51
|
Does Chlorination Promote Antimicrobial Resistance in Waterborne Pathogens? Mechanistic Insight into Co-Resistance and Its Implication for Public Health. Antibiotics (Basel) 2022; 11:antibiotics11050564. [PMID: 35625208 PMCID: PMC9137585 DOI: 10.3390/antibiotics11050564] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 11/16/2022] Open
Abstract
Chemical agents including chlorine and antibiotics are used extensively to control infectious microorganisms. While antibiotics are mainly used to treat bacterial infections, chlorine is widely used for microbial inactivation in the post-secondary disinfection steps of water treatment. The extensive use of these agents has been acknowledged as a driving force for the expansion of antimicrobial resistance (AMR) and has prompted discourse on their roles in the evolution and proliferation of resistant pathogens in the aquatic milieus. We live in a possible “post-antibiotic” era when resistant microbes spread at startling levels with dire predictions relating to a potential lack of effective therapeutic antibacterial drugs. There have been reports of enhancement of resistance among some waterborne pathogens due to chlorination. In this context, it is pertinent to investigate the various factors and mechanisms underlying the emergence and spread of resistance and the possible association between chlorination and AMR. We, therefore, reflect on the specifics of bacterial resistance development, the mechanisms of intrinsic and acquired resistance with emphasis on their environmental and public health implications, the co-selection for antibiotic resistance due to chlorination, biofilm microbiology, and multidrug efflux activity. In-depth knowledge of the molecular basis of resistance development in bacteria will significantly contribute to the more rational utilization of these biocidal agents and aid in filling identified knowledge gap toward curbing resistance expansion.
Collapse
|
52
|
Effects of glucose oxidase and its combination with B. amyloliquefaciens SC06 on intestinal microbiota, immune response and antioxidative capacity in broilers. Animal 2022; 16:100473. [PMID: 35218993 DOI: 10.1016/j.animal.2022.100473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023] Open
Abstract
Glucose oxidase (GOD) is an aerobic dehydrogenase, which catalyses the oxidation of β-D-glucose to gluconic acid and hydrogen peroxide. This study aimed to investigate the effects of dietary glucose oxidase and its combined effects with Bacillus amyloliquefaciens SC06 (BaSC06) on the intestinal microbiota, immune function and antioxidant capacity of broilers. One-day-old male Lingnan yellow-feathered broilers (n = 720) were randomly assigned to four treatment groups: Control group (basal diet), Anti group (basal diet supplemented with 200 mg/kg enramycin), GOD group (basal diet supplemented with 75 U/kg GOD), and combination of GOD and BaSC06 (GB) group (GOD diet (75 U/kg) supplemented with 1 × 108 colony-forming units BaSC06/kg feed), with six replicates per group and 30 birds per replicate. The experiment was conducted over 52 days. The results indicated a significant decrease in α-diversity (Observed species, Chao1, PD_whole_tree and Shannon) with GOD treatment, compared with the control group. GB treatment also significantly decreased the Shannon index of cecal microbiota. GOD treatment significantly decreased the α-diversity, whereas GB treatment significantly increased these indices except for the Chao1 index, compared with the Anti group. Compared with the control group, the relative abundance of Bacteroides in the GOD and GB groups was significantly increased, whereas a decrease in Firmicutes was observed. Compared with the Anti group, GOD treatment significantly increased the relative abundances of Bacteroides and Lactobacillales, while GB treatment significantly increased Lactobacillales and decreased Proteobacteria levels. In addition, GOD treatment significantly decreased interleukin-10 and interferon-γ levels, compared with the control group. In contrast, GB treatment significantly downregulated interferon-γ levels and upregulated secretory immunoglobulin A, transforming growth factor-β and interleukin-2 expression in the jejunal mucosa. GOD treatment significantly decreased transforming growth factor-β and interleukin-10 levels, whereas GB treatment markedly increased interferon-γ expression in the jejunal mucosa compared with the Anti group. Furthermore, GB treatment significantly increased the total antioxidant capability levels and the total superoxide dismutase (T-SOD) and catalase (CAT) activities compared with the control group. Meanwhile, GOD treatment significantly increased glutathione peroxidase (GSH-Px) activity in the jejunal mucosa. Total superoxide dismutase, GSH-Px and CAT activities in the Anti group were higher than in the GOD and GB groups. The malondialdehyde levels in the control group were the highest among all groups. In conclusion, our results indicated that supplementation with GOD alone and its combination with BaSC06 in diet could increase antioxidant capacity, immune function and improve the intestinal microbiota composition of broilers. Combination treatment with GOD with BaSC06 exerted stronger effects than GOD treatment only.
Collapse
|
53
|
Cantón R, Barberán J, Linares M, Molero JM, Rodríguez-González-Moro JM, Salavert M, González Del Castillo J. Decalogue for the selection of oral antibiotics for lower respiratory tract infections. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2022; 35:16-29. [PMID: 35041328 PMCID: PMC8790641 DOI: 10.37201/req/172.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 01/12/2022] [Indexed: 12/28/2022]
Abstract
Lower respiratory tract infections, including chronic obstructive pulmonary disease exacerbations (COPD-E) and community acquired pneumonia (CAP), are one of the most frequent reasons for consultation in primary care and hospital emergency departments, and are the cause of a high prescription of antimicrobial agents. The selection of the most appropriate oral antibiotic treatment is based on different aspects and includes to first consider a bacterial aetiology and not a viral infection, to know the bacterial pathogen that most frequently cause these infections and the frequency of their local antimicrobial resistance. Treatment should also be prescribed quickly and antibiotics should be selected among those with a quicker mode of action, achieving the greatest effect in the shortest time and with the fewest adverse effects (toxicity, interactions, resistance and/or ecological impact). Whenever possible, antimicrobials should be rotated and diversified and switched to the oral route as soon as possible. With these premises, the oral treatment guidelines for mild or moderate COPD-E and CAP in Spain include as first options beta-lactam antibiotics (amoxicillin and amoxicillin-clavulanate and cefditoren), in certain situations associated with a macrolide, and relegating fluoroquinolones as an alternative, except in cases where the presence of Pseudomonas aeruginosa is suspected.
Collapse
Affiliation(s)
- R Cantón
- Rafael Cantón. Servicio de Microbiología. Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS). Madrid. Spain.
| | | | | | | | | | | | - J González Del Castillo
- Juan Gonzalez del Castillo. Servicio de Urgencias, Hospital Clínico San Carlos and Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain.
| |
Collapse
|
54
|
Lai CKC, Ng RWY, Leung SSY, Hui M, Ip M. Overcoming the rising incidence and evolving mechanisms of antibiotic resistance by novel drug delivery approaches - An overview. Adv Drug Deliv Rev 2022; 181:114078. [PMID: 34896131 DOI: 10.1016/j.addr.2021.114078] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/14/2022]
Abstract
Antimicrobial resistance is a normal evolutionary process for microorganisms. Antibiotics exerted accelerated selective pressure that hasten bacterial resistance through mutation, and acquisition external genes. These genes often carry multiple antibiotic resistant determinants allowing the recipient microbe an instant "super-bug" status. The extent of Antimicrobial Resistance (AMR) has reached a level of global crisis, existing antimicrobials are no long effective in treating infections caused by AMR pathogens. The great majority of clinically available antimicrobial agents are administered through oral and intra-venous routes. Overcoming antibacterial resistance by novel drug delivery approach offered new hopes, particularly in the treatment of AMR pathogens in sites less assessible through systemic circulation such as the lung and skin. In the current review, we will revisit the mechanism and incidence of important AMR pathogens. Finally, we will discuss novel drug delivery approaches including novel local antibiotic delivery systems, hybrid antibiotics, and nanoparticle-based antibiotic delivery systems.
Collapse
Affiliation(s)
- Christopher K C Lai
- Department of Microbiology, Prince of Wales Hospital, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong Special Administrative Region.
| | - Rita W Y Ng
- Department of Microbiology, Prince of Wales Hospital, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong Special Administrative Region.
| | - Sharon S Y Leung
- School of Pharmacy, The Chinese University of Hong Kong, New Territories, Hong Kong Special Administrative Region.
| | - Mamie Hui
- Department of Microbiology, Prince of Wales Hospital, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong Special Administrative Region.
| | - Margaret Ip
- Department of Microbiology, Prince of Wales Hospital, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong Special Administrative Region.
| |
Collapse
|
55
|
Urbaniec J, Xu Y, Hu Y, Hingley-Wilson S, McFadden J. Phenotypic heterogeneity in persisters: a novel 'hunker' theory of persistence. FEMS Microbiol Rev 2022; 46:fuab042. [PMID: 34355746 PMCID: PMC8767447 DOI: 10.1093/femsre/fuab042] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/04/2021] [Indexed: 12/11/2022] Open
Abstract
Persistence has been linked to treatment failure since its discovery over 70 years ago and understanding formation, nature and survival of this key antibiotic refractory subpopulation is crucial to enhancing treatment success and combatting the threat of antimicrobial resistance (AMR). The term 'persistence' is often used interchangeably with other terms such as tolerance or dormancy. In this review we focus on 'antibiotic persistence' which we broadly define as a feature of a subpopulation of bacterial cells that possesses the non-heritable character of surviving exposure to one or more antibiotics; and persisters as cells that possess this characteristic. We discuss novel molecular mechanisms involved in persister cell formation, as well as environmental factors which can contribute to increased antibiotic persistence in vivo, highlighting recent developments advanced by single-cell studies. We also aim to provide a comprehensive model of persistence, the 'hunker' theory which is grounded in intrinsic heterogeneity of bacterial populations and a myriad of 'hunkering down' mechanisms which can contribute to antibiotic survival of the persister subpopulation. Finally, we discuss antibiotic persistence as a 'stepping-stone' to AMR and stress the urgent need to develop effective anti-persister treatment regimes to treat this highly clinically relevant bacterial sub-population.
Collapse
Affiliation(s)
- J Urbaniec
- Department of Microbial Sciences and University of Surrey, Guildford, Surrey, GU27XH, UK
| | - Ye Xu
- Department of Microbial Sciences and University of Surrey, Guildford, Surrey, GU27XH, UK
| | - Y Hu
- Farnborough Sensonic limited, Farnborough road, GU14 7NA, UK
| | - S Hingley-Wilson
- Department of Microbial Sciences and University of Surrey, Guildford, Surrey, GU27XH, UK
| | - J McFadden
- Department of Microbial Sciences and University of Surrey, Guildford, Surrey, GU27XH, UK
- Quantum biology doctoral training centre, University of Surrey, Guildford, Surrey, GU27XH, UK
| |
Collapse
|
56
|
Babosan A, Skurnik D, Muggeo A, Pier G, Baharoglu Z, Jové T, Ploy MC, Griveau S, Bedioui F, Vergnolle S, Moussalih S, de Champs C, Mazel D, Guillard T. A qnr-plasmid allows aminoglycosides to induce SOS in Escherichia coli. eLife 2022; 11:69511. [PMID: 35037621 PMCID: PMC8789287 DOI: 10.7554/elife.69511] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 01/12/2022] [Indexed: 11/16/2022] Open
Abstract
The plasmid-mediated quinolone resistance (PMQR) genes have been shown to promote high-level bacterial resistance to fluoroquinolone antibiotics, potentially leading to clinical treatment failures. In Escherichia coli, sub-minimum inhibitory concentrations (sub-MICs) of the widely used fluoroquinolones are known to induce the SOS response. Interestingly, the expression of several PMQR qnr genes is controlled by the SOS master regulator, LexA. During the characterization of a small qnrD-plasmid carried in E. coli, we observed that the aminoglycosides become able to induce the SOS response in this species, thus leading to the elevated transcription of qnrD. Our findings show that the induction of the SOS response is due to nitric oxide (NO) accumulation in the presence of sub-MIC of aminoglycosides. We demonstrated that the NO accumulation is driven by two plasmid genes, ORF3 and ORF4, whose products act at two levels. ORF3 encodes a putative flavin adenine dinucleotide (FAD)-binding oxidoreductase which helps NO synthesis, while ORF4 codes for a putative fumarate and nitrate reductase (FNR)-type transcription factor, related to an O2-responsive regulator of hmp expression, able to repress the Hmp-mediated NO detoxification pathway of E. coli. Thus, this discovery, that other major classes of antibiotics may induce the SOS response could have worthwhile implications for antibiotic stewardship efforts in preventing the emergence of resistance.
Collapse
Affiliation(s)
- Anamaria Babosan
- Inserm UMR-S 1250 P3Cell, SFR CAP-Santé, Université de Reims-Champagne-Ardenne, Reims, France
| | - David Skurnik
- Institut Necker-Enfants Malades, Inserm U1151-Equipe 11, Université Paris Descartes, Paris, France
| | - Anaëlle Muggeo
- Inserm UMR-S 1250 P3Cell, SFR CAP-Santé, Université de Reims-Champagne-Ardenne, Reims, France
| | - Gerald Pier
- Department of Medicine, Brigham and Women's Hospital, Boston, United States
| | - Zeynep Baharoglu
- Unité Plasticité du Génome Bactérien, CNRS UMR3525, Institut Pasteur, Paris, France
| | - Thomas Jové
- CHU Limoges, RESINFIT, UMR 1092, Université de Limoges, Inserm, Limoges, France
| | - Marie-Cécile Ploy
- CHU Limoges, RESINFIT, UMR 1092, Université de Limoges, Inserm, Limoges, France
| | - Sophie Griveau
- Institute of Chemistry for Life and Health Sciences, PSL Research University, CNRS, Paris, France
| | - Fethi Bedioui
- Institute of Chemistry for Life and Health Sciences, PSL Research University, CNRS, Paris, France
| | | | - Sophie Moussalih
- Inserm UMR-S 1250 P3Cell, SFR CAP-Santé, Université de Reims-Champagne-Ardenne, Reims, France
| | - Christophe de Champs
- Inserm UMR-S 1250 P3Cell, SFR CAP-Santé, Université de Reims-Champagne-Ardenne, Reims, France
| | - Didier Mazel
- Unité Plasticité du Génome Bactérien, CNRS UMR3525, Institut Pasteur, Paris, France
| | - Thomas Guillard
- Inserm UMR-S 1250 P3Cell, SFR CAP-Santé, Université de Reims-Champagne-Ardenne, Reims, France
| |
Collapse
|
57
|
Optimizing antimicrobial use: challenges, advances and opportunities. Nat Rev Microbiol 2021; 19:747-758. [PMID: 34158654 DOI: 10.1038/s41579-021-00578-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2021] [Indexed: 02/06/2023]
Abstract
An optimal antimicrobial dose provides enough drug to achieve a clinical response while minimizing toxicity and development of drug resistance. There can be considerable variability in pharmacokinetics, for example, owing to comorbidities or other medications, which affects antimicrobial pharmacodynamics and, thus, treatment success. Although current approaches to antimicrobial dose optimization address fixed variability, better methods to monitor and rapidly adjust antimicrobial dosing are required to understand and react to residual variability that occurs within and between individuals. We review current challenges to the wider implementation of antimicrobial dose optimization and highlight novel solutions, including biosensor-based, real-time therapeutic drug monitoring and computer-controlled, closed-loop control systems. Precision antimicrobial dosing promises to improve patient outcome and is important for antimicrobial stewardship and the prevention of antimicrobial resistance.
Collapse
|
58
|
Udo EE, Boswihi SS, Mathew B, Noronha B, Verghese T. Resurgence of Chloramphenicol Resistance in Methicillin-Resistant Staphylococcus aureus Due to the Acquisition of a Variant Florfenicol Exporter ( fexAv)-Mediated Chloramphenicol Resistance in Kuwait Hospitals. Antibiotics (Basel) 2021; 10:antibiotics10101250. [PMID: 34680830 PMCID: PMC8532628 DOI: 10.3390/antibiotics10101250] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 12/27/2022] Open
Abstract
Following a surge in the prevalence of chloramphenicol-resistant methicillin-resistant Staphylococcus aureus (MRSA) in Kuwait hospitals, this study investigated the genotypes and antibiotic resistance of the chloramphenicol-resistant isolates to ascertain whether they represented new or a resurgence of sporadic endemic clones. Fifty-four chloramphenicol-resistant MRSA isolates obtained in 2014–2015 were investigated. Antibiotic resistance was tested by disk diffusion and MIC determination. Molecular typing was performed using spa typing, multilocus sequence typing, and DNA microarray. Curing and transfer experiments were used to determine the genetic location of resistance determinants. All 54 isolates were resistant to chloramphenicol (MIC: 32–56 mg/L) but susceptible to florfenicol. Two chloramphenicol-resistance determinants, florfenicol exporter (fexA) and chloramphenicol acetyl transferase (cat), were detected. The fexA-positive isolates belonged to CC5-ST627-VI-t688/t450/t954 (n = 45), CC5-ST5-V-t688 (n = 6), whereas the cat-positives isolates were CC8-ST239-III-t037/t860 (n = 3). While cat was carried on 3.5–4.4 kb plasmids, the location of fexA could not be established. DNA sequencing of fexA revealed 100% sequence similarity to a previously reported fexA variant that confers chloramphenicol but not florfenicol resistance. The resurgence of chloramphenicol resistance was due to the introduction and spread of closely related fexA-positive CC5-ST5-V and CC5-ST627-VI clones.
Collapse
|
59
|
Jing H, Chen Y, Liang W, Chen M, Qiu C, Guo MY. Effects of Selenium on MAC-T Cells in Bovine Mastitis: Transcriptome Analysis of Exosomal mRNA Interactions. Biol Trace Elem Res 2021; 199:2904-2912. [PMID: 33098075 DOI: 10.1007/s12011-020-02439-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/14/2020] [Indexed: 12/18/2022]
Abstract
Selenium, a micronutrient, is indispensable for maintaining normal metabolic functions in animals and plants. Selenium has shown promise in terms of its effect on the immune function, ability to control inflammation, and ability to improve bovine mammary gland health. Bovine mastitis remains a major threat to dairy herds globally and has economically significant impacts. The exosomes are a new mode of intercellular communication. Exosomal transfer of mRNAs, microRNAs, and proteins between cells affects the protein production of recipient cells. The development of novel high-throughput omics approaches and bioinformatics tools will help us understand the effects of selenium on immunobiology. However, the differential expression of mRNAs in bovine mammary epithelial cell-derived exosomes has rarely been studied. In the present study, differences in the exosomal transcriptome between control and selenium-treated MAC-T cells were identified by RNA sequencing and transcriptome analysis. The results of mRNA profiling revealed 1978 genes in exosomes that were differentially expressed between the selenium-treated and control cells. We selected and analyzed 91 genes that are involved in inflammation, redox reactions, and immune cell function related to mastitis. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed enrichment pathways involved in selenoproteins and the Ras/PI3K/AKT, MAPK, and FOXO signaling pathways. Our results revealed that selenium may play a crucial role in immune and inflammatory regulation by influencing the differential expression of exosomal mRNAs of key genes in bovine mastitis.
Collapse
Affiliation(s)
- Hongyuan Jing
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yu Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Wan Liang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Miaoyu Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Changwei Qiu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Meng-Yao Guo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
60
|
Testing as Prevention of Resistance in Bacteria Causing Sexually Transmitted Infections-A Population-Based Model for Germany. Antibiotics (Basel) 2021; 10:antibiotics10080929. [PMID: 34438979 PMCID: PMC8388946 DOI: 10.3390/antibiotics10080929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/18/2021] [Accepted: 07/27/2021] [Indexed: 12/30/2022] Open
Abstract
Prescribed antibiotic treatments which do not match the therapeutic requirements of potentially co-existing undetected sexually transmitted infections (STIs) can facilitate the selection of antibiotic-drug-resistant clones. To reduce this risk, this modelling assessed the potential applicability of reliable rapid molecular test assays targeting bacterial STI prior to the prescription of antibiotic drugs. The modelling was based on the prevalence of three bacterial STIs in German heterosexual and men-having-sex-with-men (MSM) populations, as well as on reported test characteristics of respective assays. In the case of the application of rapid molecular STI assays for screening, the numbers needed to test in order to correctly identify any of the included bacterial STIs ranged from 103 to 104 for the heterosexual population and from 5 to 14 for the MSM population. The number needed to harm—defined as getting a false negative result for any of the STIs and a false positive signal for another one, potentially leading to an even more inappropriate adaptation of antibiotic therapy than without any STI screening—was at least 208,995 for the heterosexuals and 16,977 for the MSM. Therefore, the screening approach may indeed be suitable to avoid unnecessary selective pressure on bacterial causes of sexually transmitted infections.
Collapse
|
61
|
Mirzaei M, Furxhi I, Murphy F, Mullins M. A Machine Learning Tool to Predict the Antibacterial Capacity of Nanoparticles. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1774. [PMID: 34361160 PMCID: PMC8308172 DOI: 10.3390/nano11071774] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/13/2021] [Accepted: 07/06/2021] [Indexed: 12/22/2022]
Abstract
The emergence and rapid spread of multidrug-resistant bacteria strains are a public health concern. This emergence is caused by the overuse and misuse of antibiotics leading to the evolution of antibiotic-resistant strains. Nanoparticles (NPs) are objects with all three external dimensions in the nanoscale that varies from 1 to 100 nm. Research on NPs with enhanced antimicrobial activity as alternatives to antibiotics has grown due to the increased incidence of nosocomial and community acquired infections caused by pathogens. Machine learning (ML) tools have been used in the field of nanoinformatics with promising results. As a consequence of evident achievements on a wide range of predictive tasks, ML techniques are attracting significant interest across a variety of stakeholders. In this article, we present an ML tool that successfully predicts the antibacterial capacity of NPs while the model's validation demonstrates encouraging results (R2 = 0.78). The data were compiled after a literature review of 60 articles and consist of key physico-chemical (p-chem) properties and experimental conditions (exposure variables and bacterial clustering) from in vitro studies. Following data homogenization and pre-processing, we trained various regression algorithms and we validated them using diverse performance metrics. Finally, an important attribute evaluation, which ranks the attributes that are most important in predicting the outcome, was performed. The attribute importance revealed that NP core size, the exposure dose, and the species of bacterium are key variables in predicting the antibacterial effect of NPs. This tool assists various stakeholders and scientists in predicting the antibacterial effects of NPs based on their p-chem properties and diverse exposure settings. This concept also aids the safe-by-design paradigm by incorporating functionality tools.
Collapse
Affiliation(s)
- Mahsa Mirzaei
- Department of Accounting and Finance, Kemmy Business School, University of Limerick, V94PH93 Limerick, Ireland; (M.M.); (F.M.); (M.M.)
| | - Irini Furxhi
- Department of Accounting and Finance, Kemmy Business School, University of Limerick, V94PH93 Limerick, Ireland; (M.M.); (F.M.); (M.M.)
- Transgero Limited, Cullinagh, Newcastle West, V42V384 Limerick, Ireland
| | - Finbarr Murphy
- Department of Accounting and Finance, Kemmy Business School, University of Limerick, V94PH93 Limerick, Ireland; (M.M.); (F.M.); (M.M.)
- Transgero Limited, Cullinagh, Newcastle West, V42V384 Limerick, Ireland
| | - Martin Mullins
- Department of Accounting and Finance, Kemmy Business School, University of Limerick, V94PH93 Limerick, Ireland; (M.M.); (F.M.); (M.M.)
| |
Collapse
|
62
|
Zhu F, Pan J, Zou Q, Wu M, Wang H, Xu G. Electron beam irradiation of typical sulfonamide antibiotics in the aquatic environment: Kinetics, removal mechanisms, degradation products and toxicity assessment. CHEMOSPHERE 2021; 274:129713. [PMID: 33545585 DOI: 10.1016/j.chemosphere.2021.129713] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/14/2021] [Accepted: 01/17/2021] [Indexed: 06/12/2023]
Abstract
Due to their widespread use and harmful effects on aquatic environment, sulfonamide antibiotics (SAs) have become an emerging pollutant of great concern around the world. In this study, we investigated the degradation process and mechanism of sulfamerazine (SMR), sulfadiazine (SDZ), and sulfapyridine (SPD) by electron-beam irradiation (EBI). The results showed that the three SAs were well suited to the pseudo-first-order reaction kinetics, and they could be almost completely removed with high efficiency (5 kGy). Among the environmental factors, pH (3.0) and O2 atmosphere can further enhance the removal of the sulfonamides (SAs), while NO2- has the most pronounced degrading inhibitory effects among the many ions, these results illustrate that hydroxyl radicals play a dominant role. Compared with SMR and SDZ, the degree of mineralization of lower molecular weight SPD is obvious (45%). LC-MS and DFT calculations indicate that the concentrations of degradation products of the three SAs show a tendency to increase and then decrease, demonstrating that EBI can achieve efficient removal and further mineralization of SAs. Meanwhile, the results of the common product 4-Aminophenol produced during the degradation process further indicate that HO is the predominant reactive oxygen species (ROS). In addition, acute toxicity experiments with luminescent bacteria and predictions of ECOSAR procedures proved the toxic effects greatly decreased after the degradation. This study provides new ideas for achieving efficient and profound removal of emerging pollutants from the aquatic environment.
Collapse
Affiliation(s)
- Feng Zhu
- School of Environmental and Chemical Engineering, Shanghai University, 99 Shangda Road, Shanghai, 200444, China; Key Laboratory of Organic Compound Pollution Control Engineering, Ministry of Education, Shanghai, 200444, PR China.
| | - Jiali Pan
- School of Environmental and Chemical Engineering, Shanghai University, 99 Shangda Road, Shanghai, 200444, China.
| | - Qi Zou
- School of Environmental and Chemical Engineering, Shanghai University, 99 Shangda Road, Shanghai, 200444, China.
| | - Minghong Wu
- School of Environmental and Chemical Engineering, Shanghai University, 99 Shangda Road, Shanghai, 200444, China; Key Laboratory of Organic Compound Pollution Control Engineering, Ministry of Education, Shanghai, 200444, PR China.
| | - Hongyong Wang
- Key Laboratory of Organic Compound Pollution Control Engineering, Ministry of Education, Shanghai, 200444, PR China; Shanghai Institute of Applied Radiation, Shanghai University, 20 Chengzhong Road, Shanghai, 200444, China.
| | - Gang Xu
- School of Environmental and Chemical Engineering, Shanghai University, 99 Shangda Road, Shanghai, 200444, China; Key Laboratory of Organic Compound Pollution Control Engineering, Ministry of Education, Shanghai, 200444, PR China.
| |
Collapse
|
63
|
Qazi SH, Yousafzai MT, Saddal NS, Dehraj IF, Thobani RS, Akhtar A, Syed JR, Kazi AM, Hotwani A, Rahman N, Mehmood J, Andrews JR, Luby SP, Garrett DO, Qamar FN. Burden of Ileal Perforations Among Surgical Patients Admitted in Tertiary Care Hospitals of Three Asian countries: Surveillance of Enteric Fever in Asia Project (SEAP), September 2016-September 2019. Clin Infect Dis 2021; 71:S232-S238. [PMID: 33258928 PMCID: PMC7705870 DOI: 10.1093/cid/ciaa1309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background Typhoid fever is caused by Salmonella enterica subspecies enterica serovar Typhi (S. Typhi) and can lead to systemic illness and complications. We aimed to characterize typhoid-related ileal perforation in the context of the population-based Surveillance of Enteric Fever in Asia Project (SEAP) in Bangladesh, Nepal and Pakistan. Methods Between September 2016 and September 2019, all cases of nontraumatic ileal perforation with a clinical diagnosis of typhoid were enrolled from 4 tertiary care hospitals in Karachi, 2 pediatric hospitals in Bangladesh, and 2 hospitals in Nepal. Sociodemographic data were collected from patients or their caregivers, and clinical and outcome data were retrieved from medical records. Tissue samples were collected for histopathology and blood cultures where available. Results Of the 249 enrolled cases, 2 from Bangladesh, 5 from Nepal and 242 from Pakistan. In Pakistan, most of the cases were in the 0–15 (117/242; 48%) and 16–30 (89/242; 37%) age groups. In all countries, males were most affected: Pakistan 74.9% (180/242), Nepal 80% (4/5), and Bangladesh 100% (2/2). Blood culture was done on 76 cases; 8 (11%) were positive for S. Typhi, and all were extensively drug resistant (XDR) S. Typhi. Tissue cultures was done on 86 patients; 3 (3%) were positive for S. Typhi, and all were XDR S. Typhi, out of 86 samples tested for histopathology 4 (5%) revealed ileal perforation with necrosis. Culture or histopathology confirmed total 15 (11%) enteric fever cases with ileal perforation are similar to the clinically diagnosed cases. There were 16/242 (7%) deaths from Pakistan. Cases of ileal perforation who survived were more likely to have sought care before visiting the sentinel hospital (P = .009), visited any hospital for treatment (P = .013) compared to those who survived. Conclusions Although surveillance differed substantially by country, one reason for the higher number of ileal perforation cases in Pakistan could be the circulation of XDR strain of S. Typhi in Karachi.
Collapse
Affiliation(s)
- Saqib H Qazi
- Section of Pediatric Surgery, Department of Surgery, Aga Khan University, Karachi, Pakistan
| | - Mohammad T Yousafzai
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Irum F Dehraj
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Rozina S Thobani
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Afshan Akhtar
- Aga Khan University Medical College, Karachi, Pakistan
| | - Jamal R Syed
- National Institute of Child Health, Karachi, Pakistan
| | - Abdul M Kazi
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Aneeta Hotwani
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Najeeb Rahman
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Junaid Mehmood
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Jason R Andrews
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, California, USA
| | - Stephen P Luby
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, California, USA
| | - Denise O Garrett
- Applied Epidemiology, Sabin Vaccine Institute, Washington, DC, USA
| | - Farah N Qamar
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
64
|
Cui ZH, He HL, Wu SB, Dong CL, Lu SY, Shan TJ, Fang LX, Liao XP, Liu YH, Sun J. Rapid Screening of Essential Oils as Substances Which Enhance Antibiotic Activity Using a Modified Well Diffusion Method. Antibiotics (Basel) 2021; 10:antibiotics10040463. [PMID: 33923861 PMCID: PMC8072922 DOI: 10.3390/antibiotics10040463] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 11/30/2022] Open
Abstract
Antimicrobial resistance is recognized as one of the major global health challenges of the 21st century. Synergistic combinations for antimicrobial therapies can be a good strategy for the treatment of multidrug resistant infections. We examined the ability of a group of 29 plant essential oils as substances which enhance the antibiotic activity. We used a modified well diffusion method to establish a high-throughput screening method for easy and rapid identification of high-level enhancement combinations against bacteria. We found that 25 essential oils possessed antibacterial activity against Escherichia Coli ATCC 25922 and methicillin-resistant Staphylococcus aureus (MRSA) 43300 with MICs that ranged from 0.01% to 2.5% v/v. We examined 319 (11 × 29) combinations in a checkerboard assay with E. Coli ATCC 25922 and MRSA 43300, and the result showed that high-level enhancement combinations were 48 and 44, low-level enhancement combinations were 214 and 211, and no effects combinations were 57 and 64, respectively. For further verification we randomly chose six combinations that included orange and Petitgrain essential oils in a standard time-killing assay. The results are in great agreement with those of the well diffusion assays. Therefore, the modified diffusion method was a rapid and effective method to screen high-level enhancement combinations of antibiotics and essential oils.
Collapse
Affiliation(s)
- Ze-Hua Cui
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou 510642, China; (Z.-H.C.); (H.-L.H.); (S.-B.W.); (S.-Y.L.); (L.-X.F.); (X.-P.L.); (Y.-H.L.)
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Hui-Ling He
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou 510642, China; (Z.-H.C.); (H.-L.H.); (S.-B.W.); (S.-Y.L.); (L.-X.F.); (X.-P.L.); (Y.-H.L.)
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Shuai-Bin Wu
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou 510642, China; (Z.-H.C.); (H.-L.H.); (S.-B.W.); (S.-Y.L.); (L.-X.F.); (X.-P.L.); (Y.-H.L.)
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Chun-Liu Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China;
| | - Si-Ya Lu
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou 510642, China; (Z.-H.C.); (H.-L.H.); (S.-B.W.); (S.-Y.L.); (L.-X.F.); (X.-P.L.); (Y.-H.L.)
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Ti-Jiang Shan
- College of Forestry and Landscape Architecture, South China Agricultural University, Guangzhou 510642, China;
| | - Liang-Xing Fang
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou 510642, China; (Z.-H.C.); (H.-L.H.); (S.-B.W.); (S.-Y.L.); (L.-X.F.); (X.-P.L.); (Y.-H.L.)
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Xiao-Ping Liao
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou 510642, China; (Z.-H.C.); (H.-L.H.); (S.-B.W.); (S.-Y.L.); (L.-X.F.); (X.-P.L.); (Y.-H.L.)
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Ya-Hong Liu
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou 510642, China; (Z.-H.C.); (H.-L.H.); (S.-B.W.); (S.-Y.L.); (L.-X.F.); (X.-P.L.); (Y.-H.L.)
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Jian Sun
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou 510642, China; (Z.-H.C.); (H.-L.H.); (S.-B.W.); (S.-Y.L.); (L.-X.F.); (X.-P.L.); (Y.-H.L.)
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Correspondence:
| |
Collapse
|
65
|
Targeting Superoxide dismutase confers enhanced Reactive Oxygen Species mediated eradication of Polymyxin B induced Acinetobacter baumannii persisters. Antimicrob Agents Chemother 2021; 95:AAC.02180-20. [PMID: 33593839 PMCID: PMC8092903 DOI: 10.1128/aac.02180-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Bacterial persisters represent non-inheritable drug tolerant population that are linked to recalcitrance of infections in healthcare settings. The rise of antibiotic resistance and depletion of new antibiotics in drug discovery pipeline has made the task of persister eradication more daunting. In the present study, we report that treatment of Acinetobacter baumannii with the last resort antibiotic polymyxin B displays continuous variation in tolerance among different clinical isolates. Mechanistically, Polymyxin B persisters exhibit disruption of proton motive force led delocalisation of cell division protein to attain a growth arrested phenotype. Tolerance studies on mutant strains revealed that superoxide dismutase (sodB) activity is a major contributor in tolerance of A. baumannii to polymyxin B. Using a dual fluorescence based persister detection system, screening of various antibiotics was performed to eradicate polymyxin B induced persisters of A. baumannii Rifampicin exhibited eradication of polymyxin B tolerant population by log reduction of 6 in magnitude against different clinical isolates of A. baumannii We establish that enhanced generation of ROS by rifampicin leads to clearance of these polymyxin B persisters. It was further demonstrated, as a proof of concept, that rifampicin potentiates the killing of polymyxin B persisters in murine wound infection model. We found that the effects were linked to significant down regulation of sodB by rifampicin, which contributes to higher generation of ROS in polymyxin B tolerant cells. In view of these results, we propose that the combination of polymyxin B and rifampicin is an effective antipersister strategy in clearing polymyxin B induced A. baumannii persisters.
Collapse
|
66
|
Wasa A, Land JG, Gorthy R, Krumdieck S, Bishop C, Godsoe W, Heinemann JA. Antimicrobial and biofilm-disrupting nanostructured TiO2 coating demonstrating photoactivity and dark activity. FEMS Microbiol Lett 2021; 368:6232155. [PMID: 33864459 DOI: 10.1093/femsle/fnab039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/13/2021] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial materials are tools used to reduce the transmission of infectious microorganisms. Photo-illuminated titania (TiO2) is a known antimicrobial material. Used as a coating on door handles and similar surfaces, it may reduce viability and colonization by pathogens and limit their spread. We tested the survival of Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus and Saccharomyces cerevisiae on a nano-structured TiO2-based thin film, called 'NsARC', and on stainless steel under a variety of light wavelengths and intensities. There was significantly less survival (P <0.001) of all the organisms tested on NsARC compared to inert uncoated stainless steel under all conditions. NsARC was active in the dark and possible mechanisms for this are suggested. NsARC inhibited biofilm formation as confirmed by scanning electron microscopy. These results suggest that NsARC can be used as a self-cleaning and self-sterilizing antimicrobial surface coating for the prevention and reduction in the spread of potentially infectious microbes.
Collapse
Affiliation(s)
- Alibe Wasa
- School of Biological Sciences, University of Canterbury, New Zealand
| | - Johann G Land
- Department of Mechanical Engineering, University of Canterbury, New Zealand
| | - Rukmini Gorthy
- Department of Mechanical Engineering, University of Canterbury, New Zealand
| | - Susan Krumdieck
- Department of Mechanical Engineering, University of Canterbury, New Zealand
| | - Catherine Bishop
- Department of Mechanical Engineering, University of Canterbury, New Zealand
| | - William Godsoe
- Bio-Protection Centre, Lincoln University, 85084 Ellesmere Junction Road, Lincoln 7647, New Zealand
| | - Jack A Heinemann
- School of Biological Sciences, University of Canterbury, New Zealand
| |
Collapse
|
67
|
Ara B, Urmi UL, Haque TA, Nahar S, Rumnaz A, Ali T, Alam MS, Mosaddek ASM, Rahman NAA, Haque M, Islam S. Detection of mobile colistin-resistance gene variants ( mcr-1 and mcr-2) in urinary tract pathogens in Bangladesh: the last resort of infectious disease management colistin efficacy is under threat. Expert Rev Clin Pharmacol 2021; 14:513-522. [PMID: 33691556 DOI: 10.1080/17512433.2021.1901577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background: Currently, colistin-resistant pathogens emerged has become a global health concern. This study assessed the distribution of mcr-1 to mcr-5 variants with the phenotypic colistin-resistance in bacterial isolates from urinary tract infection (UTI) patients in Bangladesh.Methods: A cross-sectional study was conducted between April 2017 and March 2018 to enroll uncomplicated UTI patients, and 142 urine samples were analyzed. Uropathogens were identified using the API-20E biochemical panel and 16s rRNA gene sequencing. Polymerase chain reactions detected the mcr gene variants in the UTI isolates. The phenotypic colistin-susceptibility was determined by the Kirby-Bauer disc-diffusion method and the minimal inhibitory concentration (MIC) measurement.Results: The combined carriage of mcr-1 and mcr-2 genes in 11.4% (14/123) of urinary tract pathogens. The mcr-positive pathogens include five Escherichia coli, three Klebsiella pneumoniae, three Pseudomonas putida, two Enterobacter cloacae, and one Enterobacter hormaechei. The mcr-positive variant showed significantly higher phenotypic colistin resistance with MIC between >16 µg/mL and >128 µg/mL (p< 0.001). Over 85% of colistin-resistant isolates showed MDR phenomena.Conclusions: The emergence of the clinical MDR pathogens with resistance to a highly selective drug may lead to a lack of treatment options for the infectious diseases and spread of infection to the unaffected cohorts.
Collapse
Affiliation(s)
- Bayasrin Ara
- Department of Microbiology, Jahangirnagar University, Dhaka, Bangladesh
| | - Umme Laila Urmi
- Department of Microbiology, Jahangirnagar University, Dhaka, Bangladesh
| | - Tanjum Ara Haque
- Department of Microbiology, Jahangirnagar University, Dhaka, Bangladesh
| | - Shamsun Nahar
- Department of Microbiology, Jahangirnagar University, Dhaka, Bangladesh
| | - Adity Rumnaz
- Department of Microbiology, Jahangirnagar University, Dhaka, Bangladesh
| | - Tamanna Ali
- Department of Microbiology, Jahangirnagar University, Dhaka, Bangladesh
| | | | | | - Nor Azlina A Rahman
- Department of Physical Rehabilitation Sciences, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan, Malaysia
| | - Mainul Haque
- Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kuala Lumpur, Malaysia
| | - Salequl Islam
- Department of Microbiology, Jahangirnagar University, Dhaka, Bangladesh
| |
Collapse
|
68
|
Cantón R. [Current microbiological aspects of community respiratory infection beyond COVID-19]. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2021; 34:81-92. [PMID: 33749214 PMCID: PMC8019468 DOI: 10.37201/req/049.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 03/21/2021] [Indexed: 12/22/2022]
Abstract
From a microbiological point of view, both empirical and targeted antimicrobial treatment in respiratory infection is based on the sensitivity profile of isolated microorganisms and the possible resistance mechanisms that they may present. The latter may vary in different geographic areas according to prescription profiles and vaccination programs. Beta-lactam antibiotics, fluoroquinolones, and macrolides are the most commonly used antimicrobials during the exacerbations of chronic obstructive pulmonary disease and community-acquired pneumonia. In their prescription, different aspects such as intrinsic activity, bactericidal effect or their ability to prevent the development of resistance must be taken into account. The latter is related to the PK/PD parameters, the mutant prevention concentration and the so-called selection window. More recently, the potential ecological impact has grown in importance, not only on the intestinal microbiota, but also on the respiratory one. Maintaining the state of eubiosis requires the use of antimicrobials with a low profile of action on anaerobic bacteria. With their use, the resilience of the bacterial populations belonging to the microbiota, the state of resistance of colonization and the collateral damage related to the emergence of resistance to the antimicrobials in pathogens causing the infections and in the bacterial populations integrating the microbiota.
Collapse
Affiliation(s)
- R Cantón
- Rafael Cantón. Servicio de Microbiología. Hospital Universitario Ramón y Cajal e Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS). Madrid. Spain.
| |
Collapse
|
69
|
Selection of Resistant Bacteria in Mallards Exposed to Subinhibitory Concentrations of Ciprofloxacin in Their Water Environment. Antimicrob Agents Chemother 2021; 65:AAC.01858-20. [PMID: 33318021 DOI: 10.1128/aac.01858-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/10/2020] [Indexed: 12/22/2022] Open
Abstract
Emergence and selection of antibiotic resistance following exposure to high antibiotic concentrations have been repeatedly shown in clinical and agricultural settings, whereas the role of the weak selective pressures exerted by antibiotic levels below the MIC (sub-MIC) in aquatic environments due to anthropogenic contamination remains unclear. Here, we studied how exposure to sub-MIC levels of ciprofloxacin enriches for Escherichia coli with reduced susceptibility to ciprofloxacin using a mallard colonization model. Mallards were inoculated with two isogenic extended-spectrum-β-lactamase (ESBL)-encoding E. coli strains, differing only by a gyrA mutation that results in increased MICs of ciprofloxacin, and exposed to different levels of ciprofloxacin in their swimming water. Changes in the ratios of mutant to parental strains excreted in feces over time and ESBL plasmid spread within the gut microbiota from individual birds were investigated. Results show that in vivo selection of gyrA mutants occurred in mallards during exposure to ciprofloxacin at concentrations previously found in aquatic environments. During colonization, resistance plasmids were readily transferred between strains in the intestines of the mallards, but conjugation frequencies were not affected by ciprofloxacin exposure. Our results highlight the potential for enrichment of resistant bacteria in wildlife and underline the importance of reducing antibiotic pollution in the environment.
Collapse
|
70
|
Jiang L, Xie N, Chen M, Liu Y, Wang S, Mao J, Li J, Huang X. Synergistic Combination of Linezolid and Fosfomycin Closing Each Other's Mutant Selection Window to Prevent Enterococcal Resistance. Front Microbiol 2021; 11:605962. [PMID: 33633692 PMCID: PMC7899970 DOI: 10.3389/fmicb.2020.605962] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/30/2020] [Indexed: 12/21/2022] Open
Abstract
Enterococci, the main pathogens associated with nosocomial infections, are resistant to many common antibacterial drugs including β-lactams, aminoglycosides, etc. Combination therapy is considered an effective way to prevent bacterial resistance. Preliminary studies in our group have shown that linezolid combined with fosfomycin has synergistic or additive antibacterial activity against enterococci, while the ability of the combination to prevent resistance remains unknown. In this study, we determined mutant prevention concentration (MPC) and mutant selection window (MSW) of linezolid, fosfomycin alone and in combination including different proportions for five clinical isolates of Enterococcus and characterized the resistance mechanism for resistant mutants. The results indicated that different proportions of linezolid combined with fosfomycin had presented different MPCs and MSWs. Compared with linezolid or fosfomycin alone, the combination can restrict the enrichment of resistant mutants at a lower concentration. A rough positive correlation between the selection index (SI) of the two agents in combination and the fractional inhibitory concentration index (FICI) of the combination displayed that the smaller FICI of linezolid and fosfomycin, the more probable their MSWs were to close each other. Mutations in ribosomal proteins (L3 and L4) were the mechanisms for linezolid resistant mutants. Among the fosfomycin-resistant mutants, only two strains have detected the MurA gene mutation related to fosfomycin resistance. In conclusion, the synergistic combination of linezolid and fosfomycin closing each other’s MSW could effectively suppress the selection of enterococcus resistant mutants, suggesting that the combination may be an alternative for preventing enterococcal resistance. In this study, the resistance mechanism of fosfomycin remains to be further studied.
Collapse
Affiliation(s)
- Lifang Jiang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Na Xie
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Mingtao Chen
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Yanyan Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shuaishuai Wang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Jun Mao
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Jiabin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaohui Huang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| |
Collapse
|
71
|
The Minimum Inhibitory Concentration of Antibiotics: Methods, Interpretation, Clinical Relevance. Pathogens 2021; 10:pathogens10020165. [PMID: 33557078 PMCID: PMC7913839 DOI: 10.3390/pathogens10020165] [Citation(s) in RCA: 346] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/21/2021] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
Inefficiency of medical therapies used in order to cure patients with bacterial infections requires not only to actively look for new therapeutic strategies but also to carefully select antibiotics based on variety of parameters, including microbiological. Minimal inhibitory concentration (MIC) defines in vitro levels of susceptibility or resistance of specific bacterial strains to applied antibiotic. Reliable assessment of MIC has a significant impact on the choice of a therapeutic strategy, which affects efficiency of an infection therapy. In order to obtain credible MIC, many elements must be considered, such as proper method choice, adherence to labeling rules, and competent interpretation of the results. In this paper, two methods have been discussed: dilution and gradient used for MIC estimation. Factors which affect MIC results along with the interpretation guidelines have been described. Furthermore, opportunities to utilize MIC in clinical practice, with pharmacokinetic /pharmacodynamic parameters taken into consideration, have been investigated. Due to problems related to PK determination in individual patients, statistical estimation of the possibility of achievement of the PK/PD index, based on the Monte Carlo, was discussed. In order to provide comprehensive insights, the possible limitations of MIC, which scientists are aware of, have been outlined.
Collapse
|
72
|
Pharmacokinetic and Pharmacodynamic Optimization of Antibiotic Therapy in Cystic Fibrosis Patients: Current Evidences, Gaps in Knowledge and Future Directions. Clin Pharmacokinet 2021; 60:409-445. [PMID: 33486720 DOI: 10.1007/s40262-020-00981-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 10/22/2022]
Abstract
Antibiotic therapy is one of the main treatments for cystic fibrosis (CF). It aims to eradicate bacteria during early infection, calms down the inflammatory process, and leads to symptom resolution of pulmonary exacerbations. CF can modify both the pharmacokinetic (PK) and pharmacodynamic (PD) profiles of antibiotics, therefore specific PK/PD endpoints should be determined in the context of CF. Currently available data suggest that optimal PK/PD targets cannot be attained in sputum with intravenous aminoglycosides. Continuous infusion appears preferable for β-lactam antibiotics, but optimal concentrations in sputum are unlikely to be reached, with some possible exceptions such as meropenem and ceftolozane. Usual doses are likely suboptimal for fluoroquinolones and linezolid, whereas daily doses of 45-60 mg/kg and 200 mg could be convenient for vancomycin and doxycycline, respectively. Weekly azithromycin doses of 22-30 mg/kg could also be appropriate for its anti-inflammatory effect. The difficulty with achieving optimal concentrations supports the use of combined treatments and the inhaled administration route, as very high local concentrations, concomitantly with low systemic exposure, can be obtained with the inhaled route for aminoglycosides, colistin, and fluoroquinolones, thus minimizing the risk of toxicity.
Collapse
|
73
|
Effects of Antibiotic Treatment with Piperacillin/Tazobactam versus Ceftriaxone on the Composition of the Murine Gut Microbiota. Antimicrob Agents Chemother 2021; 65:AAC.01504-20. [PMID: 33168609 DOI: 10.1128/aac.01504-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 11/04/2020] [Indexed: 12/11/2022] Open
Abstract
Effective antimicrobial stewardship requires a better understanding of the impact of different antibiotics on the gut microflora. Studies with humans are confounded by large interindividual variability and difficulty in identifying control cohorts. However, controlled murine models can provide valuable information. In this study, we examined the impact of a penicillin-like antibiotic (piperacillin-tazobactam [TZP]) or a third-generation cephalosporin (ceftriaxone [CRO]) on the murine gut microbiota by analysis of changes in fecal microbiome composition by 16S rRNA amplicon sequencing and standard microbiology. Resistance to colonization by multidrug-resistant Escherichia coli sequence type 131 (ST131) and Klebsiella pneumoniae ST258 was also tested. Changes in microbiome composition and a significant (P < 0.05) decrease in diversity occurred in all treated mice, but dysbiosis was more marked and prolonged after CRO exposure, with a persistent rise in Proteobacteria Enterobacteriaceae blooms occurred in all antibiotic-treated mice, but for TZP, unlike CRO, these were significant only under direct antibiotic pressure. At the height of dysbiosis after antibiotic termination, the murine gut was highly susceptible to colonization with both multidrug-resistant enterobacterial pathogens. Cohabitation of treated mice with untreated individuals had a notable mitigating effect on dysbiosis of treated guts. The administration of a third-generation cephalosporin caused a more severe imbalance in the murine fecal microflora than that caused by a penicillin/β-lactam inhibitor combination with comparable activity against medically important virulent bacteria. At the height of dysbiosis, both antibiotic treatments equally led to microbial instability associated with loss of resistance to gut colonization by antibiotic-resistant pathogens.
Collapse
|
74
|
Tuan LC, Thanh LTH, Duc Huy N, Thuy Trang DT, Nhat Le BN, Nhat Linh NL, Khanh NV, Nu Bao Tien HT, Hoang Loc N. Antagonistic Activity Against Pathogenic Vibrio Isolates of Bioflocculant-Producing Bacteria Isolated from Shrimp Ponds. Pak J Biol Sci 2021; 24:1322-1332. [PMID: 34989209 DOI: 10.3923/pjbs.2021.1322.1332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
<b>Background and Objectives:</b> Biofloc culture system has been used in aquaculture as an effective technology for water treatment due to many advantages of being biodegradable and environmentally friendly. This study aims to isolate bioflocculant-producing bacteria antagonistic to pathogenic <i>Vibrio</i> species from Pacific white shrimp ponds in Thua Thien Hue, Vietnam. <b>Materials and Methods:</b> <i>Vibrio</i> isolates were isolated by screening on medium with and without antibiotics. The resistance of <i>Vibrio</i> to antimicrobial agents was assessed by Minimum Inhibitory Concentration (MIC). Bioflocs formed in shrimp cultures were used to screen bioflocculant-producing bacteria. The identification of bacteria was performed by 16S rRNA sequencing. The flocculating activity was measured by a test with kaolin clay suspension. To evaluate the antagonistic activity against <i>Vibrio</i> isolates, an agar well diffusion assay was used. <b>Results:</b> The screening results have found that <i>Vibrio</i> isolates such as <i>V. parahaemolyticus</i> KS02 and <i>V. alginolyticus</i> KS08 from shrimp ponds can be resistant to many antibiotics with the highest resistance rate up to 66.49%. Four bioflocculant-producing isolates were obtained and identified as <i>Bacillus</i> species. Among them, <i>Bacillus velezensis </i>B9 when grown in YPG medium supplemented with 3% sucrose and 0.7% peptone had the highest bioflocculation with an activity of 49.2%. Two isolates of <i>B. subtilis</i> B2 and <i>Bacillus</i> sp. B6 had quite strong antagonistic activities against vibriosis shown in the zones of inhibition on the assay plates with diameters of about 20 mm. <b>Conclusion:</b> The present study has found some <i>Bacillus</i> isolates had bioflocculant-producing efficiency and inhibited pathogenic <i>Vibrio</i> bacteria. These <i>Bacillus</i> isolates will potentially be used as inoculum for bioflocculation to improve shrimp production.
Collapse
|
75
|
Seifi T, Kamali AR. Anti-pathogenic activity of graphene nanomaterials: A review. Colloids Surf B Biointerfaces 2020; 199:111509. [PMID: 33340933 DOI: 10.1016/j.colsurfb.2020.111509] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/26/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022]
Abstract
Graphene and its derivatives are promising candidates for a variety of biological applications, among which, their anti-pathogenic properties are highly attractive due to the outstanding physicochemical characteristics of these novel nanomaterials. The antibacterial, antiviral and antifungal performances of graphene are increasingly becoming more important due to the pathogen's resistance to existing drugs. Despite this, the factors influencing the antibacterial activity of graphene nanomaterials, and consequently, the mechanisms involved are still controversial. This review aims to systematically summarize the literature, discussing various factors that affect the antibacterial performance of graphene materials, including the shape, size, functional group and the electrical conductivity of graphene flakes, as well as the concentration, contact time and the pH value of the graphene suspensions used in related microbial tests. We discuss the possible surface and edge interactions between bacterial cells and graphene nanomaterials, which cause antibacterial effects such as membrane/oxidative/photothermal stresses, charge transfer, entrapment and self-killing phenomena. This article reviews the anti-pathogenic activity of graphene nanomaterials, comprising their antibacterial, antiviral, antifungal and biofilm-forming performance, with an emphasis on the antibacterial mechanisms involved.
Collapse
Affiliation(s)
- Tahereh Seifi
- Energy and Environmental Materials Research Centre (E(2)MC), School of Metallurgy, Northeastern University, Shenyang, 110819, China
| | - Ali Reza Kamali
- Energy and Environmental Materials Research Centre (E(2)MC), School of Metallurgy, Northeastern University, Shenyang, 110819, China.
| |
Collapse
|
76
|
Islam S, Urmi UL, Rana M, Sultana F, Jahan N, Hossain B, Iqbal S, Hossain MM, Mosaddek ASM, Nahar S. High abundance of the colistin resistance gene mcr-1 in chicken gut-bacteria in Bangladesh. Sci Rep 2020; 10:17292. [PMID: 33057111 PMCID: PMC7560609 DOI: 10.1038/s41598-020-74402-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 09/26/2020] [Indexed: 12/19/2022] Open
Abstract
Colistin is considered a last-resort reserved drug for the treatment of critical human infections by Gram-negative bacteria. Phenotypic colistin-resistance is strongly associated with plasmid-mediated mobile colistin resistance (mcr) genes. The mcr-bearing Enterobacteriaceae have been detected in many countries from environments, animals, and humans. This study investigated phenotypic colistin-resistance and the distribution of mcr-1, mcr-2, mcr-3, mcr-4, and mcr-5 genes in chicken-gut bacteria in Bangladesh. Bacteria were isolated from poultry- and native-chicken droppings, and their susceptibilities to colistin were determined by agar dilution and E-test minimal inhibitory concentration (MIC) measurements. Multiplex polymerase chain reactions detected mcr-1 to mcr-5 genes. Overall, 61.7% (92/149) of the isolates showed colistin resistance by agar dilution assessment (MIC > 2.0 μg/mL). The phenotypic resistance was observed considerably higher in poultry-chicken isolates (64.6%, 64/99) than in native-chicken isolates (56%, 28/50; p = 0.373). All the resistant isolates showed MIC levels between > 2 and > 128 μg/mL. The mcr-genes (mcr-1and mcr-2 combined) were detected more in poultry gut bacteria (36.4%) than native-chicken isolates (20%, p = 0.06). Despite bacteria sources, mcr-genes appeared to be significantly associated with phenotypic colistin-resistance phenomena (p < 0.001). Prior colistin usage led to a substantial increase in the proportion of bacteria with mcr-genes and phenotypic resistance (p < 0.001).
Collapse
Affiliation(s)
- Salequl Islam
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh.
| | - Umme Laila Urmi
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Masud Rana
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Fahmida Sultana
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Nusrat Jahan
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Billal Hossain
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Samiul Iqbal
- Department of Oral Maxillofacial Surgery, Faculty of Dentistry, BSMMU, Dhaka, 1210, Bangladesh
| | - Md Moyazzem Hossain
- Department of Statistics, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Abu Syed Md Mosaddek
- Department of Pharmacology, Uttara Adhunik Medical College, Uttara, Dhaka, 1230, Bangladesh
| | - Shamsun Nahar
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh.
| |
Collapse
|
77
|
Swain SS, Paidesetty SK, Padhy RN. Phytochemical conjugation as a potential semisynthetic approach toward reactive and reuse of obsolete sulfonamides against pathogenic bacteria. Drug Dev Res 2020; 82:149-166. [PMID: 33025605 DOI: 10.1002/ddr.21746] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 09/16/2020] [Accepted: 09/20/2020] [Indexed: 12/20/2022]
Abstract
The emergence and reemergence of multidrug-resistant (MDR) bacteria and mycobacteria in community and hospital periphery have directly enhanced the hospitalization costs, morbidity and mortality, globally. The appearance of MDR pathogens, the currently used antibiotics, remains insufficient, and the development of potent antibacterial(s) is merely slow. Thus, the development of active antibacterials is the call of the day. The sulfonamides class of antibacterials was the most successful synthesized drug in the 19th century. Mechanically, sulfonamides were targeting bacterial folic acid biosynthesis and today, those are obsolete or clinically inactive. Nevertheless, the magic sulfonamide pharmacophore has been used continuously in several mainstream antibacterial, antidiabetic, antiviral drugs. Concomitantly, thousands of phytochemicals with antimicrobial potencies have been recorded and were commanded as alternate antibacterials toward control of MDR pathogens. However, none/very few isolated phytochemicals have gone up to the pure-drug stage due to the lack of the desired drug-likeness values and the required pharmacokinetic properties. Thus, chemical modification of parent drug remains as the versatile approach in antibacterial drug development. Improvement of clinically inactive sulfa drugs with suitable phytochemicals to develop active, low-toxic drug molecules followed by medicinal chemistry could be prudent. This review highlights such "sulfonamide-phytochemical" hybrid drug development research works for utilizing inactive sulfonamides and phytochemicals; the ingenious cost-effective and resource-saving hybrid drug concept could be a new trend in current antibacterial drug discovery to reactive the obsolete antibacterials.
Collapse
Affiliation(s)
- Shasank S Swain
- Central Research Laboratory, Institute of Medical Sciences and Sum Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | - Sudhir K Paidesetty
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | - Rabindra N Padhy
- Central Research Laboratory, Institute of Medical Sciences and Sum Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| |
Collapse
|
78
|
Li X, Liu C, Mao Z, Li Q, Qi S, Zhou F. Short-course versus long-course antibiotic treatment in patients with uncomplicated gram-negative bacteremia: A systematic review and meta-analysis. J Clin Pharm Ther 2020; 46:173-180. [PMID: 32981149 PMCID: PMC7820952 DOI: 10.1111/jcpt.13277] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/27/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022]
Abstract
What is known and objective Gram‐negative bacteremia remains a major health problem around the world. The optimal duration of antibiotic treatment has been poorly defined, and there are significant differences of opinion between clinicians. We conducted this systematic review and meta‐analysis to compare the clinical outcomes of short‐course and long‐course treatments in patients with uncomplicated gram‐negative bacteremia. Methods We searched public databases (PubMed, EMBASE and Cochrane Library) to identify eligible studies. The primary outcomes were all‐cause mortality and the incidence of recurrent bacteremia through day 30. We used the Cochrane risk of bias assessment tool to evaluate the risk of bias for randomized controlled trials (RCTs) and the Newcastle‐Ottawa Scale for non‐RCTs. Results and discussion Six studies involving 2689 patients were included in the systematic review and meta‐analysis. No significant difference was found between short‐course and long‐course antibiotic treatments in 30‐day mortality (risk ratio [RR] 0.85; 95% confidence interval [CI] 0.65‐1.13; P = .26), 30‐day recurrent bacteremia (RR 1.07; 95% CI 0.68‐1.67; P = .78), 90‐day mortality (RR 0.84; 95% CI 0.57‐1.24; P = .38), 90‐day recurrent bacteremia (RR 0.98; 95% CI 0.50‐1.89; P = .94), adverse events (RR 1.14; 95% CI 0.89‐1.45; P = .30), Clostridium difficile infection (RR 0.86; 95% CI 0.40‐1.86; P = .71) or resistance development (RR 1.19; 95% CI 0.66‐2.14; P = .57). What is new and conclusion Short‐course was non‐inferior to long‐course antibiotic treatments for patients with uncomplicated gram‐negative bacteremia. Considering the drug‐related side effects and cost‐effectiveness, a shorter duration of antibiotic treatment may be preferable for this particular population. However, additional high‐quality RCTs are needed to further assess whether a shorter course of antibiotic treatment is of greater benefit for patients with uncomplicated gram‐negative bacteremia.
Collapse
Affiliation(s)
- Xiaoming Li
- Medical School of Chinese PLA, Beijing, China.,Department of Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Chao Liu
- Medical School of Chinese PLA, Beijing, China
| | - Zhi Mao
- Department of Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Qinglin Li
- Department of Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Shuang Qi
- Medical School of Chinese PLA, Beijing, China.,Department of Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Feihu Zhou
- Department of Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
79
|
Sun G, Hu C, Mei Q, Luo M, Chen X, Li Z, Liu Y, Deng Z, Zhang Z, Sun Y. Uncovering the cytochrome P450-catalyzed methylenedioxy bridge formation in streptovaricins biosynthesis. Nat Commun 2020; 11:4501. [PMID: 32908132 PMCID: PMC7481197 DOI: 10.1038/s41467-020-18336-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Streptovaricin C is a naphthalenic ansamycin antibiotic structurally similar to rifamycins with potential anti-MRSA bioactivities. However, the formation mechanism of the most fascinating and bioactivity-related methylenedioxy bridge (MDB) moiety in streptovaricins is unclear. Based on genetic and biochemical evidences, we herein clarify that the P450 enzyme StvP2 catalyzes the MDB formation in streptovaricins, with an atypical substrate inhibition kinetics. Furthermore, X-ray crystal structures in complex with substrate and structure-based mutagenesis reveal the intrinsic details of the enzymatic reaction. The mechanism of MDB formation is proposed to be an intramolecular nucleophilic substitution resulting from the hydroxylation by the heme core and the keto-enol tautomerization via a crucial catalytic triad (Asp89-His92-Arg72) in StvP2. In addition, in vitro reconstitution uncovers that C6-O-methylation and C4-O-acetylation of streptovaricins are necessary prerequisites for the MDB formation. This work provides insight for the MDB formation and adds evidence in support of the functional versatility of P450 enzymes. Streptovaricin C is an antibiotic containing a methylenedioxy bridge (MDB) moiety essential for its activity. Here, the authors show that a P450 monooxygenase StvP2 catalyses MDB formation, report its crystal structure in complex with the substrate, and elucidate mechanistic details of MDB formation.
Collapse
Affiliation(s)
- Guo Sun
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 430071, Wuhan, P. R. China
| | - Chaoqun Hu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 430071, Wuhan, P. R. China
| | - Qing Mei
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 430071, Wuhan, P. R. China
| | - Minghe Luo
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 430071, Wuhan, P. R. China
| | - Xu Chen
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 430071, Wuhan, P. R. China
| | - Zhengyuan Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 430071, Wuhan, P. R. China
| | - Yuanzhen Liu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 430071, Wuhan, P. R. China
| | - Zixin Deng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 430071, Wuhan, P. R. China
| | - Zhengyu Zhang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 430071, Wuhan, P. R. China.
| | - Yuhui Sun
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, 430071, Wuhan, P. R. China.
| |
Collapse
|
80
|
Al Salah DMM, Ngweme GN, Laffite A, Otamonga JP, Mulaji C, Poté J. Hospital wastewaters: A reservoir and source of clinically relevant bacteria and antibiotic resistant genes dissemination in urban river under tropical conditions. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 200:110767. [PMID: 32470679 DOI: 10.1016/j.ecoenv.2020.110767] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/21/2020] [Accepted: 05/14/2020] [Indexed: 05/12/2023]
Abstract
The occurrence and dissemination of antibiotic resistant genes (ARGs) that are associated with clinical pathogens and the evaluation of associated risks are still under-investigated in developing countries under tropical conditions. In this context, cultivable and molecular approaches were performed to assess the dissemination of bacteria and the antibiotic resistance genes in aquatic environment in Kinshasa, Democratic Republic of the Congo. Cultivable approach quantified β-lactam, carbapenem resistant, and total Escherichia coli and Enterobacteriaceae in river sediments and surface waters that receive raw hospital effluents. The molecular approach utilized Quantitative Polymerase Chain Reaction (qPCR) to quantify the total bacteria and the richness of relevant bacteria (Escherichia coli, Enterococcus, and Pseudomonas), and antibiotic resistance genes (ARGs: blaOXA-48, blaCTX-M, blaIMP, blaTEM) in sediment samples. Statistical analysis were employed to highlight the significance of hospital contribution and seasonal variation of bacteria and ARGs into aquatic ecosystems in suburban municipalities of Kinshasa, Democratic Republic of the Congo. The contribution of hospitals to antibiotic resistance proliferation is higher in the dry season than during the wet season (p < 0.05). Hospital similarly contributed Escherichia coli, Enterococcus, and Pseudomonas and ARGs significantly to the sediments in both seasons (p < 0.05). The organic matter content correlated positively with E. coli (r = 0.50, p < 0.05). The total bacterial load correlated with Enterococcus, and Pseudomonas (0.49 < r < 0.69, p < 0.05). Each ARG correlated with the total bacterial load or at least one relevant bacteria (0.41 < r < 0.81, p < 0.05). Our findings confirm that hospital wastewaters contributed significantly to antibiotic resistance profile and the significance of this contribution increased in the dry season. Moreover, our analysis highlights this risk from untreated hospital wastewaters in developing countries, which presents a great threat to public health.
Collapse
Affiliation(s)
- Dhafer Mohammed M Al Salah
- University of Geneva, Faculty of Sciences, Earth and Environmental Sciences, Institute F. A. Forel and Institute of Environmental Sciences, Bd Carl-Vogt 66, CH-1211, Geneva 4, Switzerland; King Abdulaziz City for Science and Technology, Joint Centers of Excellence Program, Prince Turki the 1st St, Riyadh, 11442, Saudi Arabia
| | - Georgette N Ngweme
- School of Public Health, Faculty of Medicine, University of Kinshasa, B.P. 11850, Kinshasa XI, Congo
| | - Amandine Laffite
- University of Geneva, Faculty of Sciences, Earth and Environmental Sciences, Institute F. A. Forel and Institute of Environmental Sciences, Bd Carl-Vogt 66, CH-1211, Geneva 4, Switzerland
| | - Jean-Paul Otamonga
- Université Pédagogique Nationale (UPN), Croisement Route de Matadi et Avenue de La Libération. Quartier Binza/UPN, B.P. 8815 Kinshasa, Congo
| | - Crispin Mulaji
- Faculty of Science, Department of Chemistry, University of Kinshasa, B.P. 190, Kinshasa XI, Congo
| | - John Poté
- University of Geneva, Faculty of Sciences, Earth and Environmental Sciences, Institute F. A. Forel and Institute of Environmental Sciences, Bd Carl-Vogt 66, CH-1211, Geneva 4, Switzerland; Université Pédagogique Nationale (UPN), Croisement Route de Matadi et Avenue de La Libération. Quartier Binza/UPN, B.P. 8815 Kinshasa, Congo; Faculty of Science, Department of Chemistry, University of Kinshasa, B.P. 190, Kinshasa XI, Congo.
| |
Collapse
|
81
|
Katale BZ, Misinzo G, Mshana SE, Chiyangi H, Campino S, Clark TG, Good L, Rweyemamu MM, Matee MI. Genetic diversity and risk factors for the transmission of antimicrobial resistance across human, animals and environmental compartments in East Africa: a review. Antimicrob Resist Infect Control 2020; 9:127. [PMID: 32762743 PMCID: PMC7409632 DOI: 10.1186/s13756-020-00786-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 07/21/2020] [Indexed: 12/30/2022] Open
Abstract
Background The emergence and spread of antimicrobial resistance (AMR) present a challenge to disease control in East Africa. Resistance to beta-lactams, which are by far the most used antibiotics worldwide and include the penicillins, cephalosporins, monobactams and carbapenems, is reducing options for effective control of both Gram-positive and Gram-negative bacteria. The World Health Organization, Food and Agricultural Organization and the World Organization for Animal Health have all advocated surveillance of AMR using an integrated One Health approach. Regional consortia also have strengthened collaboration to address the AMR problem through surveillance, training and research in a holistic and multisectoral approach. This review paper contains collective information on risk factors for transmission, clinical relevance and diversity of resistance genes relating to extended-spectrum beta-lactamase-producing (ESBL) and carbapenemase-producing Enterobacteriaceae, and Methicillin-resistant Staphylococcus aureus (MRSA) across the human, animal and environmental compartments in East Africa. Main body The review of the AMR literature (years 2001 to 2019) was performed using search engines such as PubMed, Scopus, Science Direct, Google and Web of Science. The search terms included ‘antimicrobial resistance and human-animal-environment’, ‘antimicrobial resistance, risk factors, genetic diversity, and human-animal-environment’ combined with respective countries of East Africa. In general, the risk factors identified were associated with the transmission of AMR. The marked genetic diversity due to multiple sequence types among drug-resistant bacteria and their replicon plasmid types sourced from the animal, human and environment were reported. The main ESBL, MRSA and carbapenem related genes/plasmids were the blaCTX-Ms (45.7%), SCCmec type III (27.3%) and IMP types (23.8%), respectively. Conclusion The high diversity of the AMR genes suggests there may be multiple sources of resistance bacteria, or the possible exchange of strains or a flow of genes amongst different strains due to transfer by mobile genetic elements. Therefore, there should be harmonized One Health guidelines for the use of antibiotics, as well as regulations governing their importation and sale. Moreover, the trend of ESBLs, MRSA and carbapenem resistant (CAR) carriage rates is dynamic and are on rise over time period, posing a public health concern in East Africa. Collaborative surveillance of AMR in partnership with regional and external institutions using an integrated One Health approach is required for expert knowledge and technology transfer to facilitate information sharing for informed decision-making.
Collapse
Affiliation(s)
- Bugwesa Z Katale
- Department of Microbiology and Immunology, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania. .,Tanzania Commission for Science and Technology, Dar es Salaam, Tanzania. .,SACIDS Foundation for One Health (SACIDS), Sokoine University of Agriculture, Morogoro, Tanzania.
| | - Gerald Misinzo
- SACIDS Foundation for One Health (SACIDS), Sokoine University of Agriculture, Morogoro, Tanzania.,Department of Veterinary Microbiology, Parasitology and Biotechnology, College of Veterinary Medicine and Biomedical Sciences, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Stephen E Mshana
- SACIDS Foundation for One Health (SACIDS), Sokoine University of Agriculture, Morogoro, Tanzania.,Department of Microbiology and Immunology, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Harriet Chiyangi
- Department of Microbiology and Immunology, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania.,SACIDS Foundation for One Health (SACIDS), Sokoine University of Agriculture, Morogoro, Tanzania
| | - Susana Campino
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Taane G Clark
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.,Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Liam Good
- Department of Pathobiology and Population Sciences, Royal Veterinary College, London, UK
| | - Mark M Rweyemamu
- SACIDS Foundation for One Health (SACIDS), Sokoine University of Agriculture, Morogoro, Tanzania.,Department of Veterinary Microbiology, Parasitology and Biotechnology, College of Veterinary Medicine and Biomedical Sciences, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Mecky I Matee
- Department of Microbiology and Immunology, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania.,SACIDS Foundation for One Health (SACIDS), Sokoine University of Agriculture, Morogoro, Tanzania
| |
Collapse
|
82
|
MCR-1 Gene Encoded Colistin-Resistant Escherichia coli in Raw Chicken Meat and Bean Sprouts in Malaysia. Int J Microbiol 2020; 2020:8853582. [PMID: 32774381 PMCID: PMC7407033 DOI: 10.1155/2020/8853582] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/03/2020] [Accepted: 07/01/2020] [Indexed: 11/17/2022] Open
Abstract
This study was conducted to detect the presence of colistin-resistant Escherichia coli (E. coli) in raw chicken meat and bean sprouts collected from local markets and to determine the antimicrobial resistance patterns of the E. coli isolates. A total of 100 samples, comprised of 50 raw chicken meat and 50 bean sprouts, were collected and processed. Kirby-Bauer method was used to determine the antimicrobial resistance patterns, and PCR amplification was used to detect E. coli species-specific and colistin resistance (mcr-1 and mcr-2) genes. The results showed that 52.1% (12/23) of the E. coli isolated from raw chicken meat were positive for the colistin resistance encoding gene, mcr-1, whereas all the E. coli isolates from bean sprouts were negative for colistin resistance encoding genes. The findings show that chicken meat contaminated with colistin-resistant E. coli may pose public health risk to the consumers. Hence, prudent usage of antibiotics and hygienic handling of food items helps to prevent and combat the risks of spreading of colistin-resistant E. coli and the public health risks it may pose. More comprehensive and large-scale studies focusing on all the possible sources of colistin-resistant E. coli are recommended.
Collapse
|
83
|
de Groot PWJ, Fernández-Pereira J, Sabariegos R, Clemente-Casares P, Parra-Martínez J, Cid VJ, Moreno DA. Optimizing Small World Initiative service learning by focusing on antibiotics-producing actinomycetes from soil. FEMS Microbiol Lett 2020; 366:5717303. [PMID: 31995181 DOI: 10.1093/femsle/fnaa019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 01/28/2020] [Indexed: 12/20/2022] Open
Abstract
Small World Initiative and Tiny Earth are popular citizen science programs that are implemented worldwide in response to the global antibiotic resistance crisis. When starting up the program in Albacete (Spain), we noted that rates of isolated antibiotic-producing bacteria are generally low. To make the activity more stimulating for participating students, we modified the protocol to obtain more positive results by focusing on isolation of actinomycetes, the main producers of most clinically used antibiotics. Adaptations involved redesigning culture media, incubation times and temperatures, and modification of the ESKAPE antibiosis experiment by employing an agar-transplantation step. Of 390 bacterial isolates tested, almost 6% tested positive in antibiosis experiments and DNA sequence analysis confirmed that all positives are actinomycetes, demonstrating that our protocol is efficient toward isolating antibiotic-producing actinomycetes from soil. Evaluation forms filled by participating students indicated that the program was received very positively and that our modifications contribute to make this educational program more stimulating and efficient.
Collapse
Affiliation(s)
- Piet W J de Groot
- Universidad de Castilla-La Mancha, Facultad de Farmacia, Avda. Dr. José María Sánchez Ibáñez s/n, E-02008 Albacete, Spain.,Universidad de Castilla-La Mancha, Facultad de Medicina, c/ Almansa 14, E-02008 Albacete, Spain.,Universidad de Castilla-La Mancha, Centro Regional de Investigaciones Biomédicas, c/ Almansa 14, E-02008 Albacete, Spain.,Fundación Parque Científico y Tecnológico de Castilla-La Mancha, Paseo de la Innovación 1, E-02006 Albacete, Spain
| | - Jordan Fernández-Pereira
- Universidad de Castilla-La Mancha, Facultad de Medicina, c/ Almansa 14, E-02008 Albacete, Spain.,Universidad de Castilla-La Mancha, Centro Regional de Investigaciones Biomédicas, c/ Almansa 14, E-02008 Albacete, Spain
| | - Rosario Sabariegos
- Universidad de Castilla-La Mancha, Facultad de Medicina, c/ Almansa 14, E-02008 Albacete, Spain.,Universidad de Castilla-La Mancha, Centro Regional de Investigaciones Biomédicas, c/ Almansa 14, E-02008 Albacete, Spain
| | - Pilar Clemente-Casares
- Universidad de Castilla-La Mancha, Facultad de Farmacia, Avda. Dr. José María Sánchez Ibáñez s/n, E-02008 Albacete, Spain.,Universidad de Castilla-La Mancha, Centro Regional de Investigaciones Biomédicas, c/ Almansa 14, E-02008 Albacete, Spain
| | - Javier Parra-Martínez
- Complejo Hospitalario Universitario de Albacete (CHUA), Servicio de Microbiología, c/ Hermanos Falcó 37, E-02006 Albacete, Spain
| | - Víctor J Cid
- Universidad Complutense de Madrid, Departamento de Microbiología y Parasitología, Facultad de Farmacia, Pza. Ramón y Cajal s/n. E-28040, Madrid, Spain
| | - Diego A Moreno
- Universidad de Castilla-La Mancha, Facultad de Farmacia, Avda. Dr. José María Sánchez Ibáñez s/n, E-02008 Albacete, Spain.,Universidad Politécnica de Madrid, Escuela Técnica Superior de Ingenieros Industriales, c/ José Gutiérrez Abascal 2, E-28006 Madrid, Spain
| |
Collapse
|
84
|
Nowrotek M, Jałowiecki Ł, Płaza G. Fluoroquinolone Resistance and Virulence Properties Among Wastewater Aeromonas caviae Isolates. Microb Drug Resist 2020; 27:179-189. [PMID: 32552456 DOI: 10.1089/mdr.2019.0287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The study provides data on antibiotic resistance as well as the virulence characteristics of Aeromonas caviae isolated from raw and treated wastewater. The isolates were identified as A. caviae by 16S rRNA gene sequencing. In the analyzed strains, high frequency for the following genes was observed: aac(6')-Ib-cr, qnrB, and qnrD. The presence of qnrA and ogxB genes was not found in any strain. The higher frequency of the investigated genes was observed in strains from raw wastewater (RW). The strains of A. caviae showed multiple antibiotic resistance evaluated by the disk diffusion method. Multiple antibiotic resistance indices ranged from 0.36 to 0.69. Susceptibility to six heavy metals (Cd+2, Zn+2, Cu+2, Co+2, Mn+2, and Ni+2) was recorded for all the isolates. The order of metal resistance of A. caviae was Co > Cu > Zn > Cd > Ni > Mn. All the strains of A. caviae showed β-hemolytic activity. Enzymes of amylase, cellulase, and lipase were produced by all isolates. Only the strains from RW had the ability to form biofilms and showed motility. The obtained results indicate that wastewater is a potential source and/or reservoir of virulent and multidrug-resistant A. caviae as "high-risk isolates."
Collapse
Affiliation(s)
- Monika Nowrotek
- Environmental Microbiology Unit, Institute for Ecology of Industrial Areas, Katowice, Poland
| | - Łukasz Jałowiecki
- Environmental Microbiology Unit, Institute for Ecology of Industrial Areas, Katowice, Poland
| | - Grażyna Płaza
- Environmental Microbiology Unit, Institute for Ecology of Industrial Areas, Katowice, Poland
| |
Collapse
|
85
|
Post SJ, Keohane CE, Rossiter LM, Kaplan AR, Khowsathit J, Matuska K, Karanicolas J, Wuest WM. Target-Based Design of Promysalin Analogues Identifies a New Putative Binding Cleft in Succinate Dehydrogenase. ACS Infect Dis 2020; 6:1372-1377. [PMID: 32286041 PMCID: PMC7293565 DOI: 10.1021/acsinfecdis.0c00024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Promysalin is a small-molecule natural product that specifically inhibits growth of the Gram-negative pathogen Pseudomonas aeruginosa (PA). This activity holds promise in the treatment of multidrug resistant infections found in immunocompromised patients with chronic illnesses, such as cystic fibrosis. In 2015, our lab completed the first total synthesis; subsequent analogue design and SAR investigation enabled identification of succinate dehydrogenase (Sdh) as the biological target in PA. Herein, we report the target-guided design of new promysalin analogues with varying alkyl chains, one of which is on par with our most potent analogue to date. Computational docking revealed that some analogues have a different orientation in the Sdh binding pocket, placing the terminal carbon proximal to a tryptophan residue. This inspired the design of an extended side chain analogue bearing a terminal phenyl moiety, providing a basis for the design of future analogues.
Collapse
Affiliation(s)
- Savannah J Post
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Colleen E Keohane
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Lauren M Rossiter
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Anna R Kaplan
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Jittasak Khowsathit
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, United States
| | - Katie Matuska
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - John Karanicolas
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, United States
| | - William M Wuest
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, United States
- Emory Antibiotic Resistance Center, Emory School of Medicine, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
86
|
Liu L, Wang Q, Lin H, Das R, Wang S, Qi H, Yang J, Xue Y, Mao D, Luo Y. Amoxicillin Increased Functional Pathway Genes and Beta-Lactam Resistance Genes by Pathogens Bloomed in Intestinal Microbiota Using a Simulator of the Human Intestinal Microbial Ecosystem. Front Microbiol 2020; 11:1213. [PMID: 32582117 PMCID: PMC7287123 DOI: 10.3389/fmicb.2020.01213] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
Antibiotics are frequently used to treat bacterial infections; however, they affect not only the target pathogen but also commensal gut bacteria. They may cause the dysbiosis of human intestinal microbiota and consequent metabolic alterations, as well as the spreading of antibiotic resistant bacteria and antibiotic resistance genes (ARGs). In vitro experiments by simulator of the human intestinal microbial ecosystem (SHIME) can clarify the direct effects of antibiotics on different regions of the human intestinal microbiota, allowing complex human microbiota to be stably maintained in the absence of host cells. However, there are very few articles added the antibiotics into this in vitro model to observe the effects of antibiotics on the human intestinal microbiota. To date, no studies have focused on the correlations between the bloomed pathogens caused by amoxicillin (AMX) exposure and increased functional pathway genes as well as ARGs. This study investigated the influence of 600 mg day-1 AMX on human intestinal microbiota using SHIME. The impact of AMX on the composition and function of the human intestinal microbiota was revealed by 16S rRNA gene sequencing and high-throughput quantitative PCR. The results suggested that: (i) AMX treatment has tremendous influence on the overall taxonomic composition of the gut microbiota by increasing the relative abundance of Klebsiella [linear discriminant analysis (LDA) score = 5.26] and Bacteroides uniformis (LDA score = 4.75), as well as taxonomic diversity (Simpson, P = 0.067, T-test; Shannon, P = 0.061, T-test), and decreasing the members of Parabacteroides (LDA score = 4.18), Bifidobacterium (LDA score = 4.06), and Phascolarctobacterium (LDA score = 3.95); (ii) AMX exposure significantly enhanced the functional pathway genes and beta-lactam resistance genes, and the bloomed pathogens were strongly correlated with the metabolic and immune system diseases gene numbers (R = 0.98, P < 0.001) or bl2_len and bl2be_shv2 abundance (R = 0.94, P < 0.001); (iii) the changes caused by AMX were "SHIME-compartment" different with more significant alteration in ascending colon, and the effects were permanent, which could not be restored after 2-week AMX discontinuance. Overall results demonstrated negative side-effects of AMX, which should be considered for AMX prescription.
Collapse
Affiliation(s)
- Lei Liu
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
| | - Qing Wang
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
- Hebei Key Laboratory of Air Pollution Cause and Impact (preparatory), College of Energy and Environmental Engineering, Hebei University of Engineering, Handan, China
| | - Huai Lin
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
| | - Ranjit Das
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
| | - Siyi Wang
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
| | - Hongmei Qi
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
| | - Jing Yang
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
| | - Yingang Xue
- Key Laboratory of Environmental Protection of Water Environment Biological Monitoring of Jiangsu Province, Changzhou Environmental Monitoring Center, Changzhou, China
| | - Daqing Mao
- School of Medicine, Nankai University, Tianjin, China
| | - Yi Luo
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin, China
| |
Collapse
|
87
|
Acosta MM, Bram JT, Sim D, Read AF. Effect of drug dose and timing of treatment on the emergence of drug resistance in vivo in a malaria model. Evol Med Public Health 2020; 2020:196-210. [PMID: 33209305 PMCID: PMC7652304 DOI: 10.1093/emph/eoaa016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/15/2020] [Accepted: 05/26/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND OBJECTIVES There is a significant interest in identifying clinically effective drug treatment regimens that minimize the de novo evolution of antimicrobial resistance in pathogen populations. However, in vivo studies that vary treatment regimens and directly measure drug resistance evolution are rare. Here, we experimentally investigate the role of drug dose and treatment timing on resistance evolution in an animal model. METHODOLOGY In a series of experiments, we measured the emergence of atovaquone-resistant mutants of Plasmodium chabaudi in laboratory mice, as a function of dose or timing of treatment (day post-infection) with the antimalarial drug atovaquone. RESULTS The likelihood of high-level resistance emergence increased with atovaquone dose. When varying the timing of treatment, treating either very early or late in infection reduced the risk of resistance. When we varied starting inoculum, resistance was more likely at intermediate inoculum sizes, which correlated with the largest population sizes at time of treatment. CONCLUSIONS AND IMPLICATIONS (i) Higher doses do not always minimize resistance emergence and can promote the emergence of high-level resistance. (ii) Altering treatment timing affects the risk of resistance emergence, likely due to the size of the population at the time of treatment, although we did not test the effect of immunity whose influence may have been important in the case of late treatment. (iii) Finding the 'right' dose and 'right' time to maximize clinical gains and limit resistance emergence can vary depending on biological context and was non-trivial even in our simplified experiments. LAY SUMMARY In a mouse model of malaria, higher drug doses led to increases in drug resistance. The timing of drug treatment also impacted resistance emergence, likely due to the size of the population at the time of treatment.
Collapse
Affiliation(s)
- Mónica M Acosta
- Department of Biology, Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
| | - Joshua T Bram
- Department of Biology, Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
| | - Derek Sim
- Department of Biology, Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
| | - Andrew F Read
- Department of Biology, Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
88
|
Hansen E, Karslake J, Woods RJ, Read AF, Wood KB. Antibiotics can be used to contain drug-resistant bacteria by maintaining sufficiently large sensitive populations. PLoS Biol 2020; 18:e3000713. [PMID: 32413038 PMCID: PMC7266357 DOI: 10.1371/journal.pbio.3000713] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/02/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
Standard infectious disease practice calls for aggressive drug treatment that rapidly eliminates the pathogen population before resistance can emerge. When resistance is absent, this elimination strategy can lead to complete cure. However, when resistance is already present, removing drug-sensitive cells as quickly as possible removes competitive barriers that may slow the growth of resistant cells. In contrast to the elimination strategy, a containment strategy aims to maintain the maximum tolerable number of pathogens, exploiting competitive suppression to achieve chronic control. Here, we combine in vitro experiments in computer-controlled bioreactors with mathematical modeling to investigate whether containment strategies can delay failure of antibiotic treatment regimens. To do so, we measured the "escape time" required for drug-resistant Escherichia coli populations to eclipse a threshold density maintained by adaptive antibiotic dosing. Populations containing only resistant cells rapidly escape the threshold density, but we found that matched resistant populations that also contain the maximum possible number of sensitive cells could be contained for significantly longer. The increase in escape time occurs only when the threshold density-the acceptable bacterial burden-is sufficiently high, an effect that mathematical models attribute to increased competition. The findings provide decisive experimental confirmation that maintaining the maximum number of sensitive cells can be used to contain resistance when the size of the population is sufficiently large.
Collapse
Affiliation(s)
- Elsa Hansen
- Center for Infectious Disease Dynamics, Department of Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Jason Karslake
- Department of Biophysics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Robert J. Woods
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Andrew F. Read
- Center for Infectious Disease Dynamics, Huck Institutes of the Life Sciences and Departments of Biology and Entomology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Kevin B. Wood
- Department of Biophysics, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Physics, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
89
|
Cefotiam Treatment in Children: Evidence of Subtherapeutic Levels. Ther Drug Monit 2020; 42:733-736. [PMID: 32251152 DOI: 10.1097/ftd.0000000000000759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Cefotiam, a second-generation cephalosporin, is a broad-spectrum antibiotic with good antibacterial action against both gram-negative and gram-positive bacteria. It is used widely in clinical practice, although bacterial drug resistance makes its clinical use problematic. The authors hypothesized that subtherapeutic concentrations of cefotiam leads to bacterial resistance. The present study was conducted to evaluate whether the standard cefotiam dosing regimen resulted in a subtherapeutic concentrations in children. METHOD Data were prospectively collected from pediatric patients with suspected or confirmed community-acquired pneumonia who were receiving cefotiam at the standard dosing regimen (40-80 mg/kg, 2 or 3 times daily). A blood sample was collected after 70%-100% of the dosing interval, and plasma concentrations were determined by high-performance liquid chromatography using an ultraviolet detector. RESULTS The data from 88 patients (age, 3.0 ± 2.8 years; weight, 15.4 ± 8.3 kg) were used for analysis. The average of cefotiam concentrations was 0.06 mcg/mL (range: <0.05-0.79 mcg/mL). Most patients (n = 72, 81.8%) had concentrations below 0.1 mcg/mL; only 2 patients had concentrations higher than 0.4 mcg/mL. CONCLUSIONS The standard dosing regimen for cefotiam resulted in extremely low plasma concentrations in children; such low concentrations may lead to antimicrobial drug resistance. Thus, an increase in cefotiam dosage in children to 80 mg/kg 4 times daily is recommended (maximum dose on the label).
Collapse
|
90
|
DeNegre AA, Myers K, Fefferman NH. Impact of Strain Competition on Bacterial Resistance in Immunocompromised Populations. Antibiotics (Basel) 2020; 9:antibiotics9030114. [PMID: 32156072 PMCID: PMC7148506 DOI: 10.3390/antibiotics9030114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/27/2020] [Accepted: 03/04/2020] [Indexed: 01/21/2023] Open
Abstract
Despite the risk of emerging drug resistance that occurs with the frequent use of antimicrobial agents, targeted and prophylactic antibiotics have been considered crucial to opportunistic infection management among the HIV/AIDS-immunocompromised. As we recently demonstrated, the disrupted selective pressures that occur in AIDS-prevalent host populations increase the probability of novel emergence. This effect is concerning, given that bacterial strains unresponsive to first-line antibiotics can be particularly dangerous to hosts whose immune response is insufficient to fight infection in the absence of antibiotic support. While greater host susceptibility within a highly immunocompromised population may offer a fitness advantage to drug-resistant bacterial strains, this advantage could be mitigated by increased morbidity and mortality among the AIDS-immunocompromised. Using a Susceptible-Exposed-Infectious-Recovered (SEIR) epidemiological model parameterized to reflect conditions in an AIDS-prevalent host population, we examine the evolutionary relationship between drug-sensitive and -resistant strains of Mycobacterium tuberculosis. We explore this relationship when the fitness of the resistant strain is varied relative to that of the sensitive strain to investigate the likely long-term multi-strain dynamics of the AIDS-mediated increased emergence of drug resistance.
Collapse
Affiliation(s)
- Ashley A. DeNegre
- Department of Ecology, Evolution and Natural Resources, Rutgers University, New Brunswick, NJ 08901, USA;
- The Command, Control and Interoperability Center for Advanced Data Analysis (CCICADA), Rutgers University, New Brunswick, NJ 08901, USA
| | - Kellen Myers
- Department of Ecology & Evolutionary Biology, University of Tennessee, Knoxville, TN 37996, USA;
- Department of Mathematics, University of Tennessee, Knoxville, TN 37996, USA
- National Institute for Mathematical and Biological Synthesis (NIMBioS), University of Tennessee, Knoxville, TN 37996, USA
- Department of Mathematics, Tusculum University, Greeneville, TN 37745, USA
| | - Nina H. Fefferman
- Department of Ecology, Evolution and Natural Resources, Rutgers University, New Brunswick, NJ 08901, USA;
- The Command, Control and Interoperability Center for Advanced Data Analysis (CCICADA), Rutgers University, New Brunswick, NJ 08901, USA
- Department of Ecology & Evolutionary Biology, University of Tennessee, Knoxville, TN 37996, USA;
- Department of Mathematics, University of Tennessee, Knoxville, TN 37996, USA
- National Institute for Mathematical and Biological Synthesis (NIMBioS), University of Tennessee, Knoxville, TN 37996, USA
- Correspondence:
| |
Collapse
|
91
|
Role of antibiotic stress in phenotypic switching to persister cells of antibiotic-resistant Staphylococcus aureus. ANN MICROBIOL 2020. [DOI: 10.1186/s13213-020-01552-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Abstract
Purpose
This study was designed to evaluate phenotypic and genotypic properties of persister cells formed by Staphylococcus aureus ATCC 15564 (SAWT), oxacillin-induced S. aureus (SAOXA), ciprofloxacin-induced S. aureus (SACIP), and clinically isolated multidrug-resistant S. aureus CCARM 3080 (SAMDR).
Methods
The dose-dependent biphasic killing patterns were observed for SAWT, SAOXA, SACIP, and SAMDR in response to twofold minimum inhibitory concentrate (MIC) of ciprofloxacin. The surviving cells of SAWT, SAOXA, SACIP, and SAMDR after twofold MIC of ciprofloxacin treatment were analyzed using a metabolic-based assay to estimate the fractions of persister cells.
Results
The least persister formation was induced in SACIP after twofold MIC of ciprofloxacin treatment, showing 58% of persistence. The lowest fitness cost of resistance was observed for the recovered persister cells of SACIP (relative fitness = 0.95), followed by SAMDR (relative fitness = 0.70), while the highest fitness cost was observed for SAWT (relative fitness = 0.26). The mRNA transcripts were analyzed by RT-PCR assay in recovered persister cells pre-incubated with ciprofloxacin. The highest expression levels of stress-related genes (dnaK and groEL) and efflux pump-related genes (mepR, norA, and norB) were observed in the recovered persister cells of SAOXA and SAMDR.
Conclusion
This study provides valuable information for understanding crosstalk between antibiotic resistance, tolerance, and persistence in different antibiotic-resistant S. aureus strains.
Collapse
|
92
|
Rudnick W, Science M, Thirion DJG, Abdesselam K, Choi KB, Pelude L, Amaratunga K, Comeau JL, Dalton B, Delport J, Dhami R, Embree J, Émond Y, Evans G, Frenette C, Fryters S, German G, Grant JM, Happe J, Katz K, Kibsey P, Kosar J, Langley JM, Lee BE, Lefebvre MA, Leis JA, McGeer A, Neville HL, Simor A, Slayter K, Suh KN, Tse-Chang A, Weiss K, Conly J. Antimicrobial use among adult inpatients at hospital sites within the Canadian Nosocomial Infection Surveillance Program: 2009 to 2016. Antimicrob Resist Infect Control 2020; 9:32. [PMID: 32054539 PMCID: PMC7020554 DOI: 10.1186/s13756-020-0684-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/23/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Antimicrobial resistance is a growing threat to the world's ability to prevent and treat infections. Links between quantitative antibiotic use and the emergence of bacterial resistance are well documented. This study presents benchmark antimicrobial use (AMU) rates for inpatient adult populations in acute-care hospitals across Canada. METHODS In this retrospective surveillance study, acute-care adult hospitals participating in the Canadian Nosocomial Infection Surveillance Program (CNISP) submitted annual AMU data on all systemic antimicrobials from 2009 to 2016. Information specific to intensive care units (ICUs) and non-ICU wards were available for 2014-2016. Data were analyzed using defined daily doses (DDD) per 1000 patient days (DDD/1000pd). RESULTS Between 2009 and 2016, 16-18 CNISP adult hospitals participated each year and provided their AMU data (22 hospitals participated in ≥1 year of surveillance; 11 in all years). From 2009 to 2016, there was a significant reduction in use (12%) (from 654 to 573 DDD/1000pd, p = 0.03). Fluoroquinolones accounted for the majority of this decrease (47% reduction in combined oral and intravenous use, from 129 to 68 DDD/1000pd, p < 0.002). The top five antimicrobials used in 2016 were cefazolin (78 DDD/1000pd), piperacillin-tazobactam (53 DDD/1000pd), ceftriaxone (49 DDD/1000pd), vancomycin (combined oral and intravenous use was 44 DDD/1000pd; 7% of vancomycin use was oral), and ciprofloxacin (combined oral and intravenous use: 42 DDD/1000pd). Among the top 10 antimicrobials used in 2016, ciprofloxacin and metronidazole use decreased significantly between 2009 and 2016 by 46% (p = 0.002) and 26% (p = 0.002) respectively. Ceftriaxone (85% increase, p = 0.0008) and oral amoxicillin-clavulanate (140% increase, p < 0.0001) use increased significantly but contributed only a small component (8.6 and 5.0%, respectively) of overall use. CONCLUSIONS This study represents the largest collection of dispensed antimicrobial use data among inpatients in Canada to date. Between 2009 and 2016, there was a significant 12% decrease in AMU, driven primarily by a 47% decrease in fluoroquinolone use. Modest absolute increases in parenteral ceftriaxone and oral amoxicillin-clavulanate use were noted but contributed a small amount of total AMU. Ongoing national surveillance is crucial for establishing benchmarks and antimicrobial stewardship guidelines.
Collapse
Affiliation(s)
- Wallis Rudnick
- Public Health Agency of Canada, 130 Colonnade Rd, Ottawa, ON K2E 7L9 Canada
| | | | - Daniel J. G. Thirion
- Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, QC H3T 1J4 Canada
- McGill University Health Centre, 1001 Boulevard Décarie, Montréal, QC H4A 3J1 Canada
| | - Kahina Abdesselam
- Public Health Agency of Canada, 130 Colonnade Rd, Ottawa, ON K2E 7L9 Canada
| | - Kelly B. Choi
- Public Health Agency of Canada, 130 Colonnade Rd, Ottawa, ON K2E 7L9 Canada
| | - Linda Pelude
- Public Health Agency of Canada, 130 Colonnade Rd, Ottawa, ON K2E 7L9 Canada
| | - Kanchana Amaratunga
- Public Health Agency of Canada, 130 Colonnade Rd, Ottawa, ON K2E 7L9 Canada
- The Ottawa Hospital, 501 Smyth Rd, Ottawa, ON K1H 8L6 Canada
| | - Jeannette L. Comeau
- IWK Health Centre, 5980 University Ave, Halifax, NS B3K 6R8 Canada
- Dalhousie University, 6299 South St, Halifax, NS B3H 4R2 Canada
| | - Bruce Dalton
- Alberta Health Services, 1620 29 St NW, Calgary, AB T2N 4L7 Canada
| | - Johan Delport
- London Health Sciences Centre, 800 Commissioners Rd E, London, ON N6A 5W9 Canada
| | - Rita Dhami
- London Health Sciences Centre, 800 Commissioners Rd E, London, ON N6A 5W9 Canada
- University of Waterloo, 200 University Ave W, Waterloo, ON N2L 3G1 Canada
- University of Western Ontario, 1151 Richmond St, London, ON N6A 3K7 Canada
| | - Joanne Embree
- University of Manitoba, Winnipeg, MB R3T 2N2 Canada
- Shared Health Manitoba, Winnipeg, MB R3T 2N2 Canada
- Children’s Hospital Winnipeg, 840 Sherbrook St, Winnipeg, MB R3E 0Z3 Canada
| | - Yannick Émond
- Hôpital Maisonneuve-Rosemont, 5415 Boulevard de l’Assomption, Montréal, QC H1T 2M4 Canada
| | - Gerald Evans
- Kingston General Hospital, 76 Stuart St, Kingston, ON K7L 2V7 Canada
| | - Charles Frenette
- McGill University Health Centre, 1001 Boulevard Décarie, Montréal, QC H4A 3J1 Canada
| | - Susan Fryters
- Alberta Health Services, 10240 Kingsway Avenue, Edmonton, AB T5H 3V9 Canada
| | - Greg German
- Health PEI, 16 Garfield St, Charlottetown, PEI C1A 6A5 Canada
| | - Jennifer M. Grant
- University of British Columbia, 2329 West Mall, Vancouver, BC V6T 1Z4 Canada
| | - Jennifer Happe
- Infection Prevention and Control Canada, Red Deer, AB T4N 6R2 Canada
| | - Kevin Katz
- North York General Hospital, 4001 Leslie St, North York, ON M2K 1E1 Canada
| | - Pamela Kibsey
- Royal Jubilee Hospital, 1952 Bay St, Victoria, BC V8R 1J8 Canada
| | - Justin Kosar
- Saskatchewan Health Authority, Saskatoon, SK S7N 0W8 Canada
| | - Joanne M. Langley
- IWK Health Centre, 5980 University Ave, Halifax, NS B3K 6R8 Canada
- Dalhousie University, 6299 South St, Halifax, NS B3H 4R2 Canada
| | - Bonita E. Lee
- Stollery Children’s Hospital, Edmonton, AB T6G 2B7 Canada
- University of Alberta, Edmonton, AB T6G 2R7 Canada
| | - Marie-Astrid Lefebvre
- McGill University Health Centre, 1001 Boulevard Décarie, Montréal, QC H4A 3J1 Canada
| | - Jerome A. Leis
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5 Canada
| | - Allison McGeer
- Sinai Health System, 600 University Ave, Toronto, ON M5G 1X5 Canada
- University of Toronto, 27 King’s College Cir, Toronto, ON M5S Canada
- Dalla Lana School of Public Health, University of Toronto, 155 College St, Toronto, ON M5T 3M7 Canada
| | - Heather L. Neville
- Nova Scotia Health Authority, 1276 South Park St, Halifax, NS B3H 2Y9 Canada
| | - Andrew Simor
- University of Toronto, 27 King’s College Cir, Toronto, ON M5S Canada
- Sunnybrook Health Sciences Centre, 2015 Bayview Ave, Toronto, ON M4N 3M5 Canada
| | - Kathryn Slayter
- IWK Health Centre, 5980 University Ave, Halifax, NS B3K 6R8 Canada
| | - Kathryn N. Suh
- The Ottawa Hospital, 501 Smyth Rd, Ottawa, ON K1H 8L6 Canada
| | - Alena Tse-Chang
- Stollery Children’s Hospital, Edmonton, AB T6G 2B7 Canada
- University of Alberta, Edmonton, AB T6G 2R7 Canada
| | - Karl Weiss
- SMBD-Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1E2 Canada
| | - John Conly
- Alberta Health Services, 1620 29 St NW, Calgary, AB T2N 4L7 Canada
- University of Calgary, 3330 Hospital Dr NW, Calgary, AB T2N 4N1 Canada
| | - the Canadian Nosocomial Infection Surveillance Program
- Public Health Agency of Canada, 130 Colonnade Rd, Ottawa, ON K2E 7L9 Canada
- SickKids, 555 University Ave, Toronto, ON M5G 1X8 Canada
- Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, QC H3T 1J4 Canada
- McGill University Health Centre, 1001 Boulevard Décarie, Montréal, QC H4A 3J1 Canada
- The Ottawa Hospital, 501 Smyth Rd, Ottawa, ON K1H 8L6 Canada
- IWK Health Centre, 5980 University Ave, Halifax, NS B3K 6R8 Canada
- Dalhousie University, 6299 South St, Halifax, NS B3H 4R2 Canada
- Alberta Health Services, 1620 29 St NW, Calgary, AB T2N 4L7 Canada
- London Health Sciences Centre, 800 Commissioners Rd E, London, ON N6A 5W9 Canada
- University of Waterloo, 200 University Ave W, Waterloo, ON N2L 3G1 Canada
- University of Western Ontario, 1151 Richmond St, London, ON N6A 3K7 Canada
- University of Manitoba, Winnipeg, MB R3T 2N2 Canada
- Shared Health Manitoba, Winnipeg, MB R3T 2N2 Canada
- Children’s Hospital Winnipeg, 840 Sherbrook St, Winnipeg, MB R3E 0Z3 Canada
- Hôpital Maisonneuve-Rosemont, 5415 Boulevard de l’Assomption, Montréal, QC H1T 2M4 Canada
- Kingston General Hospital, 76 Stuart St, Kingston, ON K7L 2V7 Canada
- Alberta Health Services, 10240 Kingsway Avenue, Edmonton, AB T5H 3V9 Canada
- Health PEI, 16 Garfield St, Charlottetown, PEI C1A 6A5 Canada
- University of British Columbia, 2329 West Mall, Vancouver, BC V6T 1Z4 Canada
- Infection Prevention and Control Canada, Red Deer, AB T4N 6R2 Canada
- North York General Hospital, 4001 Leslie St, North York, ON M2K 1E1 Canada
- Royal Jubilee Hospital, 1952 Bay St, Victoria, BC V8R 1J8 Canada
- Saskatchewan Health Authority, Saskatoon, SK S7N 0W8 Canada
- Stollery Children’s Hospital, Edmonton, AB T6G 2B7 Canada
- University of Alberta, Edmonton, AB T6G 2R7 Canada
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5 Canada
- Sinai Health System, 600 University Ave, Toronto, ON M5G 1X5 Canada
- University of Toronto, 27 King’s College Cir, Toronto, ON M5S Canada
- Dalla Lana School of Public Health, University of Toronto, 155 College St, Toronto, ON M5T 3M7 Canada
- Nova Scotia Health Authority, 1276 South Park St, Halifax, NS B3H 2Y9 Canada
- Sunnybrook Health Sciences Centre, 2015 Bayview Ave, Toronto, ON M4N 3M5 Canada
- SMBD-Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1E2 Canada
- University of Calgary, 3330 Hospital Dr NW, Calgary, AB T2N 4N1 Canada
| |
Collapse
|
93
|
Serrano HDA, Mariezcurrena-Berasain MA, Del Carmen Gutiérrez Castillo A, Carranza BV, Pliego AB, Rojas MT, Anele UY, Salem AZM, Rivas-Caceres RR. Antimicrobial resistance of three common molecularly identified pathogenic bacteria to Allium aqueous extracts. Microb Pathog 2020; 142:104028. [PMID: 32045644 DOI: 10.1016/j.micpath.2020.104028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023]
Abstract
The aim of this work was to evaluate the in vitro bacterial inhibition of different types of garlic on Escherichia coli ATCC 25922, Listeria monocytogenes and Staphylococcus aureus. The bacterial strains were molecularly identified using gen 16S rDNA molecular identification. Four different types of garlics were used: 1) white, 2) Japanese, 3) elephant and 3) black, and these were evaluated at two different concentrations (0.25 and 0.125 g/mL) per garlic type. Bioactive compounds present in the garlics were identified using high-performance liquid chromatography coupled to ultraviolet detector (HPLC-UV), and total polyphenols were quantified by the Folin-Ciocalteu technique. The Kirby-Bauber method was used for the bacterial evaluation. Aqueous extract of black garlic had the highest amount of polyphenols 6.26 ± 0.21 mg GAE/mL. The area of inhibition was measured and classified as sensitive, intermediate or resistant. Using the disc diffusion assay, higher concentration (0.25 g/mL) of aqueous extract of white garlic had the highest antibacterial activity area, with 21.46 ± 3.94 mm for L. monocytogenes, 20.61 ± 2.47 mm for S. aureus and 17.83 ± 2.21 mm for E. coli. White garlic had comparable antimicrobial activity as the control (tetracycline at 30 μg) as indicated by the size of the inhibition halos. Based on your results, white garlic can be used as an alternative to synthetic antimicrobials.
Collapse
Affiliation(s)
- Héctor D Arzate Serrano
- Universidad Autónoma del Estado de México, Facultad de Medicina Veterinaria y Zootecnia, Toluca, México, C.P. 50090, Mexico
| | - María A Mariezcurrena-Berasain
- Universidad Autónoma del Estado de México, Facultad de Medicina Veterinaria y Zootecnia, Toluca, México, C.P. 50090, Mexico.
| | | | - Benjamín Valladares Carranza
- Universidad Autónoma del Estado de México, Facultad de Medicina Veterinaria y Zootecnia, Toluca, México, C.P. 50090, Mexico
| | - Alberto Barbabosa Pliego
- Universidad Autónoma del Estado de México, Facultad de Medicina Veterinaria y Zootecnia, Toluca, México, C.P. 50090, Mexico
| | - Martín Talavera Rojas
- Universidad Autónoma del Estado de México, Facultad de Medicina Veterinaria y Zootecnia, Toluca, México, C.P. 50090, Mexico
| | - Uchenna Y Anele
- North Carolina Agricultural and Technical State University, Greensboro, NC, 27411, USA
| | - Abdelfattah Z M Salem
- Universidad Autónoma del Estado de México, Facultad de Medicina Veterinaria y Zootecnia, Toluca, México, C.P. 50090, Mexico
| | | |
Collapse
|
94
|
Popov G, Evdokimova E, Stogios PJ, Savchenko A. Structure of the full-length Serratia marcescens acetyltransferase AAC(3)-Ia in complex with coenzyme A. Protein Sci 2020; 29:803-808. [PMID: 31876342 DOI: 10.1002/pro.3811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022]
Abstract
Acyl-coenzyme A-dependent N-acetyltransferases (AACs) catalyze the modification of aminoglycosides rendering the bacteria carrying such enzymes resistant to this class of antibiotics. Here we present the crystal structure of AAC(3)-Ia enzyme from Serratia marcescens in complex with coenzyme A determined to 1.8 Å resolution. This enzyme served as an architype for the AAC enzymes targeting the amino group at Position 3 of aminoglycoside main aminocyclitol ring. The structure of this enzyme has been previously determined only in truncated form and was interpreted as distinct from subsequently characterized AACs. The reason for the unusual arrangement of secondary structure elements of AAC(3)-Ia was not further investigated. By determining the full-length structure of AAC(3)-Ia we establish that this enzyme adopts the canonical AAC fold conserved across this family and it does not undergo through significant rearrangement of secondary structure elements upon ligand binding as was proposed previously. In addition, our results suggest that the C-terminal tail in AAC(3)-Ia monomer forms intramolecular hydrogen bonds that contributes to formation of stable dimer, representing the predominant oligomeric state for this enzyme.
Collapse
Affiliation(s)
- Georgy Popov
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Elena Evdokimova
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada.,Center for Structural Genomics for Infectious Diseases, University of Calgary, Calgary, Canada
| | - Peter J Stogios
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada.,Center for Structural Genomics for Infectious Diseases, University of Calgary, Calgary, Canada
| | - Alexei Savchenko
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada.,Center for Structural Genomics for Infectious Diseases, University of Calgary, Calgary, Canada
| |
Collapse
|
95
|
Ruden S, Rieder A, Chis Ster I, Schwartz T, Mikut R, Hilpert K. Synergy Pattern of Short Cationic Antimicrobial Peptides Against Multidrug-Resistant Pseudomonas aeruginosa. Front Microbiol 2019; 10:2740. [PMID: 31849888 PMCID: PMC6901909 DOI: 10.3389/fmicb.2019.02740] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 11/11/2019] [Indexed: 12/18/2022] Open
Abstract
With the rise of various multidrug-resistant (MDR) pathogenic bacteria, worldwide health care is under pressure to respond. Conventional antibiotics are failing and the development of novel classes and alternative strategies is a major priority. Antimicrobial peptides (AMPs) cannot only kill MDR bacteria, but also can be used synergistically with conventional antibiotics. We selected 30 short AMPs from different origins and measured their synergy in combination with polymyxin B, piperacillin, ceftazidime, cefepime, meropenem, imipenem, tetracycline, erythromycin, kanamycin, tobramycin, amikacin, gentamycin, and ciprofloxacin. In total, 403 unique combinations were tested against an MDR Pseudomonas aeruginosa isolate (PA910). As a measure of the synergistic effects, fractional inhibitory concentrations (FICs) were determined using microdilution assays with FICs ranges between 0.25 and 2. A high number of combinations between peptides and polymyxin B, erythromycin, and tetracycline were found to be synergistic. Novel variants of indolicidin also showed a high frequency in synergist interaction. Single amino acid substitutions within the peptides can have a very strong effect on the ability to synergize, making it possible to optimize future drugs toward synergistic interaction.
Collapse
Affiliation(s)
- Serge Ruden
- Institute of Biological Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Annika Rieder
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Irina Chis Ster
- Institute of Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Thomas Schwartz
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Kai Hilpert
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Infection and Immunity, St George's, University of London, London, United Kingdom.,Institute of Microstructure Technology, Karlsruhe Institute of Technology, Karlsruhe, Germany
| |
Collapse
|
96
|
Ben-Shimol S, Givon-Lavi N, Greenberg D, Stein M, Megged O, Bar-Yochai A, Negari S, Dagan R, On Behalf Of The Israel Bacteremia And Meningitis Active Surveillance Group. Impact of pneumococcal conjugate vaccines introduction on antibiotic resistance of Streptococcus pneumoniae meningitis in children aged 5 years or younger, Israel, 2004 to 2016. ACTA ACUST UNITED AC 2019; 23. [PMID: 30482264 PMCID: PMC6341944 DOI: 10.2807/1560-7917.es.2018.23.47.1800081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background Empiric treatment of pneumococcal meningitis includes ceftriaxone with vancomycin to overcome ceftriaxone resistant disease. The addition of vancomycin bears a risk of adverse events, including increased antibiotic resistance. We assessed antibiotic resistance rates in pneumococcal meningitis before and after pneumococcal conjugate vaccine (PCV) implementation. Methods All pneumococcal meningitis episodes in children aged 5 years and younger, from 2004 to 2016, were extracted from the nationwide bacteremia and meningitis surveillance database. For comparison purposes, we defined pre-PCV period as 2004–2008 and PCV13 period as 2014–2016. Minimal inhibitory concentration (MIC) > 0.06 and > 0.5 μg/mL were defined as penicillin and ceftriaxone resistance, respectively. Results Overall, 325 episodes were identified. Pneumococcal meningitis incidence rates declined non-significantly by 17%, comparing PCV13 and pre-PCV periods. Throughout the study, 90% of isolates were tested for antibiotic susceptibility, with 26.6%, 2.1% and 0% of isolates resistant to penicillin, ceftriaxone and vancomycin, respectively. Mean proportions (± SD) of meningitis caused by penicillin-resistant pneumococci were 40.5% ± 8.0% and 9.6% ± 7.4% in the pre-PCV and the PCV13 periods, respectively, resulting in an overall 83.9% reduction (odd ratio:0.161; 95% confidence interval: 0.059–0.441) in penicillin resistance rates. The proportions of meningitis caused by ceftriaxone resistant pneumococci were 5.0% ± 0.8% in the pre-PCV period, but no ceftriaxone resistant isolates were identified since 2010. Conclusions PCV7/PCV13 sequential introduction resulted in > 80% reduction of penicillin- resistant pneumococcal meningitis and complete disappearance of ceftriaxone resistant disease. These trends should be considered by the treating physician when choosing an empiric treatment for pneumococcal meningitis.
Collapse
Affiliation(s)
- Shalom Ben-Shimol
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer-Sheva, Israel
| | - Noga Givon-Lavi
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer-Sheva, Israel
| | - David Greenberg
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer-Sheva, Israel
| | - Michal Stein
- Infectious Diseases and Infection Control Unit, Hillel Yaffe Medical Center, Hadera, Israel and Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Orli Megged
- Pediatric Infectious Diseases Unit, Shaare Zedek Medical Center, affiliated with Hebrew University-Hadassah School of Medicine, Jerusalem, Israel
| | - Avihu Bar-Yochai
- Infectious Disease Unit, Assaf Harofe Medical Center, Zerifin, Israel
| | - Shahar Negari
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,The Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer-Sheva, Israel
| | - Ron Dagan
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | |
Collapse
|
97
|
Antonelli P, Belluco S, Mancin M, Losasso C, Ricci A. Genes conferring resistance to critically important antimicrobials in Salmonella enterica isolated from animals and food: A systematic review of the literature, 2013–2017. Res Vet Sci 2019; 126:59-67. [DOI: 10.1016/j.rvsc.2019.08.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/12/2019] [Accepted: 08/12/2019] [Indexed: 01/03/2023]
|
98
|
Das L, Virmani R, Sharma V, Rawat D, Singh Y. Human Milk Microbiota: Transferring the Antibiotic Resistome to Infants. Indian J Microbiol 2019; 59:410-416. [PMID: 31762502 DOI: 10.1007/s12088-019-00824-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 08/30/2019] [Indexed: 12/13/2022] Open
Abstract
Commensal bacterial population is believed to be a reservoir for antibiotic resistance genes (ARGs). The infant gut microbiota has relatively higher abundance of ARGs than the adults. These genes can get transferred from commensals to pathogens by horizontal gene transfer, which magnifies the spectrum of antibiotic resistance in the environment. The presence of ARGs in neo-nates and infants, with no prior antibiotic exposure, questions their origin in the naïve commensal population. Breast milk microbiota that is responsible for the initial seeding of infant gut microbiota has also been found to harbour a vast array of ARGs. This review discusses the recent findings that indicate the potential of breast milk microbiota to act as a vehicle for transmission of ARGs to infants.
Collapse
Affiliation(s)
- Lahari Das
- Department of Zoology, University of Delhi, Delhi, 110007 India
| | - Richa Virmani
- Department of Zoology, University of Delhi, Delhi, 110007 India
| | - Vishal Sharma
- Department of Zoology, University of Delhi, Delhi, 110007 India
| | - Deepti Rawat
- Department of Zoology, University of Delhi, Delhi, 110007 India
| | - Yogendra Singh
- Department of Zoology, University of Delhi, Delhi, 110007 India
| |
Collapse
|
99
|
Persulfate activation for efficient degradation of norfloxacin by a rGO-Fe3O4 composite. J Taiwan Inst Chem Eng 2019. [DOI: 10.1016/j.jtice.2019.05.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
100
|
Cave R, Misra R, Chen J, Wang S, Mkrtchyan HV. Whole genome sequencing revealed new molecular characteristics in multidrug resistant staphylococci recovered from high frequency touched surfaces in London. Sci Rep 2019; 9:9637. [PMID: 31371820 PMCID: PMC6675788 DOI: 10.1038/s41598-019-45886-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 06/13/2019] [Indexed: 01/15/2023] Open
Abstract
The rise of antibiotic resistance (AMR) is one of the most important public health threats worldwide.Today, increasing attention is being paid to multidrug resistant staphylococci isolated from healthcare and non-healthcare environments as the treatment of these bacteria has become increasingly difficult. In this study, we compared staphylococci isolates recovered from high frequency touched surfaces from public areas in the community and hospitals in East and West London. 281 out of 600 (46.83%) staphylococci isolates recovered were multidrug resistant, of which 49 (8.17%) were mecA positive. There was significantly higher proportion of multidrug resistant staphylococci (P = 0.0002) in East London (56.7%) compared to West London (49.96%). The most common species identified as multidrug resistant were S. epidermidis, S. haemolyticus and S. hominis, whereas penicillin, fusidic acid and erythromycin were the most frequent antibiotics the isolates were resistant to. Whole genome sequenced of mecA positive isolates revealed that S. sciuri isolates carried the mecA1 gene, which has only 84.43% homology with mecA. In addition, other frequently identified resistance genes included blaZ, qacA/B and dfrC. We have also identified a diverse range of SCCmec types, many of which were untypable due to carrying a novel combination of ccr genes or multiple ccr complexes.
Collapse
Affiliation(s)
- Rory Cave
- University of East London, School of Health, Sport and Bioscience, Water Lane, London, E15 4LZ, UK
| | - Raju Misra
- Natural History Museum, Core Research Laboratories, Molecular Biology, Cromwell Rd, London, SW7 5BD, UK
| | - Jiazhen Chen
- Department of Infectious Disease, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Rd., Shanghai, 200040, China
| | - Shiyong Wang
- Department of Infectious Disease, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Rd., Shanghai, 200040, China
| | - Hermine V Mkrtchyan
- University of East London, School of Health, Sport and Bioscience, Water Lane, London, E15 4LZ, UK.
| |
Collapse
|