meat Federation, North Sydney, NSW, Australia
| | - Stanley Pang
- Antimicrobial Resistance and Infectious Diseases Laboratory, Murdoch University, Murdoch, WA, Australia
- PathWest Laboratory Medicine, Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Rebecca Abraham
- Antimicrobial Resistance and Infectious Diseases Laboratory, Murdoch University, Murdoch, WA, Australia
| | - Geoffrey W Coombs
- Antimicrobial Resistance and Infectious Diseases Laboratory, Murdoch University, Murdoch, WA, Australia
- PathWest Laboratory Medicine, Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Taha Harris
- Birling Avian Laboratories, Bringelly, NSW, Australia
| | - Anthony Pavic
- Birling Avian Laboratories, Bringelly, NSW, Australia
| | - Sam Abraham
- Antimicrobial Resistance and Infectious Diseases Laboratory, Murdoch University, Murdoch, WA, Australia
| |
Collapse
|
72
|
Chen H, Wang X, Yin Y, Li S, Zhang Y, Wang Q, Wang H. Molecular characteristics of oxazolidinone resistance in enterococci from a multicenter study in China. BMC Microbiol 2019; 19:162. [PMID: 31299904 PMCID: PMC6626368 DOI: 10.1186/s12866-019-1537-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 06/30/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Linezolid-resistant enterococci pose great challenges in clinical practice. The aim of this study is to study the mechanisms underlying the resistance and genetic environment of antimicrobial resistance gene of linezolid-resistant enterococci. RESULTS The linezolid MICs of 16 enterococci were 4 mg/L to 16 mg/L. Four strains belonged to multi-drug resistant (MDR) bacteria. The sequence types (STs) of 13 enterococci strains performed WGS were diverse: 3 ST476, 1 ST86, ST116, ST480, ST59, ST416, ST21, ST67, ST16, ST585 and ST18. None of them carried multi-drug resistance gene cfr. Only one strain had the G2658 T mutation of target 23S rRNA gene. Thirteen (13/16, 81.3%) strains harbored the novel oxazolidinone resistance gene optrA. WGS analysis showed that the optrA gene was flanked by sequence IS1216E insertion in 13 strains, and optrA was adjacent to transposons Tn558 in two strains and Tn554 in one strain. The optrA gene was identified to be co-localized with fexA, the resistance genes mediated florfenicol resistance in 13 strains, and ermA1, the resistance genes mediated erythromycin resistance in 9 strains, indicating that linezolid-resistant strains may be selected due to non-oxazolidinone antibiotics (i.e. macrolides and florfenicol) usage. CONCLUSION Our findings demonstrate the high diversity of optrA-carrying genetic platforms. The mobile genetic elements (MGEs) may play an important role in the dissemination of optrA into the enterococci isolates of human origin. The genetic evidence of transferable feature and co-selection of optrA should be gave more attention in clinical practice.
Collapse
Affiliation(s)
- Hongbin Chen
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing, 100044 People’s Republic of China
| | - Xiaojuan Wang
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing, 100044 People’s Republic of China
| | - Yuyao Yin
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing, 100044 People’s Republic of China
| | - Shuguang Li
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing, 100044 People’s Republic of China
| | - Yawei Zhang
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing, 100044 People’s Republic of China
| | - Qi Wang
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing, 100044 People’s Republic of China
| | - Hui Wang
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing, 100044 People’s Republic of China
| |
Collapse
|
73
|
Weßels C, Strommenger B, Klare I, Bender J, Messler S, Mattner F, Krakau M, Werner G, Layer F. Emergence and control of linezolid-resistant Staphylococcus epidermidis in an ICU of a German hospital. J Antimicrob Chemother 2019; 73:1185-1193. [PMID: 29438544 DOI: 10.1093/jac/dky010] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/02/2018] [Indexed: 11/13/2022] Open
Abstract
Objectives To investigate an outbreak of linezolid-resistant Staphylococcus epidermidis (LRSE) in an interdisciplinary ICU, linezolid consumption and infection control measures taken. Methods Routine surveillance of nosocomial infections revealed colonization and infection with LRSE affecting 14 patients during a 15 month period. LRSE isolates were analysed with respect to their clonal relatedness, antimicrobial susceptibility, the presence of cfr and/or mutations in the 23S rRNA, rplC, rplD and rplV genes. cfr plasmids were characterized by Illumina sequencing. Medical records were reviewed and antibiotic consumption was determined. Results Molecular typing identified the presence of three different LRSE clusters: PFGE type I/ST168 (n = 5), PFGE type II/ST5 (n = 10) and PFGE type III/ST2 (n = 1). Ten strains harboured the cfr gene; we also detected mutations in the respective ribosomal protein genes. WGS revealed an almost identical 39 kb cfr plasmid obtained from strains of different genetic background (ST2, ST5, ST168) that shows high similarity to the recently published LRSE plasmid p12-02300. Due to an increase in the number of patients treated for infections with MRSA, a significant increase in linezolid usage was noted from January to July 2014 (from 5.55 to 20.41 DDDs/100 patient-days). Conclusions Here, we report the molecular epidemiology of LRSE in an ICU. Our results suggest the selection of resistant mutants under linezolid treatment as well as the spread of cfr-carrying plasmids. The reduction of linezolid usage and the strengthening of contact precautions proved to be effective infection control measures.
Collapse
Affiliation(s)
- Christina Weßels
- Institute of Hospital Hygiene, City of Cologne Hospitals, Cologne, Germany
| | - Birgit Strommenger
- National Reference Centre for Staphylococci and Enterococci, Division 13: Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode Branch, Wernigerode, Germany
| | - Ingo Klare
- National Reference Centre for Staphylococci and Enterococci, Division 13: Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode Branch, Wernigerode, Germany
| | - Jennifer Bender
- National Reference Centre for Staphylococci and Enterococci, Division 13: Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode Branch, Wernigerode, Germany
| | - Sabine Messler
- Labor im Sommershof, Praxis für Laboratoriumsmedizin Dr. med. Christiane Boogen, Cologne, Germany
| | - Frauke Mattner
- Institute of Hospital Hygiene, City of Cologne Hospitals, Cologne, Germany
| | - Michael Krakau
- Department of Internal Medicine, City of Cologne Hospitals, Cologne, Germany
| | - Guido Werner
- National Reference Centre for Staphylococci and Enterococci, Division 13: Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode Branch, Wernigerode, Germany
| | - Franziska Layer
- National Reference Centre for Staphylococci and Enterococci, Division 13: Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode Branch, Wernigerode, Germany
| |
Collapse
|
74
|
Layer F, Vourli S, Karavasilis V, Strommenger B, Dafopoulou K, Tsakris A, Werner G, Pournaras S. Dissemination of linezolid-dependent, linezolid-resistant Staphylococcus epidermidis clinical isolates belonging to CC5 in German hospitals. J Antimicrob Chemother 2019; 73:1181-1184. [PMID: 29360979 DOI: 10.1093/jac/dkx524] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/14/2017] [Indexed: 11/13/2022] Open
Abstract
Objectives Linezolid-resistant Staphylococcus epidermidis (LRSE) and linezolid-dependent ST22 strains have been shown to predominate in tertiary care facilities all over Greece. We report herein the dissemination of ST22 but also ST2, ST5 and ST168 linezolid-dependent LRSE clones in four unrelated German hospitals. Methods Fourteen LRSE clinical isolates recovered during 2012-14 from five distantly located German hospitals were tested by for MIC determination broth microdilution and Etest, PCR/sequencing for cfr and for mutations in 23S rRNA, rplC, rplD and rplV genes, MLST, PFGE and growth curves without and with linezolid at 16 and 32 mg/L. Results Most (11, 78.6%) isolates had linezolid MICs >256 mg/L. Five isolates carried the cfr gene. Eight isolates belonged to ST22, two isolates each to ST168 and ST2 and one isolate each to ST5 and ST23. Ten isolates [seven belonging to ST22 and one to each of ST2, ST5 and ST168; all these STs belong to clonal complex (CC) 5] exhibited linezolid-dependent growth, growing significantly faster in linezolid-containing broth. Four isolates were non-dependent (one belonging to each of ST22, ST2, ST23 and ST168). Four isolates came from three different hospitals, whereas four and six isolates were recovered during outbreaks of LRSE in two distinct hospitals. Conclusions The multi-clonal dissemination of CC5 linezolid-dependent LRSE throughout German hospitals along with the clonal expansion of ST22 linezolid-dependent LRSE in Greek hospitals is of particular concern. It is plausible that this characteristic is inherent and provides a selective advantage to CC5 LRSE under linezolid pressure, contributing to their dissemination throughout hospitals in these countries.
Collapse
Affiliation(s)
- Franziska Layer
- Robert Koch Institute, Department of Infectious Diseases, National Centre for Staphylococci and Enterococci, Wernigerode, Germany
| | - Sophia Vourli
- Laboratory of Clinical Microbiology, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Birgit Strommenger
- Robert Koch Institute, Department of Infectious Diseases, National Centre for Staphylococci and Enterococci, Wernigerode, Germany
| | - Konstantina Dafopoulou
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanassios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Guido Werner
- Robert Koch Institute, Department of Infectious Diseases, National Centre for Staphylococci and Enterococci, Wernigerode, Germany
| | - Spyros Pournaras
- Laboratory of Clinical Microbiology, ATTIKON University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Department of Microbiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
75
|
Ogami C, Tsuji Y, To H, Yamamoto Y. Pharmacokinetics, toxicity and clinical efficacy of linezolid in Japanese pediatric patients. J Infect Chemother 2019; 25:979-986. [PMID: 31208925 DOI: 10.1016/j.jiac.2019.05.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/25/2019] [Accepted: 05/21/2019] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The aims of the present study were (a) to evaluate the pharmacokinetics of linezolid, and (b) to assess the toxicity and clinical efficacy of linezolid in Japanese pediatric patients. PATIENTS AND METHODS Routine clinical data including serum linezolid total and unbound concentrations were collected from 15 pediatric patients (0-13 years old). Pharmacokinetics of linezolid was assumed to follow one-compartment with the first-order absorption model. The relationship between risk for thrombocytopenia and linezolid concentrations, and the variations in C-reactive protein (CRP) concentrations and body temperatures were evaluated as clinical efficacy assessment. RESULTS Body weight (WT) and maturation of body function were significant covariates for pharmacokinetics of linezolid in pediatric patients. The elimination half-life of linezolid in a pediatric patient with a WT of 9.9 kg and age of 24 months (median of this study) was 3.0 h. Thrombocytopenia was detected in three patients (21.4%), and the minimum concentrations (Cmin) in these patients were significantly higher than those in patients without thrombocytopenia (P < 0.05). The CRP concentrations decreased more than 50% in all pediatric patients after the treatment with linezolid, however body temperatures at the end of treatment were higher than 37.5 °C in 6 patients (42.9%). CONCLUSIONS Although dose adjustment based on body size was performed for pediatric patients, thrombocytopenia was detected in 21.4% of pediatric patients, and higher Cmin was associated with the risk of thrombocytopenia. These results encourage the implementation of individual dose adjustment based on linezolid serum concentrations for safe and appropriate treatment with linezolid.
Collapse
Affiliation(s)
- Chika Ogami
- Department of Medical Pharmaceutics, Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Yasuhiro Tsuji
- Center for Pharmacist Education, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba, 274-8555, Japan.
| | - Hideto To
- Department of Medical Pharmaceutics, Faculty of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Yoshihiro Yamamoto
- Department of Clinical Infectious Diseases, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
76
|
Kang ZZ, Lei CW, Kong LH, Wang YL, Ye XL, Ma BH, Wang XC, Li C, Zhang Y, Wang HN. Detection of transferable oxazolidinone resistance determinants in Enterococcus faecalis and Enterococcus faecium of swine origin in Sichuan Province, China. J Glob Antimicrob Resist 2019; 19:333-337. [PMID: 31136832 DOI: 10.1016/j.jgar.2019.05.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/13/2019] [Accepted: 05/17/2019] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVES The aim of this study was to detect transferable oxazolidinone resistance determinants (cfr, optrA and poxtA) in Enterococcus faecalis and Enterococcus faecium isolates of swine origin in Sichuan Province, China. METHODS A total of 158 enterococcal isolates (93 E. faecalis and 65 E. faecium) isolated from 25 large-scale swine farms (2016-2017) were screened for the presence of cfr, optrA and poxtA by PCR. The genetic environments of cfr, optrA and poxtA were characterised by whole-genome sequencing. Transfer of oxazolidinone resistance determinants was determined by conjugation or electrotransformation experiments. RESULTS The transferable oxazolidinone resistance determinants cfr, optrA and poxtA were detected in zero, six and one enterococcal isolates, respectively. The poxtA gene in one E. faecalis isolate was located on a 37 990-bp plasmid that co-harboured fexB, cat, tet(L) and tet(M) and could be conjugated to E. faecalis JH2-2. One E. faecalis isolate harboured two different OptrA variants, including one variant with a single substitution (Q219H) that has not been reported previously. Two optrA-carrying plasmids, pC25-1 (45 581bp) and pC54 (64 500bp), shared a 40 494-bp identical region containing the genetic context IS1216E-fexA-optrA-erm(A)-IS1216E that could be electrotransformed into Staphylococcus aureus. Four different chromosomal optrA gene clusters were found in five strains, in which optrA was associated with Tn554 or Tn558 inserted into the radC gene. CONCLUSION This study highlights the fact that mobile genetic elements, such as plasmids, IS1216E, Tn554 and Tn558, may facilitate the horizontal transmission of optrA and poxtA genes.
Collapse
Affiliation(s)
- Zhuang-Zhuang Kang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, and Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, China
| | - Chang-Wei Lei
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, and Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, China
| | - Ling-Han Kong
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, and Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, China
| | - Yu-Long Wang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, and Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, China
| | - Xiao-Lan Ye
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, and Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, China
| | - Bo-Heng Ma
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, and Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, China
| | - Xue-Chun Wang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, and Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, China
| | - Cui Li
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, and Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, China
| | - Yu Zhang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, and Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, China
| | - Hong-Ning Wang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, and Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, China.
| |
Collapse
|
77
|
Talaga-Ćwiertnia K, Bulanda M. DRUG RESISTANCE IN THE GENUS ENTEROCOCCUS - CURRENT PROBLEM IN HUMANS AND ANIMALS. ADVANCEMENTS OF MICROBIOLOGY 2019. [DOI: 10.21307/pm-2018.57.3.244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
78
|
Zhang Y, Wang Y, Van Driel ML, McGuire TM, Zhang T, Dong Y, Liu Y, Liu L, Hao R, Cao L, Xing J, Dong Y. Network meta-analysis and pharmacoeconomic evaluation of antibiotics for the treatment of patients infected with complicated skin and soft structure infection and hospital-acquired or ventilator-associated penumonia. Antimicrob Resist Infect Control 2019; 8:72. [PMID: 31080587 PMCID: PMC6501412 DOI: 10.1186/s13756-019-0518-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/04/2019] [Indexed: 01/18/2023] Open
Abstract
Background Infections due to methicillin-resistant Staphylococcus aureus (MRSA) cause serious health risks and significant economic burdens and the preferred drugs are still controversial. Methods We performed a network meta-analysis (NMA) to compare the efficacy and safety of antibiotics used to treat inpatients with complicated skin and soft structure infections (cSSSI) or hospital-acquired or ventilator-associated pneumonia (HAP/VAP). We also developed a decision tree model to assess the cost-effectiveness of antibiotics. Results Forty-nine randomized controlled trials met the inclusion criteria (34 for cSSSI, 15 for HAP/VAP) and compared the efficacy and safety of 16 antibiotics. For cSSSI, NMA indicated that for clinical cure, linezolid was superior than vancomycin (odds ratio (OR) 1.55, 95% confidence interval (CI) 1.19-2.02), while tedizolid (OR 1.39, CI 0.70-2.76) was similar to vancomycin. In terms of safety, there were no significant differences between any two interventions on total adverse events. Based on drug and hospital costs in America, the incremental cost-effectiveness ratios (ICERs) per life-year saved for linezolid and tedizolid compared with vancomycin were US$2833 and US$5523. For HAP/VAP, there were no significant effects either for clinical cure or for safety endpoints between linezolid and vancomycin in NMA. ICERs per life-year saved for linezolid compared with vancomycin were US$2185. Conclusion In these clinical trials, considering efficacy, safety, and cost-effectivenes, linezolid and tedizolid showed their superiority in MRSA cSSSI; while linezolid might be recommended to treat MRSA pneumonia. Although vancomycin was not cost-effective in pharmacoeconomic evaluation, it is still the first-line treatment for MRSA infection in the clinical practice. This study might provide new insights of therapeutic choices for patients with MRSA infections whilst awaiting the arrival of higher quality evidence.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 China
| | - Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 China
- School of Pharmacy, University of Queensland, Brisbane, Queensland Australia
| | - Mieke L. Van Driel
- Primary Care Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland Australia
| | - Treasure M. McGuire
- School of Pharmacy, University of Queensland, Brisbane, Queensland Australia
- Faculty of Health Sciences & Medicine, Bond University, Gold Coast, Queensland Australia
- Mater Pharmacy Services, Mater Health Services, Brisbane, Queensland Australia
| | - Tao Zhang
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 China
| | - Yuzhu Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 China
| | - Yang Liu
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 China
| | - Leichao Liu
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 China
| | - Ruifang Hao
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 China
| | - Lu Cao
- Department of Pharmacy, Xi’an Jiaotong University, Xi’an, 710061 China
| | - Jianfeng Xing
- Department of Pharmacy, Xi’an Jiaotong University, Xi’an, 710061 China
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061 China
| |
Collapse
|
79
|
Fernandes GFDS, Salgado HRN, Santos JLD. A critical review of HPLC-based analytical methods for quantification of Linezolid. Crit Rev Anal Chem 2019; 50:196-211. [PMID: 31017000 DOI: 10.1080/10408347.2019.1605876] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Linezolid is a synthetic antimicrobial agent belonging to the oxazolidinone class. Since its approval in the year 2000 until now, linezolid remains the main representative drug for the oxazolidinone class of drugs, which is used in therapy due to its unique mode of action, which involves inhibition of protein synthesis. As linezolid holds great importance in antimicrobial therapy, it is necessary to compile the various analytical methods that have been reported in the literature for its analysis. Analytical techniques used for pharmaceutical analyses and therapeutic drug monitoring play an important role in comprehending the aspects regarding bioavailability, bioequivalence, and therapeutic monitoring during patient follow-ups. Even though linezolid has had the approval for clinical use for more than 18 years now, most of the analytical methods for its determination reported in the scientific literature are the ones which utilize HPLC. Therefore, the present review provides a summary of the HPLC-based methods used in the determination and quantification of linezolid in different matrices since the time of its discovery.
Collapse
Affiliation(s)
- Guilherme Felipe Dos Santos Fernandes
- Institute of Chemistry, São Paulo State University, Araraquara, Brazil.,School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | | | - Jean Leandro Dos Santos
- Institute of Chemistry, São Paulo State University, Araraquara, Brazil.,School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| |
Collapse
|
80
|
Babiker A, Mustapha MM, Pacey MP, Shutt KA, Ezeonwuka CD, Ohm SL, Cooper VS, Marsh JW, Doi Y, Harrison LH. Use of online tools for antimicrobial resistance prediction by whole-genome sequencing in methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE). J Glob Antimicrob Resist 2019; 19:136-143. [PMID: 31005733 DOI: 10.1016/j.jgar.2019.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 03/31/2019] [Accepted: 04/06/2019] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES The antimicrobial resistance (AMR) crisis represents a serious threat to public health and has resulted in concentrated efforts to accelerate development of rapid molecular diagnostics for AMR. In combination with publicly available web-based AMR databases, whole-genome sequencing (WGS) offers the capacity for rapid detection of AMR genes. Here we studied the concordance between WGS-based resistance prediction and phenotypic susceptibility test results for methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE) clinical isolates using publicly available tools and databases. METHODS Clinical isolates prospectively collected at the University of Pittsburgh Medical Center between December 2016 and December 2017 underwent WGS. The AMR gene content was assessed from assembled genomes by BLASTn search of online databases. Concordance between the WGS-predicted resistance profile and phenotypic susceptibility as well as the sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV) were calculated for each antibiotic/organism combination, using the phenotypic results as gold standard. RESULTS Phenotypic susceptibility testing and WGS results were available for 1242 isolate/antibiotic combinations. Overall concordance was 99.3%, with a sensitivity, specificity, PPV and NPV of 98.7% (95% CI 97.2-99.5%), 99.6% (95% CI 98.8-99.9%), 99.3% (95% CI 98.0-99.8%) and 99.2% (95% CI 98.3-99.7%), respectively. Additional identification of point mutations in housekeeping genes increased the concordance to 99.4%, sensitivity to 99.3% (95% CI 98.2-99.8%) and NPV to 99.4% (95% CI 98.4-99.8%). CONCLUSION WGS can be used as a reliable predicator of phenotypic resistance both for MRSA and VRE using readily available online tools.
Collapse
Affiliation(s)
- Ahmed Babiker
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Microbial Genomic Epidemiology Laboratory, Infectious Diseases Epidemiology Research Unit, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Mustapha M Mustapha
- Microbial Genomic Epidemiology Laboratory, Infectious Diseases Epidemiology Research Unit, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marissa P Pacey
- Microbial Genomic Epidemiology Laboratory, Infectious Diseases Epidemiology Research Unit, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathleen A Shutt
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Microbial Genomic Epidemiology Laboratory, Infectious Diseases Epidemiology Research Unit, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chinelo D Ezeonwuka
- Microbial Genomic Epidemiology Laboratory, Infectious Diseases Epidemiology Research Unit, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sara L Ohm
- Microbial Genomic Epidemiology Laboratory, Infectious Diseases Epidemiology Research Unit, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vaughn S Cooper
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jane W Marsh
- Microbial Genomic Epidemiology Laboratory, Infectious Diseases Epidemiology Research Unit, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yohei Doi
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lee H Harrison
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Microbial Genomic Epidemiology Laboratory, Infectious Diseases Epidemiology Research Unit, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
81
|
Huang J, Sun J, Wu Y, Chen L, Duan D, Lv X, Wang L. Identification and pathogenicity of an XDR Streptococcus suis isolate that harbours the phenicol-oxazolidinone resistance genes optrA and cfr, and the bacitracin resistance locus bcrABDR. Int J Antimicrob Agents 2019; 54:43-48. [PMID: 30981924 DOI: 10.1016/j.ijantimicag.2019.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/29/2019] [Accepted: 04/06/2019] [Indexed: 12/30/2022]
Abstract
One hundred and seven Streptococcus suis isolates were collected from healthy pigs or asymptomatic carriers in Jiangsu, China in 2016-2017. Thirty-eight percent of the isolates were linezolid-resistant and all carried the optrA gene. Among them, one isolate, SFJ44, was resistant to all 20 of the antibiotics tested, except for ceftiofur, and thus exhibited an extensively-drug-resistant phenotype. This isolate carried the optrA gene and the bacitracin resistance locus bcrABDR on an antibiotic-resistance-associated genomic island (ARGI1), and harboured the resistance genes cfr, aadE, sat4, spw-like, aphA3, mef(A), msr(D), erm(A)-like, erm(B), tetAB(P)', tet(M) and catQ on ARGI2∼4. The IS1216E-bcrABDR-ISEnfa1 segment showed >99.9% sequence identity to corresponding sequences from other species. The cfr gene was located on ARGI4, and two IS6 family insertion sequences, IS1216E and ISTeha2, were found upstream and downstream of cfr-ΔISEnfa5, respectively. A circular intermediate of bcrABDR-ISEnfa1 was detected, suggesting the role of ISEnfa1 in dissemination of bcrABDR. Other antibiotic resistance genes might be acquired from different Gram-positive pathogens. Infection of zebrafish showed that SFJ44 exhibited a virulence level comparable to serotype 2 hypervirulent strain SC070731, highlighting the need for surveillance of the pathogenicity of multi-drug-resistant S. suis isolates. This is the first report of the co-existence of optrA and cfr, and of the bcrABDR locus in streptococci. As it has been suggested that S. suis may act as an antibiotic resistance reservoir contributing to the spread of resistance genes to major streptococcal pathogens, the potential dissemination of these resistance genes among Gram-positive bacteria is of concern and routine surveillance should be strengthened.
Collapse
Affiliation(s)
- Jinhu Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Junjie Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuanchang Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Li Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Duan Duan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xi Lv
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Liping Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
82
|
Physicochemical Compatibility and Stability of Linezolid with Parenteral Nutrition. Molecules 2019; 24:molecules24071242. [PMID: 30934964 PMCID: PMC6480230 DOI: 10.3390/molecules24071242] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 03/22/2019] [Accepted: 03/27/2019] [Indexed: 12/12/2022] Open
Abstract
Patients referred to intensive care units (ICU) require special care due to their life-threatening condition, diseases and, frequently, malnutrition. Critically ill patients manifest a range of typical physiological changes caused by predominantly catabolic reactions in the body. It is necessary to provide the patients with proper nutrition, for example by administering total parenteral nutrition (TPN). The addition of linezolid to TPN mixtures for patients treated for linezolid-sensitive infections may reduce the extent of vascular access handling, resulting in a diminished risk of unwanted catheter-related infections. The compatibility and stability studies were conducted of linezolid in parenteral nutrition mixtures of basic, high- and low-electrolytic, high- and low-energetic and immunomodulatory composition. Mixtures containing linezolid were stored at 4–6 °C and 25 °C with light protection and at 25 °C without light protection for 168 h. In order to evaluate changes in the concentration of linezolid a previously validated reversed-phase HPLC method with UV detection was used. It was found that linezolid was stable at 4–6 °C in the whole course of the study whereas at 25 °C it proved stable over a period of 24 h required for administration of parenteral nutrition mixtures. The TPN mixtures demonstrated compatibility with linezolid and suitable stability, which were not affected by time or storage conditions.
Collapse
|
83
|
Low Prevalence of Gram-Positive Isolates Showing Elevated Lefamulin MIC Results during the SENTRY Surveillance Program for 2015-2016 and Characterization of Resistance Mechanisms. Antimicrob Agents Chemother 2019; 63:AAC.02158-18. [PMID: 30670418 DOI: 10.1128/aac.02158-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 01/18/2019] [Indexed: 12/31/2022] Open
Abstract
This study investigated the molecular mechanisms possibly associated with non-wild-type MICs for lefamulin among staphylococci and streptococci included in the lefamulin surveillance program from 2015 to 2016. A total of 2,919 Staphylococcus aureus, 276 coagulase-negative staphylococci (CoNS), 3,923 Streptococcus pneumoniae, 389 β-hemolytic, and 178 viridans group streptococci isolates were included in the surveillance studies. Eleven (0.3% of all S. aureus) S. aureus isolates with lefamulin MICs above the staphylococcal epidemiological cutoff (ECOFF) value (>0.25 μg/ml) were selected for this study. Eight (72.7%) S. aureus (lefamulin MIC, 0.5 to 4 μg/ml) isolates carried vga(A or E), one isolate (MIC, 32 μg/ml) carried lsa(E), one isolate (MIC, 16 μg/ml) had an alteration in L4, and one strain (MIC, 0.5 μg/ml) did not carry any of the investigated resistance mechanisms. A total of 14 (5.1% of all CoNS) CoNS isolates had lefamulin MICs (0.5 to >32 μg/ml) above the ECOFF. Similar to S. aureus, 8 (57.1%) CoNS (lefamulin MIC, 1 to 8 μg/ml) isolates carried vga(A or B), while 2 isolates (MIC, 4 to 32 μg/ml) carried cfr High genetic diversity was observed among staphylococci, although 3 S. aureus isolates belonged to sequence type 398 (ST398). Among the 3 Streptococcus agalactiae and 3 viridans group streptococci (0.1% of all streptococci surveyed) isolates selected for additional characterization, all but 1 isolate carried lsa(E). This study documents a low occurrence of surveillance isolates exhibiting a non-wild-type MIC for lefamulin, and among these isolates, vga and lsa(E) prevailed in staphylococci and streptococci, respectively.
Collapse
|
84
|
Mendes RE, Jones RN, Woosley LN, Cattoir V, Castanheira M. Application of Next-Generation Sequencing for Characterization of Surveillance and Clinical Trial Isolates: Analysis of the Distribution of β-lactamase Resistance Genes and Lineage Background in the United States. Open Forum Infect Dis 2019; 6:S69-S78. [PMID: 30895217 PMCID: PMC6419912 DOI: 10.1093/ofid/ofz004] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background Sequencing technologies and techniques have seen remarkable transformation and innovation that have significantly affected sequencing capability. Data analyses have replaced sequencing as the main challenge. This paper provides an overview on applying next-generation sequencing (NGS) and analysis and discusses the benefits and challenges. In addition, this document shows results from using NGS and bioinformatics tools to screen for β-lactamase genes and assess the epidemiological structure of Escherichia coli– and Klebsiella pneumoniae–causing bloodstream (BSIs) and urinary tract (UTIs) infections in patients hospitalized in the United States during the SENTRY Antimicrobial Surveillance Program for 2016. Methods A total of 3525 isolates (2751 E. coli and 774 K. pneumoniae) causing BSIs (n = 892) and UTIs (n = 2633) in hospitalized patients in the United States were included. Isolates were tested for susceptibility by broth microdilution, and those that met a minimum inhibitory concentration (MIC)–based screening criteria had their genomes sequenced and analyzed. Results A total of 11.6% and 16.1% of E. coli–causing UTIs and BSIs, respectively, met the MIC-based criteria, whereas 11.0% and 13.7% of K. pneumoniae isolates causing UTIs and BSIs, respectively, met the criteria. Among E. coli, blaCTX-M variants (87.6% overall) prevailed (60.5% of CTX-M group 1 and 26.9% of group 9). A total of 60.3% of K. pneumoniae isolates carried blaCTX-M variants (52.7% and 7.6% of groups 1 and 9, respectively). Two E. coli (0.6%) and 13 K. pneumoniae (12.9%) isolates harbored blaKPC. Among KPC-producing K. pneumoniae (2 from BSIs and 11 from UTIs), 84.6% (11/13) were ST258 (CC258). Seventeen and 38 unique clonal complexes (CCs) were noted in E. coli that caused BSIs and UTIs, respectively, and CC131 (or ST131) was the most common CC among BSI (53.6%) and UTI (58.2%) isolates. Twenty-three and 26 CCs were noted among K. pneumoniae–causing BSIs and UTIs, respectively. CC258 (28.3%) prevailed in UTI pathogens, whereas CC307 (15.0%) was the most common CC among BSI isolates. Conclusions This study provides a benchmark for the distribution of β-lactamase genes and the population structure information for the most common Enterobacteriaceae species responsible for BSIs and UTIs in US medical centers during the 2016 SENTRY Program.
Collapse
Affiliation(s)
| | | | | | - Vincent Cattoir
- University Hospital of Rennes, Department of Clinical Microbiology, Rennes, France.,National Reference Center for Antimicrobial Resistance, Rennes, France.,University of Rennes 1, Unit Inserm U1230, Rennes, France
| | | |
Collapse
|
85
|
Na SH, Moon DC, Choi MJ, Oh SJ, Jung DY, Kang HY, Hyun BH, Lim SK. Detection of oxazolidinone and phenicol resistant enterococcal isolates from duck feces and carcasses. Int J Food Microbiol 2019; 293:53-59. [DOI: 10.1016/j.ijfoodmicro.2019.01.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/26/2018] [Accepted: 01/04/2019] [Indexed: 01/08/2023]
|
86
|
Vestergaard M, Frees D, Ingmer H. Antibiotic Resistance and the MRSA Problem. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0057-2018. [PMID: 30900543 PMCID: PMC11590431 DOI: 10.1128/microbiolspec.gpp3-0057-2018] [Citation(s) in RCA: 256] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Indexed: 12/29/2022] Open
Abstract
Staphylococcus aureus is capable of becoming resistant to all classes of antibiotics clinically available and resistance can develop through de novo mutations in chromosomal genes or through acquisition of horizontally transferred resistance determinants. This review covers the most important antibiotics available for treatment of S. aureus infections and a special emphasis is dedicated to the current knowledge of the wide variety of resistance mechanisms that S. aureus employ to withstand antibiotics. Since resistance development has been inevitable for all currently available antibiotics, new therapies are continuously under development. Besides development of new small molecules affecting cell viability, alternative approaches including anti-virulence and bacteriophage therapeutics are being investigated and may become important tools to combat staphylococcal infections in the future.
Collapse
Affiliation(s)
- Martin Vestergaard
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Dorte Frees
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Hanne Ingmer
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
87
|
Lu M, Yang X, Yu C, Gong Y, Yuan L, Hao L, Sun S. Linezolid in Combination With Azoles Induced Synergistic Effects Against Candida albicans and Protected Galleria mellonella Against Experimental Candidiasis. Front Microbiol 2019; 9:3142. [PMID: 30766527 PMCID: PMC6365414 DOI: 10.3389/fmicb.2018.03142] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 12/04/2018] [Indexed: 12/11/2022] Open
Abstract
The incidence of resistant Candida isolates has increased continuously in recent decades, especially Candida albicans. To overcome this resistance, research on antifungal sensitizers has attracted considerable attention. Linezolid was found to inhibit the growth of Pythium insidiosum and synergize with amphotericin B against Cryptococcus neoformans. The objective of this study was to determine the interactions of linezolid and azoles against C. albicans in vitro and in vivo. In vitro, linezolid combined with azoles induced synergistic effects not only against some susceptible C. albicans isolates, but also against all tested resistant C. albicans isolates. For all resistant isolates, exposure to the combination of linezolid with azoles induced a significant decrease in the minimum inhibitory concentrations (MIC) of azoles, from >512 to 0.5–1 μg/mL for fluconazole, from >16 to 0.25–1 μg/mL for itraconazole, and from >16 to 0.03–0.25 μg/mL for voriconazole. Additionally, linezolid synergized with fluconazole against biofilms that were preformed for ≤ 12 h from both susceptible and resistant C. albicans, and the sessile MIC of fluconazole decreased from >1024 to 1–4 μg/mL. In vivo, linezolid plus azoles prolonged the survival rate of infected Galleria mellonella larvae twofold compared with the azole monotherapy group, significantly decreased the fungal burden of the infected larvae, and reduced the damage of resistant C. albicans to the larval tissue. These findings will contribute to antifungal agent discovery and new approaches for the treatment of candidiasis caused by C. albicans.
Collapse
Affiliation(s)
- Mengjiao Lu
- School of Pharmaceutical Sciences, Shandong University, Ji'nan, China.,Department of Pharmacy, Baodi People's Hospital, Tianjin, China
| | - Xinmei Yang
- Department of Pharmacy, Qianfoshan Hospital Affiliated to Shandong University, Ji'nan, China
| | - Cuixiang Yu
- Department of Respiration Medicine, Qianfoshan Hospital Affiliated to Shandong University, Ji'nan, China
| | - Ying Gong
- School of Pharmaceutical Sciences, Shandong University, Ji'nan, China
| | - Lei Yuan
- Department of Pharmacy, Baodi People's Hospital, Tianjin, China
| | - Lina Hao
- Qilu Children's Hospital of Shandong University, Ji'nan, China
| | - Shujuan Sun
- Department of Pharmacy, Qianfoshan Hospital Affiliated to Shandong University, Ji'nan, China
| |
Collapse
|
88
|
Zhou W, Gao S, Xu H, Zhang Z, Chen F, Shen H, Zhang C. Distribution of the optrA gene in Enterococcus isolates at a tertiary care hospital in China. J Glob Antimicrob Resist 2019; 17:180-186. [PMID: 30641287 DOI: 10.1016/j.jgar.2019.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVES Linezolid-resistant Enterococcus have spread worldwide. This study investigated the prevalence of linezolid-non-susceptible Enterococcus (LNSE) and the potential mechanism and molecular epidemiology of LNSE isolates from Nanjing, China. METHODS Linezolid susceptibility of 2555 Enterococcus was retrospectively determined by Etest. Vancomycin and teicoplanin MICs were determined for LNSE by Etest. PCR and DNA sequencing were used to investigate the potential molecular mechanism. Clonal relatedness between LNSE isolates was analysed by MLST. WGS was also performed. RESULTS A total of 27 Enterococcus isolates (24 Enterococcus faecalis, 3 Enterococcus faecium) with linezolid MICs of 4-48μg/mL were identified, among which 20 E. faecalis and 3 E. faecium were positive for optrA. No mutations were found in genes encoding domain V of 23S rRNA or ribosomal proteins L3/L4; the cfr gene was not found. The 24 linezolid-non-susceptible E. faecalis were classified into eight STs (ST16, ST480, ST476, ST631, ST585, ST428, ST25 and ST689). The three linezolid-non-susceptible E. faecium were classified as ST17, ST400 and ST195. Comparison of the deduced OptrA amino acid sequences of the 23 optrA-positive isolates by PCR-based sequencing and WGS with that of the original OptrA from E. faecalis E349 revealed seven variants (KD, EDP, EDM, D, EDD, RDK and DP) in 16 isolates, with no mutations in the remaining 7 isolates. optrA was found downstream of fexA by searching the pE349 sequence based on WGS data. CONCLUSIONS Emergence of LNSE with optrA-mediated resistance and clonal dissemination of ST16 E. faecalis in our hospital may pose a potential public-health threat.
Collapse
Affiliation(s)
- Wanqing Zhou
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Gulou District, Nanjing, Jiangsu Province 210008, PR China
| | - Shuo Gao
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Gulou District, Nanjing, Jiangsu Province 210008, PR China
| | - Hongjing Xu
- Department of Laboratory Medicine, Jiangning District Hospital of Traditional Chinese Medicine, 657# Tianyin Avenue, Jiangning District, Nanjing, Jiangsu Province 211100, PR China
| | - Zhifeng Zhang
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Gulou District, Nanjing, Jiangsu Province 210008, PR China
| | - Fei Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Gulou District, Nanjing, Jiangsu Province 210008, PR China
| | - Han Shen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Gulou District, Nanjing, Jiangsu Province 210008, PR China.
| | - Chunni Zhang
- Department of Clinical Laboratory, Jinling Hospital, Nanjing University School of Medicine, Nanjing University, 305# East Zhongshan Road, Qinhuai District, Nanjing, Jiangsu Province 210008, PR China.
| |
Collapse
|
89
|
Kim SY, Jhun BW, Moon SM, Jeon K, Kwon OJ, Huh HJ, Lee NY, Shin SJ, Daley CL, Koh WJ. Genetic mutations in linezolid-resistant Mycobacterium avium complex and Mycobacterium abscessus clinical isolates. Diagn Microbiol Infect Dis 2018; 94:38-40. [PMID: 30581010 DOI: 10.1016/j.diagmicrobio.2018.10.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/15/2018] [Accepted: 10/15/2018] [Indexed: 02/08/2023]
Abstract
There are no studies evaluating the mechanisms driving linezolid resistance in nontuberculous mycobacteria. The novel mutations G2599A and A2137T in the 23S rRNA gene and mutations A439G and G443A in the rplD gene associated with linezolid resistance were identified in linezolid-resistant M. avium complex isolates.
Collapse
Affiliation(s)
- Su-Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Byung Woo Jhun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seong Mi Moon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyeongman Jeon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - O Jung Kwon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hee Jae Huh
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Nam Yong Lee
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Charles L Daley
- Division of Mycobacterial and Respiratory Infections, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Won-Jung Koh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
90
|
Emergence and Within-Host Genetic Evolution of Methicillin-Resistant Staphylococcus aureus Resistant to Linezolid in a Cystic Fibrosis Patient. Antimicrob Agents Chemother 2018; 62:AAC.00720-18. [PMID: 30275089 DOI: 10.1128/aac.00720-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 09/22/2018] [Indexed: 12/26/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infection has increased in recent years among cystic fibrosis (CF) patients. Linezolid (LZD) is one of the antistaphylococcal antibiotics widely used in this context. Although LZD resistance is rare, it has been described as often associated with long-term treatments. Thirteen MRSA strains isolated over 5 years from one CF patient were studied for LZD resistance emergence and subjected to whole-genome sequencing (WGS). Resistance emerged after three 15-day LZD therapeutic regimens over 4 months. It was associated with the mutation of G to T at position 2576 (G2576T) in all 5 rrl copies, along with a very high MIC (>256 mg/liter) and a strong increase in the generation time. Resistant strains isolated during the ensuing LZD therapeutic regimens and until 13 months after LZD stopped harbored only 3 or 4 mutated rrl copies, associated with lower MICs (8 to 32 mg/liter) and low to moderate generation time increases. Despite these differences, whole-genome sequencing allowed us to determine that all isolates, including the susceptible one isolated before LZD treatment, belonged to the same lineage. In conclusion, LZD resistance can emerge rapidly in CF patients and persist without linezolid selective pressure in colonizing MRSA strains belonging to the same lineage.
Collapse
|
91
|
Hua R, Xia Y, Wu W, Yang M, Yan J. Molecular Epidemiology and Mechanisms of 43 Low-Level Linezolid-Resistant Enterococcus faecalis Strains in Chongqing, China. Ann Lab Med 2018; 39:36-42. [PMID: 30215228 PMCID: PMC6143467 DOI: 10.3343/alm.2019.39.1.36] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/12/2018] [Accepted: 08/16/2018] [Indexed: 12/02/2022] Open
Abstract
Background Enterococcus faecalis strains with low-level resistance to linezolid (an oxazolidinone antibiotic) have become common. No large-scale study has examined the underlying mechanisms in linezolid-resistant E. faecalis (LRE) strains. We investigated these mechanisms and molecular characteristics in Chongqing, China. Methods A total of 1,120 non-duplicated E. faecalis strains collected from August 2014 to June 2017 underwent drug susceptibility testing. LRE strains were screened for optrA, cfr, and mutations in the 23S rRNA and ribosomal proteins L3 and L4 by PCR amplification and sequencing. Multi-locus sequence typing (MLST) and pulsed-field gel electrophoresis (PFGE) were used for epidemiological analysis. Results All 43 low-level LRE strains (minimum inhibitory concentration: 8–16 mg/L) harbored optrA; cfr and 23S rRNA mutations were not detected. Novel mutations in the ribosomal proteins L3 and L4—one deletion (Q103del) and four substitutions (S113L, T35A, I98V, and N79D)—were identified. Novel amino acid substitutions at positions E60K, G197D, and T285P of the OptrA protein were observed. MLST revealed 20 types of LRE strains; the most common type was ST16 (32.6%). PFGE showed 14 strains of ST16 with unique banding patterns. Eight novel sequence types (ST823 to ST830) and one allele (gki95) were identified for the first time in China. Conclusions optrA plays an important role in linezolid resistance and may serve as a marker for resistance screening. Since the L3 and L4 mutations did not simultaneously occur in the same strain, they play a negligible role in linezolid resistance. Epidemiological investigation suggested that the LRE cases were sporadic.
Collapse
Affiliation(s)
- Ruoyi Hua
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun Xia
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Wenyao Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mi Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jia Yan
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
92
|
Chen M, Pan H, Lou Y, Wu Z, Zhang J, Huang Y, Yu W, Qiu Y. Epidemiological characteristics and genetic structure of linezolid-resistant Enterococcus faecalis. Infect Drug Resist 2018; 11:2397-2409. [PMID: 30538507 PMCID: PMC6251436 DOI: 10.2147/idr.s181339] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Objectives The aim of this study was to investigate the mechanism of linezolid resistance and evaluate the risk factors for linezolid-resistant Enterococcus faecalis (LZR-Efa) infections. Methods A total of 730 E. faecalis isolates were collected, and whole-genome sequencing and bioinformatics analysis were performed. Meanwhile, risk factors related to linezolid resistance were analyzed by binary logistic regression. Results Twenty-six LZR-Efa were isolated from various clinical samples, and 24 isolates were multidrug resistant. Four isolates were daptomycin nonsusceptible, while all LZR-Efa were susceptible to vancomycin. Thirteen different sequence types (STs) were identified, and the most prevalent type was ST16 (23.1%). The genes dfrE, lsaA, and emeA were identified in all isolates. A total of 23 E. faecalis were positive for optrA gene, and six amino acids mutations were identified among 18 LZR-Efa in OptrA. The 23S rRNA mutation was found in 16 LZR-Efa isolates. However, the presence of cfr was not identified. Furthermore, there were 41 virulence genes detected, and 10 genes (ace, bopD, cpsA, cpsB, ebpB, ebpC, efaA, fss1, fss2, and srtC) were found in all isolates. A total of nine isolates were positive for multiple virulent factors (ace, asa1, cylA, efaA, esp, and gelE). There was no difference in the number of virulence factors among different specimens (P=0.825). It is of note that all patients had not been prescribed linezolid or traveled abroad previously. Moreover, previous use of carbapenems was a risk factor for LZR-Efa infections. Conclusion The main trends of LZR-Efa, with lower level of resistance, were sporadic mainly in the department of surgery. optrA and 23S rRNA were the main resistance mechanisms. In addition, carbapenems use was an independent predictor of LZR-Efa infections.
Collapse
Affiliation(s)
- Meijuan Chen
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China,
| | - Hongying Pan
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China,
| | - Yaling Lou
- Department of Infectious Diseases, Dongyang People's Hospital, Jinhua, China
| | - Zhe Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China, ,
| | - Jiajie Zhang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China,
| | - Yicheng Huang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China,
| | - Wei Yu
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China, .,State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China, ,
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China, ,
| |
Collapse
|
93
|
Kuroda M, Sekizuka T, Matsui H, Suzuki K, Seki H, Saito M, Hanaki H. Complete Genome Sequence and Characterization of Linezolid-Resistant Enterococcus faecalis Clinical Isolate KUB3006 Carrying a cfr(B)-Transposon on Its Chromosome and optrA-Plasmid. Front Microbiol 2018; 9:2576. [PMID: 30410481 PMCID: PMC6209644 DOI: 10.3389/fmicb.2018.02576] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/09/2018] [Indexed: 01/01/2023] Open
Abstract
Linezolid (LZD) has become one of the most important antimicrobial agents for infections caused by gram-positive bacteria, including those caused by Enterococcus species. LZD-resistant (LR) genetic features include mutations in 23S rRNA/ribosomal proteins, a plasmid-borne 23S rRNA methyltransferase gene cfr, and ribosomal protection genes (optrA and poxtA). Recently, a cfr gene variant, cfr(B), was identified in a Tn6218-like transposon (Tn) in a Clostridioides difficile isolate. Here, we isolated an LR Enterococcus faecalis clinical isolate, KUB3006, from a urine specimen of a patient with urinary tract infection during hospitalization in 2017. Comparative and whole-genome analyses were performed to characterize the genetic features and overall antimicrobial resistance genes in E. faecalis isolate KUB3006. Complete genome sequencing of KUB3006 revealed that it carried cfr(B) on a chromosomal Tn6218-like element. Surprisingly, this Tn6218-like element was almost (99%) identical to that of C. difficile Ox3196, which was isolated from a human in the UK in 2012, and to that of Enterococcus faecium 5_Efcm_HA-NL, which was isolated from a human in the Netherlands in 2012. An additional oxazolidinone and phenicol resistance gene, optrA, was also identified on a plasmid. KUB3006 is sequence type (ST) 729, suggesting that it is a minor ST that has not been reported previously and is unlikely to be a high-risk E. faecalis lineage. In summary, LR E. faecalis KUB3006 possesses a notable Tn6218-like-borne cfr(B) and a plasmid-borne optrA. This finding raises further concerns regarding the potential declining effectiveness of LZD treatment in the future.
Collapse
Affiliation(s)
- Makoto Kuroda
- Pathogen Genomics Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tsuyoshi Sekizuka
- Pathogen Genomics Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hidehito Matsui
- Infection Control Research Center, Kitasato University, Tokyo, Japan
| | - Katsunori Suzuki
- Division of Infection Control and Prevention, University of Occupational and Environmental Health, Kitakyusyu, Japan
| | - Hiroyuki Seki
- Division of Infection Control and Prevention, University of Occupational and Environmental Health, Kitakyusyu, Japan
| | - Mitsumasa Saito
- Division of Infection Control and Prevention, University of Occupational and Environmental Health, Kitakyusyu, Japan.,Department of Microbiology, School of Medicine, University of Occupational and Environmental Health, Kitakyusyu, Japan
| | - Hideaki Hanaki
- Infection Control Research Center, Kitasato University, Tokyo, Japan
| |
Collapse
|
94
|
Greene MH, Harris BD, Nesbitt WJ, Watson ML, Wright PW, Talbot TR, Nelson GE. Risk Factors and Outcomes Associated With Acquisition of Daptomycin and Linezolid-Nonsusceptible Vancomycin-Resistant Enterococcus. Open Forum Infect Dis 2018; 5:ofy185. [PMID: 30320147 PMCID: PMC6176497 DOI: 10.1093/ofid/ofy185] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/31/2018] [Indexed: 02/07/2023] Open
Abstract
Background Vancomycin-resistant enterococcus (VRE) causes substantial health care–associated infection with increasing reports of resistance to daptomycin or linezolid. We conducted a case–control study reporting 81 cases of daptomycin and linezolid–nonsusceptible VRE (DLVRE), a resistance pattern not previously reported. Methods We reviewed VRE isolates from June 2010 through June 2015 for nonsusceptibility to both daptomycin (minimum inhibitory concentration [MIC] > 4) and linezolid (MIC ≥ 4). We matched cases by year to control patients with VRE susceptible to both daptomycin and linezolid and performed retrospective chart review to gather risk factor and outcome data. Results We identified 81 DLVRE cases. Resistance to both daptomycin and linezolid was more common than resistance to either agent individually. Compared with susceptible VRE, DLVRE was more likely to present as bacteremia without focus (P < 0.01), with DLVRE patients more likely to be immune suppressed (P = .04), to be neutropenic (P = .03), or to have had an invasive procedure in the prior 30 days (P = .04). Any antibiotic exposure over the prior 30 days conferred a 4-fold increased risk for DLVRE (odds ratio [OR], 4.25; 95% confidence interval [CI], 1.43−12.63; P = .01); multivariate analysis implicated daptomycin days of therapy (DOT) over the past year as a specific risk factor (OR, 1.10; 95% CI, 1.01−1.19; P = .03). DLVRE cases had longer hospitalizations (P = .04) but no increased risk for in-hospital death. Conclusions DLVRE is an emerging multidrug-resistant pathogen associated with immune suppression, neutropenia, and recent invasive procedure. Prior antibiotic exposure, specifically daptomycin exposure, confers risk for acquisition of DLVRE.
Collapse
Affiliation(s)
| | - Bryan D Harris
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | - Patty W Wright
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | |
Collapse
|
95
|
Update on prevalence and mechanisms of resistance to linezolid, tigecycline and daptomycin in enterococci in Europe: Towards a common nomenclature. Drug Resist Updat 2018; 40:25-39. [DOI: 10.1016/j.drup.2018.10.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 08/10/2018] [Accepted: 10/30/2018] [Indexed: 01/04/2023]
|
96
|
Càmara J, Camoez M, Tubau F, Pujol M, Ayats J, Ardanuy C, Domínguez MÁ. Detection of the Novel optrA Gene Among Linezolid-Resistant Enterococci in Barcelona, Spain. Microb Drug Resist 2018; 25:87-93. [PMID: 30153086 DOI: 10.1089/mdr.2018.0028] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The purpose of this study was to describe the presence of the novel optrA gene among clinical isolates of enterococci in a Spanish teaching hospital (May 2016-April 2017). optrA and cfr genes were screened by PCR in all isolates showing linezolid minimal inhibitory concentration (MIC) ≥4 mg/L. The genetic relatedness of the isolates, the presence of resistance and virulence genes, and the genetic environment of optrA were assessed by whole-genome sequencing (WGS). Six of 1,640 enterococci had linezolid MIC ≥4 mg/L. Among them, the optrA gene was detected in five Enterococcus faecalis isolated from unrelated patients. Although none of them had received linezolid or chloramphenicol, all had antecedents of recent quinolone consumption. WGS analysis revealed the existence of two different genotypes: ST585 and ST474. cfr was not detected in any of the isolates. No mutations were detected among the 23S ribosomal RNA and the ribosomal proteins L3, L4, and L22. Both genotypes also carried genes related to aminoglycoside, lincosamide, macrolide, phenicol, and tetracycline resistance. Detection of optrA in a setting with low linezolid consumption and among patients without antecedents of oxazolidinone therapy is of concern.
Collapse
Affiliation(s)
- Jordi Càmara
- 1 Microbiology Department, Hospital Universitari de Bellvitge-Universitat de Barcelona-IDIBELL, L'Hospitalet de Llobregat , Barcelona, Spain
| | - Mariana Camoez
- 1 Microbiology Department, Hospital Universitari de Bellvitge-Universitat de Barcelona-IDIBELL, L'Hospitalet de Llobregat , Barcelona, Spain .,2 Spanish Network for Research in Infectious Diseases (REIPI), Instituto de Salud Carlos III , Madrid, Spain
| | - Fe Tubau
- 1 Microbiology Department, Hospital Universitari de Bellvitge-Universitat de Barcelona-IDIBELL, L'Hospitalet de Llobregat , Barcelona, Spain .,3 CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Miquel Pujol
- 2 Spanish Network for Research in Infectious Diseases (REIPI), Instituto de Salud Carlos III , Madrid, Spain .,4 Infectious Diseases Department, Hospital Universitari de Bellvitge-Universitat de Barcelona-IDIBELL, L'Hospitalet de Llobregat , Barcelona, Spain
| | - Josefina Ayats
- 1 Microbiology Department, Hospital Universitari de Bellvitge-Universitat de Barcelona-IDIBELL, L'Hospitalet de Llobregat , Barcelona, Spain .,3 CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Ardanuy
- 1 Microbiology Department, Hospital Universitari de Bellvitge-Universitat de Barcelona-IDIBELL, L'Hospitalet de Llobregat , Barcelona, Spain .,3 CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - M Ángeles Domínguez
- 1 Microbiology Department, Hospital Universitari de Bellvitge-Universitat de Barcelona-IDIBELL, L'Hospitalet de Llobregat , Barcelona, Spain .,2 Spanish Network for Research in Infectious Diseases (REIPI), Instituto de Salud Carlos III , Madrid, Spain
| |
Collapse
|
97
|
Cai J, Schwarz S, Chi D, Wang Z, Zhang R, Wang Y. Faecal carriage of optrA-positive enterococci in asymptomatic healthy humans in Hangzhou, China. Clin Microbiol Infect 2018; 25:630.e1-630.e6. [PMID: 30076974 DOI: 10.1016/j.cmi.2018.07.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/19/2018] [Accepted: 07/21/2018] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To investigate the faecal carriage of optrA-positive enterococci among asymptomatic healthy humans in Hangzhou, China, and to characterize the genetic context of optrA. METHODS A total of 3458 stool samples from healthy individuals were collected and cultured on a selective medium containing 10 mg/L florfenicol and resulting enterococci were screened for the presence of optrA by PCR. OptrA variants were determined by amino acid sequence comparison with the original OptrA from Enterococcus faecalis E349. Whole genome sequencing and PCR mapping were performed to obtain and analyse the genetic environment of optrA. RESULTS Similar optrA carriage rates (∼3.5%) were detected in samples from adults (55/1558) and children (66/1900). Linezolid resistance rates for E. faecalis, Enterococcus faecium and other Enterococcus species were 58.5% (38/65), 42.3% (11/26) and 0% (0/31), respectively. Nineteen OptrA variants exhibiting different linezolid MICs were identified. Isolates carrying wild-type OptrA and variants RDK, KLDP, KD, D, RDKP, and EDP generally demonstrated linezolid MICs ≥8 mg/L. The OptrA variants, with fexA upstream and erm(A) downstream, were flanked by IS1216E at one or both ends. The fexA-optrA(wild-type) was located downstream of a Tn554 transposon, and was inserted into the radC gene. The EDM variant was detected in 31/73 enterococci with linezolid MICs ≤4 mg/L. Despite the variable genetic context, Tn558-araC-optrA(EDM)-erm(A)-met was the most common gene array. CONCLUSIONS This study revealed a correlation between linezolid MIC, genetic context and OptrA variant. Intestinal colonization of healthy individuals by optrA-positive enterococci is a concern, and active epidemiological surveillance of optrA is warranted.
Collapse
Affiliation(s)
- J Cai
- Clinical Microbiology Laboratory, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - S Schwarz
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, Beijing, China; Institute of Microbiology and Epizootics, Centre for Infection Medicine, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - D Chi
- Department of Laboratory, Children's Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Z Wang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - R Zhang
- Clinical Microbiology Laboratory, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Y Wang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
98
|
Dortet L, Glaser P, Kassis-Chikhani N, Girlich D, Ichai P, Boudon M, Samuel D, Creton E, Imanci D, Bonnin R, Fortineau N, Naas T. Long-lasting successful dissemination of resistance to oxazolidinones in MDR Staphylococcus epidermidis clinical isolates in a tertiary care hospital in France. J Antimicrob Chemother 2018; 73:41-51. [PMID: 29092052 PMCID: PMC5890688 DOI: 10.1093/jac/dkx370] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/10/2017] [Indexed: 01/16/2023] Open
Abstract
Objectives Patient- and procedure-related changes in modern medicine have turned CoNS into one of the major nosocomial pathogens. Treatments of CoNS infections are challenging owing to the large proportion of MDR strains and oxazolidinones often remain the last active antimicrobial molecules. Here, we have investigated a long-lasting outbreak (2010-13) due to methicillin- and linezolid-resistant (LR) CoNS (n = 168), involving 72 carriers and 49 infected patients. Methods Antimicrobial susceptibilities were tested by the disc diffusion method and MICs were determined by broth microdilution or Etest. The clonal relationship of LR Staphylococcus epidermidis (LRSE) was first determined using a semi-automated repetitive element palindromic PCR (rep-PCR) method. Then, WGS was performed on all cfr-positive LRSE (n = 30) and LRSE isolates representative of each rep-PCR-defined clone (n = 17). Self-transferability of cfr-carrying plasmids was analysed by filter-mating experiments. Results This outbreak was caused by the dissemination of three clones (ST2, ST5 and ST22) of LRSE. In these clones, linezolid resistance was caused by (i) mutations in the chromosome-located genes encoding the 23S RNA and L3 and L4 ribosomal proteins, but also by (ii) the dissemination of two different self-conjugative plasmids carrying the cfr gene encoding a 23S RNA methylase. By monitoring linezolid prescriptions in two neighbouring hospitals, we highlighted that the spread of LR-CoNS was strongly associated with linezolid use. Conclusions Physicians should be aware that plasmid-encoded linezolid resistance has started to disseminate among CoNS and that rational use of oxazolidinones is critical to preserve these molecules as efficient treatment options for MDR Gram-positive pathogens.
Collapse
Affiliation(s)
- Laurent Dortet
- Department of Bacteriology-Parasitology-Hygiene, Bicêtre Hospital, Assistance Publique - Hôpitaux de Paris, Le Kremlin-Bicêtre, France.,EA7361 'Structure, dynamic, function and expression of broad spectrum β-lactamases', Paris-Sud University, LabEx Lermit, Faculty of Medicine, Le Kremlin-Bicêtre, France.,Associated French National Reference Center for Antibiotic Resistance, Le Kremlin-Bicêtre, France.,Joint Research Unit EERA 'Evolution and Ecology of Resistance to Antibiotics', Institut Pasteur-APHP-University Paris Sud, Paris, France
| | - Philippe Glaser
- Joint Research Unit EERA 'Evolution and Ecology of Resistance to Antibiotics', Institut Pasteur-APHP-University Paris Sud, Paris, France.,UMR 3525, CNRS, 75015 Paris, France
| | - Najiby Kassis-Chikhani
- Department of Hygiene, Paul Brousse Hospital, Assistance Publique - Hôpitaux de Paris, Villejuif, France
| | - Delphine Girlich
- EA7361 'Structure, dynamic, function and expression of broad spectrum β-lactamases', Paris-Sud University, LabEx Lermit, Faculty of Medicine, Le Kremlin-Bicêtre, France.,Associated French National Reference Center for Antibiotic Resistance, Le Kremlin-Bicêtre, France.,Joint Research Unit EERA 'Evolution and Ecology of Resistance to Antibiotics', Institut Pasteur-APHP-University Paris Sud, Paris, France
| | - Philippe Ichai
- Intensive Care Unit, Hepatobiliary Center, Paul Brousse Hospital, Assistance Publique - Hôpitaux de Paris, Villejuif, France
| | - Marc Boudon
- Intensive Care Unit, Hepatobiliary Center, Paul Brousse Hospital, Assistance Publique - Hôpitaux de Paris, Villejuif, France
| | - Didier Samuel
- Intensive Care Unit, Hepatobiliary Center, Paul Brousse Hospital, Assistance Publique - Hôpitaux de Paris, Villejuif, France
| | - Elodie Creton
- EA7361 'Structure, dynamic, function and expression of broad spectrum β-lactamases', Paris-Sud University, LabEx Lermit, Faculty of Medicine, Le Kremlin-Bicêtre, France.,Associated French National Reference Center for Antibiotic Resistance, Le Kremlin-Bicêtre, France.,Joint Research Unit EERA 'Evolution and Ecology of Resistance to Antibiotics', Institut Pasteur-APHP-University Paris Sud, Paris, France
| | - Dilek Imanci
- Department of Molecular Genetics and Hormonology, Assistance Publique/Hôpitaux de Paris, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Rémy Bonnin
- EA7361 'Structure, dynamic, function and expression of broad spectrum β-lactamases', Paris-Sud University, LabEx Lermit, Faculty of Medicine, Le Kremlin-Bicêtre, France.,Associated French National Reference Center for Antibiotic Resistance, Le Kremlin-Bicêtre, France.,Joint Research Unit EERA 'Evolution and Ecology of Resistance to Antibiotics', Institut Pasteur-APHP-University Paris Sud, Paris, France
| | - Nicolas Fortineau
- Department of Bacteriology-Parasitology-Hygiene, Bicêtre Hospital, Assistance Publique - Hôpitaux de Paris, Le Kremlin-Bicêtre, France.,EA7361 'Structure, dynamic, function and expression of broad spectrum β-lactamases', Paris-Sud University, LabEx Lermit, Faculty of Medicine, Le Kremlin-Bicêtre, France.,Associated French National Reference Center for Antibiotic Resistance, Le Kremlin-Bicêtre, France.,Joint Research Unit EERA 'Evolution and Ecology of Resistance to Antibiotics', Institut Pasteur-APHP-University Paris Sud, Paris, France
| | - Thierry Naas
- Department of Bacteriology-Parasitology-Hygiene, Bicêtre Hospital, Assistance Publique - Hôpitaux de Paris, Le Kremlin-Bicêtre, France.,EA7361 'Structure, dynamic, function and expression of broad spectrum β-lactamases', Paris-Sud University, LabEx Lermit, Faculty of Medicine, Le Kremlin-Bicêtre, France.,Associated French National Reference Center for Antibiotic Resistance, Le Kremlin-Bicêtre, France.,Joint Research Unit EERA 'Evolution and Ecology of Resistance to Antibiotics', Institut Pasteur-APHP-University Paris Sud, Paris, France
| |
Collapse
|
99
|
Yan J, Xia Y, Yang M, Zou J, Chen Y, Zhang D, Ma L. Quantitative Proteomics Analysis of Membrane Proteins in Enterococcus faecalis With Low-Level Linezolid-Resistance. Front Microbiol 2018; 9:1698. [PMID: 30100900 PMCID: PMC6072972 DOI: 10.3389/fmicb.2018.01698] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/09/2018] [Indexed: 11/26/2022] Open
Abstract
Despite increasing reports of low-level linezolid-resistant enterococci worldwide, the mechanism of this resistance remains poorly understood. Previous transcriptome studies of low-level linezolid-resistant Enterococcus faecalis isolates have demonstrated a number of significantly up-regulated genes potentially involved in mediation of drug resistance. However, whether the transcriptome faithfully reflects the proteome remains unknown. In this study, we performed quantitative proteomics analysis of membrane proteins in an E. faecalis isolate (P10748) with low-level linezolid-resistance in comparison with two linezolid-susceptible strains 3138 and ATCC 29212, all of which have been previously investigated by whole transcriptome analysis. A total of 8,197 peptides associated with 1,170 proteins were identified in all three isolates with false discovery rate (FDR) at 1% and P < 0.05. There were 14 significantly up-regulated and 6 significantly down-regulated proteins in strain P10748 compared to strains 3138 and ATCC 29212, which were in general positively correlated with transcription levels revealed in previous transcriptome studies. Our analysis suggests that the low-level linezolid-resistance in E. faecalis is conferred primarily by the ATP-binding cassette protein OptrA through ribosomal protection and, possibly, also by the enterococcal surface protein (Esp) and other proteins through biofilm formation. The genetic transfer of optrA is potentially regulated by the surface exclusion protein Sea1, conjugal transfer protein TraB, replication protein RepA and XRE family transcription regulator protein. This report represents the first investigation of the mechanisms of linezolid-resistance in E. faecalis by a quantitative proteomics approach.
Collapse
Affiliation(s)
- Jia Yan
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun Xia
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mi Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiaqi Zou
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yingzhu Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dawei Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liang Ma
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
100
|
Comparison of In Vitro Activity and MIC Distributions between the Novel Oxazolidinone Delpazolid and Linezolid against Multidrug-Resistant and Extensively Drug-Resistant Mycobacterium tuberculosis in China. Antimicrob Agents Chemother 2018; 62:AAC.00165-18. [PMID: 29844043 DOI: 10.1128/aac.00165-18] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/21/2018] [Indexed: 01/08/2023] Open
Abstract
Oxazolidinones are efficacious in treating mycobacterial infections, including tuberculosis (TB) caused by drug-resistant Mycobacterium tuberculosis In this study, we compared the in vitro activities and MIC distributions of delpazolid, a novel oxazolidinone, and linezolid against multidrug-resistant TB (MDR-TB) and extensively drug-resistant TB (XDR-TB) in China. Additionally, genetic mutations in 23S rRNA, rplC, and rplD genes were analyzed to reveal potential mechanisms underlying the observed oxazolidinone resistance. A total of 240 M. tuberculosis isolates were included in this study, including 120 MDR-TB isolates and 120 XDR-TB isolates. Overall, linezolid and delpazolid MIC90 values for M. tuberculosis isolates were 0.25 mg/liter and 0.5 mg/liter, respectively. Based on visual inspection, we tentatively set epidemiological cutoff (ECOFF) values for MIC determinations for linezolid and delpazolid at 1.0 mg/liter and 2.0 mg/liter, respectively. Although no significant difference in resistance rates was observed between linezolid and delpazolid among XDR-TB isolates (P > 0.05), statistical analysis revealed a significantly greater proportion of linezolid-resistant isolates than delpazolid-resistant isolates within the MDR-TB group (P = 0.036). Seven (53.85%) of 13 linezolid-resistant isolates were found to harbor mutations within the three target genes. Additionally, 1 isolate exhibited an amino acid substitution (Arg126His) within the protein encoded by rplD that contributed to high-level resistance to linezolid (MIC of >16 mg/liter), compared to a delpazolid MIC of 0.25. In conclusion, in vitro susceptibility testing revealed that delpazolid antibacterial activity was comparable to that of linezolid. A novel mutation within rplD that endowed M. tuberculosis with linezolid, but not delpazolid, resistance was identified.
Collapse
|