51
|
Haddad G, Fontanini A, Bellali S, Takakura T, Ominami Y, Hisada A, Hadjadj L, Rolain JM, Raoult D, Bou Khalil JY. Rapid Detection of Imipenem Resistance in Gram-Negative Bacteria Using Tabletop Scanning Electron Microscopy: A Preliminary Evaluation. Front Microbiol 2021; 12:658322. [PMID: 34220746 PMCID: PMC8245003 DOI: 10.3389/fmicb.2021.658322] [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: 01/25/2021] [Accepted: 05/18/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Enabling faster Antimicrobial Susceptibility Testing (AST) is critical, especially to detect antibiotic resistance, to provide rapid and appropriate therapy and to improve clinical outcomes. Although several standard and automated culture-based methods are available and widely used, these techniques take between 18 and 24 h to provide robust results. Faster techniques are needed to reduce the delay between test and results. Methods: Here we present a high throughput AST method using a new generation of tabletop scanning electron microscope, to evaluate bacterial ultra-structural modifications associated with susceptibilities to imipenem as a proof of concept. A total of 71 reference and clinical strains of Gram-negative bacteria were used to evaluate susceptibility toward imipenem after 30, 60, and 90 min of incubation. The length, width and electron density of bacteria were measured and compared between imipenem susceptible and resistant strains. Results: We correlated the presence of these morphological changes to the bacterial susceptibility and their absence to the bacterial resistance (e.g., Pseudomonas aeruginosa length without [2.24 ± 0.61 μm] and with [2.50 ± 0.68 μm] imipenem after 30 min [p = 3.032E-15]; Escherichia coli width without [0.92 ± 0.07 μm] and with [1.28 ± 0.19 μm] imipenem after 60 min [p = 1.242E-103]). We validated our method by a blind test on a series of 58 clinical isolates where all strains were correctly classified as susceptible or resistant toward imipenem. Conclusion: This method could be a potential tool for rapidly identifying carbapenem-resistance in Enterobacterales in clinical microbiology laboratories in <2 h, allowing the empirical treatment of patients to be rapidly adjusted.
Collapse
Affiliation(s)
- Gabriel Haddad
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France.,Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), UMR Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| | - Anthony Fontanini
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Sara Bellali
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Tatsuki Takakura
- Hitachi High-Tech Corporation, Analytical & Medical Solution Business Group, Ibaraki, Japan
| | - Yusuke Ominami
- Hitachi High-Tech Corporation, Nanotechnology Solutions Business Group, Toranomon Hills Business Tower, Tokyo, Japan
| | - Akiko Hisada
- Hitachi, Ltd., Research & Development Group, Tokyo, Japan
| | - Linda Hadjadj
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Jean-Marc Rolain
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France.,Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), UMR Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| | - Didier Raoult
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France.,Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), UMR Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France.,Hitachi High-Tech Corporation, Nanotechnology Solutions Business Group, Toranomon Hills Business Tower, Tokyo, Japan
| | - Jacques Yaacoub Bou Khalil
- Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France.,Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), UMR Microbes Evolution Phylogeny and Infections (MEPHI), Marseille, France
| |
Collapse
|
52
|
Gholap S, Yao C, Green O, Babjak M, Jakubec P, Malatinský T, Ihssen J, Wick L, Spitz U, Shabat D. Chemiluminescence Detection of Hydrogen Sulfide Release by β-Lactamase-Catalyzed β-Lactam Biodegradation: Unprecedented Pathway for Monitoring β-Lactam Antibiotic Bacterial Resistance. Bioconjug Chem 2021; 32:991-1000. [PMID: 33896185 PMCID: PMC8382227 DOI: 10.1021/acs.bioconjchem.1c00149] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/12/2021] [Indexed: 12/20/2022]
Abstract
β-Lactamase positive bacteria represent a growing threat to human health because of their resistance to commonly used antibiotics. Therefore, development of new diagnostic methods for identification of β-lactamase positive bacteria is of high importance for monitoring the spread of antibiotic-resistant bacteria. Here, we report the discovery of a new biodegradation metabolite (H2S), generated through β-lactamase-catalyzed hydrolysis of β-lactam antibiotics. This discovery directed us to develop a distinct molecular technique for monitoring bacterial antibiotic resistance. The technique is based on a highly efficient chemiluminescence probe, designed for detection of the metabolite, hydrogen sulfide, that is released upon biodegradation of β-lactam by β-lactamases. Such an assay can directly indicate if antibiotic bacterial resistance exists for a certain examined β-lactam. The assay was successfully demonstrated for five different β-lactam antibiotics and eight β-lactam resistant bacterial strains. Importantly, in a functional bacterial assay, our chemiluminescence probe was able to clearly distinguish between a β-lactam resistant bacterial strain and a sensitive one. As far as we know, there is no previous documentation for such a biodegradation pathway of β-lactam antibiotics. Bearing in mind the data obtained in this study, we propose that hydrogen sulfide should be considered as an emerging β-lactam metabolite for detection of bacterial resistance.
Collapse
Affiliation(s)
- Sachin
Popat Gholap
- School
of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 69978 Israel
| | - Chunyan Yao
- Biosynth
Carbosynth, Rietlistrasse
4 Postfach 125 9422 Staad, Switzerland
| | - Ori Green
- School
of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 69978 Israel
| | - Matej Babjak
- Department
of Organic Chemistry, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, 81237 Bratislava, Slovakia
| | - Pavol Jakubec
- Auchem
s.r.o., A. Hlinku 1452/3, 022 01 Čadca, Slovakia
| | | | - Julian Ihssen
- Biosynth
Carbosynth, Rietlistrasse
4 Postfach 125 9422 Staad, Switzerland
| | - Lukas Wick
- Biosynth
Carbosynth, Rietlistrasse
4 Postfach 125 9422 Staad, Switzerland
| | - Urs Spitz
- Biosynth
Carbosynth, Rietlistrasse
4 Postfach 125 9422 Staad, Switzerland
| | - Doron Shabat
- School
of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 69978 Israel
| |
Collapse
|
53
|
Rapid antimicrobial susceptibility testing by stimulated Raman scattering metabolic imaging and morphological deformation of bacteria. Anal Chim Acta 2021; 1168:338622. [PMID: 34051990 DOI: 10.1016/j.aca.2021.338622] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/16/2021] [Accepted: 05/03/2021] [Indexed: 12/19/2022]
Abstract
Methods for rapid antimicrobial susceptibility testing (AST) are urgently needed to address the emergence and spread of antimicrobial resistance. Here, we report a new method based on stimulated Raman scattering (SRS) microscopy, which measures both the metabolic activity and the morphological deformation of bacteria to determine the antimicrobial susceptibility of β-lactam antibiotics rapidly. In this approach, we quantify single bacteria's metabolic activity by the carbon-deuterium (C-D) bond concentrations in bacteria after D2O incubation. In the meantime, bacterial morphological deformation caused by β-lactam antibiotics is also measured. With these two quantifiable markers, we develop an evaluation method to perform AST of cefotaxime on 103 E. coli strains. Our method achieved a 93.2% categorical agreement and a 93.2% essential agreement with the standard reference method.
Collapse
|
54
|
Robertson J, McGoverin C, White JR, Vanholsbeeck F, Swift S. Rapid Detection of Escherichia coli Antibiotic Susceptibility Using Live/Dead Spectrometry for Lytic Agents. Microorganisms 2021; 9:924. [PMID: 33925816 PMCID: PMC8147107 DOI: 10.3390/microorganisms9050924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
Antibiotic resistance is a serious threat to public health. The empiric use of the wrong antibiotic occurs due to urgency in treatment combined with slow, culture-based diagnostic techniques. Inappropriate antibiotic choice can promote the development of antibiotic resistance. We investigated live/dead spectrometry using a fluorimeter (Optrode) as a rapid alternative to culture-based techniques through application of the LIVE/DEAD® BacLightTM Bacterial Viability Kit. Killing was detected by the Optrode in near real-time when Escherichia coli was treated with lytic antibiotics-ampicillin and polymyxin B-and stained with SYTO 9 and/or propidium iodide. Antibiotic concentration, bacterial growth phase, and treatment time used affected the efficacy of this detection method. Quantification methods of the lethal action and inhibitory action of the non-lytic antibiotics, ciprofloxacin and chloramphenicol, respectively, remain to be elucidated.
Collapse
Affiliation(s)
- Julia Robertson
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland 1023, New Zealand; (J.R.W.); (S.S.)
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Auckland 1010, New Zealand; (C.M.); (F.V.)
| | - Cushla McGoverin
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Auckland 1010, New Zealand; (C.M.); (F.V.)
- Department of Physics, The University of Auckland, Auckland 1010, New Zealand
| | - Joni R. White
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland 1023, New Zealand; (J.R.W.); (S.S.)
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Auckland 1010, New Zealand; (C.M.); (F.V.)
| | - Frédérique Vanholsbeeck
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Auckland 1010, New Zealand; (C.M.); (F.V.)
- Department of Physics, The University of Auckland, Auckland 1010, New Zealand
| | - Simon Swift
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland 1023, New Zealand; (J.R.W.); (S.S.)
| |
Collapse
|
55
|
Swami P, Sharma A, Anand S, Gupta S. DEPIS: A combined dielectrophoresis and impedance spectroscopy platform for rapid cell viability and antimicrobial susceptibility analysis. Biosens Bioelectron 2021; 182:113190. [PMID: 33866070 DOI: 10.1016/j.bios.2021.113190] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/11/2021] [Accepted: 03/18/2021] [Indexed: 10/21/2022]
Abstract
Antimicrobial resistance (AMR) is caused by inappropriate or excessive antibiotic consumption. Early diagnosis of bacterial infections can greatly curb empirical treatment and thus AMR. Current diagnostic procedures are time-consuming as they rely on gene amplification and cell culture techniques that are inherently limited by the doubling rate of the involved species. Further, biochemical methods for species identification and antibiotic susceptibility testing for drug/dose effectiveness take several days and are non-scalable. We report a real-time, label-free approach called DEPIS that combines dielectrophoresis (DEP) for bacterial enrichment and impedance spectroscopy (IS) for cell viability analysis under 60 min. Target bacteria are captured on interdigitated electrodes using DEP (30 min) and their antibiotic-induced stress response is measured using IS (another 30 min). This principle is used to generate minimum bactericidal concentration (MBC) plots by measuring impedance change due to ionic release by dying bacteria in a low conductivity buffer. The results are rapid since they rely on cell death rather than cell growth which is an intrinsically slower process. The results are also highly specific and work across all bactericidal antibiotics studied, irrespective of their cellular target or drug action mechanism. More importantly, preliminary results with clinical isolates show that methicillin-susceptible Staphylococcus aureus (MSSA) can easily be differentiated from methicillin-resistant S. aureus (MRSA) under 1 h. This rapid cell analyses approach can aid in faster diagnosis of bacterial infections and benefit the clinical decision-making process for antibiotic treatment, addressing the critical issue of AMR.
Collapse
Affiliation(s)
- Pragya Swami
- Dept. of Chemical Engineering, Indian Institute of Technology, Delhi, 110016, India
| | - Ayush Sharma
- Dept. of Chemical Engineering, Indian Institute of Technology, Delhi, 110016, India
| | - Satyam Anand
- Dept. of Chemical Engineering, Indian Institute of Technology, Delhi, 110016, India
| | - Shalini Gupta
- Dept. of Chemical Engineering, Indian Institute of Technology, Delhi, 110016, India.
| |
Collapse
|
56
|
Laure NN, Ahn J. Development of phage-based assay to differentiate ciprofloxacin resistant and sensitive Salmonella Typhimurium. Food Sci Biotechnol 2021; 30:315-320. [PMID: 33732522 DOI: 10.1007/s10068-020-00858-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/20/2020] [Accepted: 11/27/2020] [Indexed: 10/22/2022] Open
Abstract
This study was designed to evaluate the possibility of using phage-amplification assay for discriminating between antibiotic-sensitive and antibiotic-resistant Salmonella Typhimurium. The characteristics of Salmonella phage PBST32 were determined by adsorption rate, one-step growth curve, and lytic activity. The ability of phage-based method to detect S. Typhimurium ATCC 19585 (STCIP) was determined in single culture and bacterial mixtures of S. Typhimurium ATCC 19585 (STWT), Klebsiella pneumoniae, and Staphylococcus aureus. The adsorption rates of PBST32 were 95% and 93% against STWT and STCIP after 20 min, respectively. The PBST32 showed latent period of 20 min and average burst size of 90 against STWT and STCIP. The STCIP was selectively detected in mixtures of S. aureus, K. pneumoniae, and STWT by phage amplification assay. These results provide useful information for designing phage amplification method that can differentially detect antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Nana Nguefang Laure
- Department of Biomedical Science and Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, Gangwon 24341 Republic of Korea
| | - Juhee Ahn
- Department of Biomedical Science and Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, Gangwon 24341 Republic of Korea
| |
Collapse
|
57
|
Recent Development of Rapid Antimicrobial Susceptibility Testing Methods through Metabolic Profiling of Bacteria. Antibiotics (Basel) 2021; 10:antibiotics10030311. [PMID: 33803002 PMCID: PMC8002737 DOI: 10.3390/antibiotics10030311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 11/17/2022] Open
Abstract
Due to the inappropriate use and overuse of antibiotics, the emergence and spread of antibiotic-resistant bacteria are increasing and have become a major threat to human health. A key factor in the treatment of bacterial infections and slowing down the emergence of antibiotic resistance is to perform antimicrobial susceptibility testing (AST) of infecting bacteria rapidly to prescribe appropriate drugs and reduce the use of broad-spectrum antibiotics. Current phenotypic AST methods based on the detection of bacterial growth are generally reliable but are too slow. There is an urgent need for new methods that can perform AST rapidly. Bacterial metabolism is a fast process, as bacterial cells double about every 20 to 30 min for fast-growing species. Moreover, bacterial metabolism has shown to be related to drug resistance, so a comparison of differences in microbial metabolic processes in the presence or absence of antimicrobials provides an alternative approach to traditional culture for faster AST. In this review, we summarize recent developments in rapid AST methods through metabolic profiling of bacteria under antibiotic treatment.
Collapse
|
58
|
Kasas S, Malovichko A, Villalba MI, Vela ME, Yantorno O, Willaert RG. Nanomotion Detection-Based Rapid Antibiotic Susceptibility Testing. Antibiotics (Basel) 2021; 10:287. [PMID: 33801939 PMCID: PMC7999052 DOI: 10.3390/antibiotics10030287] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 02/26/2021] [Accepted: 03/07/2021] [Indexed: 01/04/2023] Open
Abstract
Rapid antibiotic susceptibility testing (AST) could play a major role in fighting multidrug-resistant bacteria. Recently, it was discovered that all living organisms oscillate in the range of nanometers and that these oscillations, referred to as nanomotion, stop as soon the organism dies. This finding led to the development of rapid AST techniques based on the monitoring of these oscillations upon exposure to antibiotics. In this review, we explain the working principle of this novel technique, compare the method with current ASTs, explore its application and give some advice about its implementation. As an illustrative example, we present the application of the technique to the slowly growing and pathogenic Bordetella pertussis bacteria.
Collapse
Affiliation(s)
- Sandor Kasas
- Laboratory of Biological Electron Microscopy, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; (A.M.); (M.I.V.)
- Unité Facultaire d’Anatomie et de Morphologie (UFAM), CUMRL, University of Lausanne, 1005 Lausanne, Switzerland
- International Joint Research Group VUB-EPFL NanoBiotechnology and NanoMedicine (NANO), Vrije Universiteit Brussel, 1050 Brussels, Belgium;
| | - Anton Malovichko
- Laboratory of Biological Electron Microscopy, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; (A.M.); (M.I.V.)
- International Joint Research Group VUB-EPFL NanoBiotechnology and NanoMedicine (NANO), Vrije Universiteit Brussel, 1050 Brussels, Belgium;
| | - Maria Ines Villalba
- Laboratory of Biological Electron Microscopy, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; (A.M.); (M.I.V.)
- International Joint Research Group VUB-EPFL NanoBiotechnology and NanoMedicine (NANO), Vrije Universiteit Brussel, 1050 Brussels, Belgium;
| | - María Elena Vela
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, and CONICET, Diagonal 113 y 64, 1900 La Plata, Argentina;
| | - Osvaldo Yantorno
- Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI-CONICET-CCT La Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 1900 La Plata, Argentina;
| | - Ronnie G. Willaert
- International Joint Research Group VUB-EPFL NanoBiotechnology and NanoMedicine (NANO), Vrije Universiteit Brussel, 1050 Brussels, Belgium;
- Research Group Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| |
Collapse
|
59
|
Emerging trends in aggregation induced emissive luminogens as bacterial theranostics. J Drug Target 2021; 29:793-807. [PMID: 33583291 DOI: 10.1080/1061186x.2021.1888111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The emergence and spread of pathogenic bacteria, particularly antibiotic-resistant strains pose grave global concerns worldwide, which demand for the rapid development of highly selective and sensitive strategies for specific bacterial detection, identification, imaging and therapy. The fascinating feature of aggregation-induced emissive molecules (AIEgens) to display fluorescence in aggregate form can be suitably coupled with nanotechnology for developing theranostic AIE dots that can offer convenient and customised functions such as sensing, imaging, detection, discrimination and cell kill of different bacterial types. The initial section of the article reveals the necessity for incorporating diagnostic imaging with antibacterial therapy, while the latter part delivers mechanistic insights on the benefits of AIE fluorophores in theranostic applications. Further, the review illustrates the recent advancements of AIEgens as theranostic nanolights in bacterial detection, identification and eradication. The review is organised according to the different classes of AIE-active bacterial theranostics such as carrier-free nanoprodrugs, nanomachines for synergistic imaging-guided cancer treatment and bacterial kill, AIE polymers, bioconjugates and nanoparticle carriers. By elucidating their design principles and applications, as well as highlighting the recent trends and perspectives that can be further explored, we hope to instill more research interest in AIE bacterial theranostics for future translational research.HighlightsCombination of aggregation induced emissive fluorophores and nanotechnology for developing bacterial theranostics.AIE theranostics with customised functions for bacterial imaging, detection, discrimination and cell kill.
Collapse
|
60
|
Clinical Implications of Polymicrobial Synergism Effects on Antimicrobial Susceptibility. Pathogens 2021; 10:pathogens10020144. [PMID: 33535562 PMCID: PMC7912749 DOI: 10.3390/pathogens10020144] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/14/2021] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
With the development of next generation sequencing technologies in recent years, it has been demonstrated that many human infectious processes, including chronic wounds, cystic fibrosis, and otitis media, are associated with a polymicrobial burden. Research has also demonstrated that polymicrobial infections tend to be associated with treatment failure and worse patient prognoses. Despite the importance of the polymicrobial nature of many infection states, the current clinical standard for determining antimicrobial susceptibility in the clinical laboratory is exclusively performed on unimicrobial suspensions. There is a growing body of research demonstrating that microorganisms in a polymicrobial environment can synergize their activities associated with a variety of outcomes, including changes to their antimicrobial susceptibility through both resistance and tolerance mechanisms. This review highlights the current body of work describing polymicrobial synergism, both inter- and intra-kingdom, impacting antimicrobial susceptibility. Given the importance of polymicrobial synergism in the clinical environment, a new system of determining antimicrobial susceptibility from polymicrobial infections may significantly impact patient treatment and outcomes.
Collapse
|
61
|
Sheka D, Alabi N, Gordon PMK. Oxford nanopore sequencing in clinical microbiology and infection diagnostics. Brief Bioinform 2021; 22:6109725. [PMID: 33483726 DOI: 10.1093/bib/bbaa403] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/26/2020] [Accepted: 12/09/2020] [Indexed: 12/16/2022] Open
Abstract
Extended turnaround times and large economic costs hinder the usage of currently applied screening methods for bacterial pathogen identification (ID) and antimicrobial susceptibility testing. This review provides an overview of current detection methods and their usage in a clinical setting. Issues of timeliness and cost could soon be circumvented, however, with the emergence of detection methods involving single molecule sequencing technology. In the context of bringing diagnostics closer to the point of care, we examine the current state of Oxford Nanopore Technologies (ONT) products and their interaction with third-party software/databases to assess their capabilities for ID and antimicrobial resistance (AMR) prediction. We outline and discuss a potential diagnostic workflow, enumerating (1) rapid sample prep kits, (2) ONT hardware/software and (3) third-party software and databases to improve the cost, accuracy and turnaround times for ID and AMR. Multiple studies across a range of infection types support that the speed and accuracy of ONT sequencing is now such that established ID and AMR prediction tools can be used on its outputs, and so it can be harnessed for near real time, close to the point-of-care diagnostics in common clinical circumstances.
Collapse
Affiliation(s)
- Dropen Sheka
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nikolay Alabi
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul M K Gordon
- Cumming School of Medicine Centre for Health Genomics and Informatics, University of Calgary
| |
Collapse
|
62
|
Jakobsen TH, Xu Y, Bay L, Schønheyder HC, Jakobsen T, Bjarnsholt T, Thomsen TR. Sampling challenges in diagnosis of chronic bacterial infections. J Med Microbiol 2021; 70. [PMID: 33410733 DOI: 10.1099/jmm.0.001302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In recent decades there has been an increase in knowledge of the distribution, species diversity and growth patterns of bacteria in human chronic infections. This has challenged standard diagnostic methods, which have undergone a development to both increase the accuracy of testing as well as to decrease the occurrence of contamination. In particular, the introduction of new technologies based on molecular techniques into the clinical diagnostic process has increased detection and identification of infectious pathogens. Sampling is the first step in the diagnostic process, making it crucial for obtaining a successful outcome. However, sampling methods have not developed at the same speed as molecular identification. The heterogeneous distribution and potentially small number of pathogenic bacterial cells in chronic infected tissue makes sampling a complicated task, and samples must be collected judiciously and handled with care. Clinical sampling is a step in the diagnostic process that may benefit from innovative methods based on current knowledge of bacteria present in chronic infections. In the present review, we describe and discuss different aspects that complicate sampling of chronic infections. The purpose is to survey representative scientific work investigating the presence and distribution of bacteria in chronic infections in relation to various clinical sampling methods.
Collapse
Affiliation(s)
- Tim Holm Jakobsen
- Costerton Biofilm Center, Institute for Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Yijuan Xu
- Bio- and Environmental Technology, Danish Technological Institute, Taastrup, Denmark
- Center for Microbial Communities, Department of Chemistry and Biosciences, Aalborg University, Aalborg, Denmark
| | - Lene Bay
- Costerton Biofilm Center, Institute for Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Carl Schønheyder
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Clinical Microbiology, Aalborg University Hospital, Aalborg, Denmark
| | - Thomas Jakobsen
- Department of Orthopaedics, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Thomas Bjarnsholt
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Costerton Biofilm Center, Institute for Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Trine Rolighed Thomsen
- Bio- and Environmental Technology, Danish Technological Institute, Taastrup, Denmark
- Center for Microbial Communities, Department of Chemistry and Biosciences, Aalborg University, Aalborg, Denmark
| |
Collapse
|
63
|
Lim T, Kim EG, Choi J, Kwon S. A high-throughput cell culture system based on capillary and centrifugal actions for rapid antimicrobial susceptibility testing. LAB ON A CHIP 2020; 20:4552-4560. [PMID: 33169754 DOI: 10.1039/d0lc00753f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Antibiotic resistance is a global threat to modern society. Rapid determination of suitable antibiotics that inhibit bacterial growth can effectively reduce antibiotic resistance and improve clinical treatment. The conventional methods of antimicrobial susceptibility testing (AST) depend on optical density measurements, which require long-time incubation. Various kinds of rapid AST systems which utilize various technologies from the field of lab on a chip have promised a great reduction in measurement time, but cannot achieve high-throughput, user-friendly testing due to the complexity of the testing system. Here, we introduce a capillary and centrifuge-based rapid AST system that reduces the time of loading the sample and culture media while achieving a high-throughput testing capacity. The capability of the proposed system is validated in a systematic analysis that includes sample loading characteristics and AST trials with standard strains. The proposed system provides a useful tool for drug testing in cell-culture systems with user-friendly and high-throughput analysis.
Collapse
Affiliation(s)
- Taegeun Lim
- QuantaMatrix Inc., Seoul National University Hospital CMI, Seoul, 03082, Republic of Korea and Department of Electrical Engineering and Computer Science, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Eun-Geun Kim
- Lowendtechnologies Co., Anyang, 14056, Republic of Korea
| | - Jungil Choi
- School of Mechanical Engineering, Kookmin University, Seoul, 02707, Republic of Korea.
| | - Sunghoon Kwon
- QuantaMatrix Inc., Seoul National University Hospital CMI, Seoul, 03082, Republic of Korea and Department of Electrical Engineering and Computer Science, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
64
|
Affordable automated phenotypic antibiotic susceptibility testing method based on a contactless conductometric sensor. Sci Rep 2020; 10:21216. [PMID: 33277561 PMCID: PMC7718250 DOI: 10.1038/s41598-020-77938-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/11/2020] [Indexed: 11/08/2022] Open
Abstract
User-friendly phenotypic antibiotic susceptibility testing (AST) methods are urgently needed in many fields including clinical medicine, epidemiological studies and drug research. Herein, we report a convenient and cost-effective phenotypic AST method based on online monitoring bacterial growth with a developed 8-channel contactless conductometric sensor (CCS). Using E. coli and V. parahaemolyticus as microorganism models, as well as enoxacin, florfenicol, ampicillin, kanamycin and sulfadiazine as antibiotic probes. The minimum inhibitory concentration (MIC) determination was validated in comparison with standard broth microdilution (BMD) assay. The total essential agreements between the CCS AST assays and the reference BMD AST assays are 68.8–92.3%. The CCS has an approximate price of $9,000 (USD). Requiring neither chemical nor biotic auxiliary materials for the assay makes the cost of each sample < $1. The MICs obtained with the automated CCS AST assays are more precise than those obtained with the manual BMD. Moreover, in 72 percent of the counterpart, the MICs obtained with the CCS AST assays are higher than that obtained with the BMD AST assays. The proposed CCS AST method has advantages in affordability, accuracy, sensitivity and user-friendliness.
Collapse
|
65
|
Genthe B, Ndlela L, Madlala T. Antimicrobial resistance screening and profiles: a glimpse from the South African perspective. JOURNAL OF WATER AND HEALTH 2020; 18:925-936. [PMID: 33328364 DOI: 10.2166/wh.2020.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
According to the Centre for Disease Dynamics Economics and Policy, South Africa represents a paradox of antibiotic management similar to other developing countries, with both overuse and underuse (resulting from lack of access) of antibiotics. In addition, wastewater reuse may contribute towards antibiotic resistance through selective pressure that increases resistance in native bacteria and on clinically relevant bacteria, increasing resistance profiles of the common pathogens. Sediments of surface water bodies and wastewater sludge provide a place where antibiotic resistance genes are transferred to other bacteria. Crop irrigation is thought to be a potential source of exposure to antibiotic-resistant bacteria through the transfer from the water or sludge into crops. The objectives of this study were to examine the antibiotic-resistance profiles of Escherishia coli from three agricultural locations in the Western Cape, South Africa. Using a classical microbiology culture approach, the resistance profiles of E. coli species isolated from river water and sediments, farm dams and their sediments and a passive algal wastewater treatment ponds and sediment used for crop irrigation were assessed for resistance to 13 commonly used antibiotics. Randomly selected E. coli isolates from the sediment and water were tested for resistance. 100% of E. coli isolates were resistant to sulphamethoxazole, highlighting its relevance in the South African context. In river water and farm dam samples, only the E. coli isolated from sediment were found to be resistant to fluoroquinolone or fluorifenicol. In the wastewater treatment ponds, the resistance profiles of E. coli isolated from sediments differed from those isolated from effluent, with 90% of the effluent isolates being resistant to ampicillin. Isolates from the sediment were less resistant (40%) to ampicillin, whereas all the isolates from the pond water and sediment samples were resistant to sulphamethoxazole. These results illustrate the importance of developing a better understanding of antibiotic resistance in agriculture and wastewater scenarios to ensure remedial measures take place where the greatest benefit can be realised especially in countries with limited financial and infrastructural resources. Moreover, the potential for passive algal treatment as an effective, feasible alternative for wastewater treatment is highlighted, with comparable resistance profiles and a reducing overall resistance in the sediment samples.
Collapse
Affiliation(s)
- B Genthe
- Water Centre, Smart Places, CSIR, P.O. Box 320, 11 Jan Celliers Road, Stellenbosch 7599, South Africa E-mail:
| | - L Ndlela
- Water Centre, Smart Places, CSIR, P.O. Box 320, 11 Jan Celliers Road, Stellenbosch 7599, South Africa E-mail:
| | - T Madlala
- Water Centre, Smart Places, CSIR, P.O. Box 320, 11 Jan Celliers Road, Stellenbosch 7599, South Africa E-mail: ; Department of Earth Science, University of Western Cape, Private Bag X17, Bellville 7535, South Africa
| |
Collapse
|
66
|
Zhang M, Hong W, Abutaleb NS, Li J, Dong P, Zong C, Wang P, Seleem MN, Cheng J. Rapid Determination of Antimicrobial Susceptibility by Stimulated Raman Scattering Imaging of D 2O Metabolic Incorporation in a Single Bacterium. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001452. [PMID: 33042757 PMCID: PMC7539191 DOI: 10.1002/advs.202001452] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/24/2020] [Indexed: 05/27/2023]
Abstract
Rapid antimicrobial susceptibility testing (AST) is urgently needed for treating infections with appropriate antibiotics and slowing down the emergence of antibiotic-resistant bacteria. Here, a phenotypic platform that rapidly produces AST results by femtosecond stimulated Raman scattering imaging of deuterium oxide (D2O) metabolism is reported. Metabolic incorporation of D2O into biomass in a single bacterium and the metabolic response to antibiotics are probed in as short as 10 min after culture in 70% D2O medium, the fastest among current technologies. Single-cell metabolism inactivation concentration (SC-MIC) is obtained in less than 2.5 h from colony to results. The SC-MIC results of 37 sets of bacterial isolate samples, which include 8 major bacterial species and 14 different antibiotics often encountered in clinic, are validated by standard minimal inhibitory concentration blindly measured via broth microdilution. Toward clinical translation, stimulated Raman scattering imaging of D2O metabolic incorporation and SC-MIC determination after 1 h antibiotic treatment and 30 min mixture of D2O and antibiotics incubation of bacteria in urine or whole blood is demonstrated.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Electrical and Computer EngineeringBoston UniversityBostonMA02215USA
- Boston University Photonics CenterBostonMA02215USA
| | - Weili Hong
- Department of Electrical and Computer EngineeringBoston UniversityBostonMA02215USA
| | - Nader S. Abutaleb
- Department of Comparative PathobiologyPurdue UniversityWest LafayetteIN47907USA
| | - Junjie Li
- Department of Electrical and Computer EngineeringBoston UniversityBostonMA02215USA
- Boston University Photonics CenterBostonMA02215USA
| | - Pu‐Ting Dong
- Boston University Photonics CenterBostonMA02215USA
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
| | - Cheng Zong
- Department of Electrical and Computer EngineeringBoston UniversityBostonMA02215USA
- Boston University Photonics CenterBostonMA02215USA
| | - Pu Wang
- Vibronix Inc.West LafayetteIN47906USA
| | - Mohamed N. Seleem
- Department of Comparative PathobiologyPurdue UniversityWest LafayetteIN47907USA
- Purdue Institute of InflammationImmunology, and Infectious DiseaseWest LafayetteIN47907USA
| | - Ji‐Xin Cheng
- Department of Electrical and Computer EngineeringBoston UniversityBostonMA02215USA
- Boston University Photonics CenterBostonMA02215USA
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Department of ChemistryBoston UniversityBostonMA02215USA
| |
Collapse
|
67
|
Likhachev IV, Kraeva LA, Samoilova AA, Rogacheva EV, Kaftyreva LA, Egorova SA, Mikhailov NV. Approbation of russian test strips for antimicrobial susceptibility testing of microorganisms by gradient diffusion method (E-test). Klin Lab Diagn 2020; 65:557-561. [PMID: 33245641 DOI: 10.18821/0869-2084-2020-65-9-557-561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The most reliable criterion for the microorganisms antibiotic susceptibility is the value of the minimum inhibitory concentration (MIC). The gradient diffusion method (epsilometric test, e-test), carried out using test strips impregnated with an antimicrobial agent, allows to obtain the quantitative value of MIC, bypassing the time-consuming steps of the traditional method of serial dilutions. We tested strips for the epsilometric test, developed at Saint-Petersburg Pasteur Institute. The quality control, carried by testing the reference strains of E. coli ATCC 25922 and S. aureus 29213, confirmed compliance of the manufacturer's declared antibiotic concentration ranges. The MIC values obtained in the study of the antibiotic susceptibility of 101 clinical isolates of microorganisms of the ESKAPE group, isolated from patients of various hospitals in St. Petersburg, were compared to 8 antimicrobial agents using tested test strips and the corresponding M.I.C. Evaluator of the Oxoid (UK). A high percentage of compliance of MIC values was demonstrated, as well as full compliance of susceptibility categories for all microorganism/antibiotic combinations. The resulting divergences in the MIC values did not exceed one step of a double serial dilution, which is permissible according to GOST R ISO 20776-1-2010. The study showed that the test strips developed at the Saint-Petersburg Pasteur Institute DNT are an acceptable alternative to import tests and, after the registration certificate will be completed, can be offered to determine the susceptibility of microorganisms to antimicrobial agents in bacteriological laboratories.
Collapse
Affiliation(s)
| | | | | | | | - L A Kaftyreva
- St. Petersburg Pasteur Institute.,State Educational Institution of the Higher Professional Education "North-Western state medical University n.a. I.I. Mechnikov" of the Ministry of Health of the Russian Federation
| | | | | |
Collapse
|
68
|
Verma P, Anand S. Antimicrobial activity as a potential factor influencing the predominance of Bacillus subtilis within the constitutive microflora of a whey reverse osmosis membrane biofilm. J Dairy Sci 2020; 103:9992-10000. [PMID: 32921470 DOI: 10.3168/jds.2020-18576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 06/30/2020] [Indexed: 11/19/2022]
Abstract
Current cleaning and sanitation protocols may not be adequately effective in cleaning separation membranes and can result in the formation of resilient multispecies biofilms. The matured biofilms may result in a bacterial predominance with resilient strains on membranes with a prolonged use. In our previous study, we isolated organisms such as Bacillus subtilis, Bacillus licheniformis, Exiguobacterium aurantiacum, and Acinetobacter radioresistens from an 18-mo-old reverse osmosis membrane. The competitive exclusion studies revealed the predominance of B. subtilis within the membrane biofilm microflora. This study investigated the antimicrobial activity of the B. subtilis isolate as a potential cause of its predominance. The culture isolate was propagated in tryptic soy broth at 37°C, and microfiltered to prepare cell-free extracts (CFE) at 8-, 10-, 12-, 14-, 16-, and 18-h intervals. The CFE were freeze-dried and suspended in minimum quantities of HPLC-grade water to prepare concentrated solutions. The antimicrobial activities of CFE were tested using the agar-well assay against the biofilm constitutive microflora. The experiments were conducted in triplicates and means were compared for significant differences using a general linear mixed model procedure. The results indicated the highest antimicrobial activity of 12-h CFE of B. subtilis against other constitutive microflora such as Exiguobacterium sp., E. auranticum, and A. radioresistens, with average inhibition zone sizes of 16.5 ± 0.00, 16.25 ± 0.66, and 20.6 ± 0.00 mm, respectively. Upon treatment with proteinase K, the CFE completely lost its antimicrobial activity, establishing it to be a proteinaceous compound. The AA profiling revealed the total crude protein in CFE to be 51% (wt/wt), with its major constituent as glutamic acid (11.30% wt/wt). The freeze-dried CFE was thermally stable on exposure to the common temperature used for sanitizer applications (23.8°C for 5 and 10 min) and over a pH range of 3.0 to 6.3. The study helped us understand the role of the antimicrobial compound produced by B. subtilis as a potential cause of its predominance within the biofilm constitutive microflora.
Collapse
Affiliation(s)
- Pratishtha Verma
- Midwest Dairy Foods Research Center, Dairy and Food Science Department, South Dakota State University, Brookings 57007
| | - Sanjeev Anand
- Midwest Dairy Foods Research Center, Dairy and Food Science Department, South Dakota State University, Brookings 57007.
| |
Collapse
|
69
|
Bolotsky A, Muralidharan R, Butler D, Root K, Murray W, Liu Z, Ebrahimi A. Organic redox-active crystalline layers for reagent-free electrochemical antibiotic susceptibility testing (ORACLE-AST). Biosens Bioelectron 2020; 172:112615. [PMID: 33166804 DOI: 10.1016/j.bios.2020.112615] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/11/2020] [Accepted: 09/13/2020] [Indexed: 12/21/2022]
Abstract
Rapid antibiotic susceptibility testing (AST) is critical in determining bacterial resistance or susceptibility to a particular antibiotic. Simple-to-use phenotype-based AST platforms can assist care-givers in timely prescription of the right antibiotic. Monitoring the change of bacterial viability by measuring electrochemical Faradaic current is a promising approach for rapid AST. However, the existing works require mixing redox-active reagents in the solution which can interfere with the antibiotics. In this paper, we developed a facile electrodeposition process for creating a redox-active crystalline layer (denoted as RZx) on pyrolytic graphite sheets (PGS), which was then utilized as the sensing layer for reagent-free electrochemical AST. To demonstrate the proof-of-principle, we tested the sensors with Escherichia coli (E. coli) K-12 treated with two antibiotics, ampicillin and kanamycin. While the sensors enable detection of bacterial metabolism mainly due to pH-sensitivity of RZx (∼ 53 mV/pH), secreted redox-active metabolites/compounds from whole cells are likely contributing to the signal as well. By monitoring the differential voltammetric signals, the sensors enable accurate prediction of the minimum inhibitory concentration (MIC) in 60 min (p < 0.03). The sensors are stable after 60 days storage in ambient conditions and enable analysis of microbial viability in complex solutions, as demonstrated in spiked milk and human whole blood.
Collapse
Affiliation(s)
- Adam Bolotsky
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, PA, 16802, USA; Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Ritvik Muralidharan
- School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Derrick Butler
- Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, USA; School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Kayla Root
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - William Murray
- Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, USA; School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Zhiwen Liu
- Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, USA; School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Aida Ebrahimi
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, PA, 16802, USA; Materials Research Institute, The Pennsylvania State University, University Park, PA, 16802, USA; School of Electrical Engineering and Computer Science, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
70
|
Liu Y, Jia Y, Yang K, Tong Z, Shi J, Li R, Xiao X, Ren W, Hardeland R, Reiter RJ, Wang Z. Melatonin overcomes MCR-mediated colistin resistance in Gram-negative pathogens. Am J Cancer Res 2020; 10:10697-10711. [PMID: 32929375 PMCID: PMC7482817 DOI: 10.7150/thno.45951] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Emergence, prevalence and widely spread of plasmid-mediated colistin resistance in Enterobacteriaceae strongly impairs the clinical efficacy of colistin against life-threatening bacterial infections. Combinations of antibiotics and FDA-approved non-antibiotic agents represent a promising means to address the widespread emergence of antibiotic-resistant pathogens. Methods: Herein, we investigated the synergistic activity between melatonin and antibiotics against MCR (mobilized colistin resistance)-positive Gram-negative pathogens through checkerboard assay and time-killing curve. Molecular mechanisms underlying its mode of action were elucidated. Finally, we assessed the in vivo efficacy of melatonin in combination with colistin against drug-resistant Gram-negative bacteria. Results: Melatonin, which has been approved for treating sleep disturbances and circadian disorders, substantially potentiates the activity of three antibiotics, particularly colistin, against MCR-expressing pathogens without enhancing its toxicity. This is evidence that the combination of colistin with melatonin enhances bacterial outer membrane permeability, promotes oxidative damage and inhibits the effect of efflux pumps. In three animal models infected by mcr-1-carrying E. coli, melatonin dramatically rescues colistin efficacy. Conclusion: Our findings revealed that melatonin serves as a promising colistin adjuvant against MCR-positive Gram-negative pathogens.
Collapse
|
71
|
Reporter Phage-Based Detection of Bacterial Pathogens: Design Guidelines and Recent Developments. Viruses 2020; 12:v12090944. [PMID: 32858938 PMCID: PMC7552063 DOI: 10.3390/v12090944] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/10/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
Fast and reliable detection of bacterial pathogens in clinical samples, contaminated food products, and water supplies can drastically improve clinical outcomes and reduce the socio-economic impact of disease. As natural predators of bacteria, bacteriophages (phages) have evolved to bind their hosts with unparalleled specificity and to rapidly deliver and replicate their viral genome. Not surprisingly, phages and phage-encoded proteins have been used to develop a vast repertoire of diagnostic assays, many of which outperform conventional culture-based and molecular detection methods. While intact phages or phage-encoded affinity proteins can be used to capture bacteria, most phage-inspired detection systems harness viral genome delivery and amplification: to this end, suitable phages are genetically reprogrammed to deliver heterologous reporter genes, whose activity is typically detected through enzymatic substrate conversion to indicate the presence of a viable host cell. Infection with such engineered reporter phages typically leads to a rapid burst of reporter protein production that enables highly sensitive detection. In this review, we highlight recent advances in infection-based detection methods, present guidelines for reporter phage construction, outline technical aspects of reporter phage engineering, and discuss some of the advantages and pitfalls of phage-based pathogen detection. Recent improvements in reporter phage construction and engineering further substantiate the potential of these highly evolved nanomachines as rapid and inexpensive detection systems to replace or complement traditional diagnostic approaches.
Collapse
|
72
|
Lee KS, Lee SM, Oh J, Park IH, Song JH, Han M, Yong D, Lim KJ, Shin JS, Yoo KH. Electrical antimicrobial susceptibility testing based on aptamer-functionalized capacitance sensor array for clinical isolates. Sci Rep 2020; 10:13709. [PMID: 32792573 PMCID: PMC7426404 DOI: 10.1038/s41598-020-70459-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/06/2020] [Indexed: 01/16/2023] Open
Abstract
To prescribe effective antibiotics to patients with bacterial infections in a timely manner and to avoid the misuse of antibiotics, a rapid antimicrobial susceptibility test (AST) is essential. However, conventional AST methods require more than 16 h to provide results; thus, we developed an electrical AST (e-AST) system, which provides results within 6 h. The proposed e-AST is based on an array of 60 aptamer-functionalized capacitance sensors that are comparable to currently available AST panels and a pattern-matching algorithm. The performance of the e-AST was evaluated in comparison with that of broth microdilution as the reference test for clinical strains isolated from septic patients. A total of 4,554 tests using e-AST showed a categorical agreement of 97% with a minor error of 2.2%, major error of 0.38%, and very major error of 0.38%. We expect that the proposed e-AST could potentially aid antimicrobial stewardship efforts and lead to improved patient outcomes.
Collapse
Affiliation(s)
- Kyo-Seok Lee
- Department of Physics, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sun-Mi Lee
- Nanomedical Graduate Program, Yonsei University, Seoul, 03722, Republic of Korea.
| | - Jeseung Oh
- Proteomtech Inc., 1101 Wooree-Venture Town, Seoul, 07573, Republic of Korea
| | - In Ho Park
- Department of Microbiology, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute and Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jun Ho Song
- Department of Physics, Yonsei University, Seoul, 03722, Republic of Korea
| | - Myeonggil Han
- Department of Microbiology, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Dongeun Yong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Kook Jin Lim
- Nanomedical Graduate Program, Yonsei University, Seoul, 03722, Republic of Korea.,Proteomtech Inc., 1101 Wooree-Venture Town, Seoul, 07573, Republic of Korea
| | - Jeon-Soo Shin
- Nanomedical Graduate Program, Yonsei University, Seoul, 03722, Republic of Korea. .,Department of Microbiology, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea. .,Severance Biomedical Science Institute and Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Kyung-Hwa Yoo
- Department of Physics, Yonsei University, Seoul, 03722, Republic of Korea. .,Nanomedical Graduate Program, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
73
|
Götz T, Dahms M, Kirchhoff J, Beleites C, Glaser U, Bohnert JA, Pletz MW, Popp J, Schlattmann P, Neugebauer U. Automated and rapid identification of multidrug resistant Escherichia coli against the lead drugs of acylureidopenicillins, cephalosporins, and fluoroquinolones using specific Raman marker bands. JOURNAL OF BIOPHOTONICS 2020; 13:e202000149. [PMID: 32410283 DOI: 10.1002/jbio.202000149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 06/11/2023]
Abstract
A Raman-based, strain-independent, semi-automated method is presented that allows the rapid (<3 hours) determination of antibiotic susceptibility of bacterial pathogens isolated from clinical samples. Applying a priori knowledge about the mode of action of the respective antibiotic, we identified characteristic Raman marker bands in the spectrum and calculated batch-wise weighted sum scores from standardized Raman intensity differences between spectra of antibiotic exposed and nonexposed samples of the same strains. The lead substances for three relevant antibiotic classes (fluoroquinolone ciprofloxacin, third-generation cephalosporin cefotaxime, ureidopenicillin piperacillin) against multidrug-resistant Gram-negative bacteria (MRGN) revealed a high sensitivity and specificity for the susceptibility testing of two Escherichia coli laboratory strains and 12 clinical isolates. The method benefits from the parallel incubation of control and treated samples, which reduces the variance due to alterations in cultivation conditions and the standardization of differences between batches leading to long-term comparability of Raman measurements.
Collapse
Affiliation(s)
- Theresa Götz
- Institute of Medical Statistics, Computer Sciences and Data Science, Jena University Hospital, Jena, Germany
| | - Marcel Dahms
- Leibniz Institute of Photonic Technology, Leibniz-IPHT, Jena, Germany
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
- InfectoGnostics Research Campus Jena e.V, Centre for Applied Research, Jena, Germany
| | - Johanna Kirchhoff
- Leibniz Institute of Photonic Technology, Leibniz-IPHT, Jena, Germany
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
- InfectoGnostics Research Campus Jena e.V, Centre for Applied Research, Jena, Germany
| | | | - Uwe Glaser
- Leibniz Institute of Photonic Technology, Leibniz-IPHT, Jena, Germany
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
- InfectoGnostics Research Campus Jena e.V, Centre for Applied Research, Jena, Germany
| | - Jürgen A Bohnert
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Mathias W Pletz
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Jürgen Popp
- Leibniz Institute of Photonic Technology, Leibniz-IPHT, Jena, Germany
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
- InfectoGnostics Research Campus Jena e.V, Centre for Applied Research, Jena, Germany
- Jena Center of Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| | - Peter Schlattmann
- Institute of Medical Statistics, Computer Sciences and Data Science, Jena University Hospital, Jena, Germany
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Ute Neugebauer
- Leibniz Institute of Photonic Technology, Leibniz-IPHT, Jena, Germany
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
- InfectoGnostics Research Campus Jena e.V, Centre for Applied Research, Jena, Germany
- Jena Center of Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
74
|
Development of an Inverted Epifluorescence Microscope for Long-Term Monitoring of Bacteria in Multiplexed Microfluidic Devices. SENSORS 2020; 20:s20154140. [PMID: 32722401 PMCID: PMC7435752 DOI: 10.3390/s20154140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 01/02/2023]
Abstract
Developing more efficient methods for antibiotic susceptibility testing is a pressing issue in novel drug development as bacterial resistance to antibiotics becomes increasingly common. Microfluidic devices have been demonstrated to be powerful platforms that allow researchers to perform multiplexed antibiotic testing. However, the level of multiplexing within microdevices is limited, evidencing the need of creating simple, low-cost and high-resolution imaging systems that can be integrated in antibiotic development pipelines. This paper describes the design and development of an epifluorescence inverted microscope that enables long-term monitoring of bacteria inside multiplexed microfluidic devices. The goal of this work is to provide a simple microscope powerful enough to allow single-cell analysis of bacteria at a reduced cost. This facilitates increasing the number of microscopes that are simultaneously used for antibiotic testing. We prove that the designed system is able to accurately detect fluorescent beads of 100 nm, demonstrating comparable features to high-end commercial microscopes and effectively achieving the resolution required for single-cell analysis of bacteria. The proposed microscope could thus increase the efficiency in antibiotic testing while reducing cost, size, weight, and power requirements, contributing to the successful development of new antibiotic drugs.
Collapse
|
75
|
Hoffman JM, Flynn AJ, Juskewitch JE, Freimuth RR. Biomedical Data Science and Informatics Challenges to Implementing Pharmacogenomics with Electronic Health Records. Annu Rev Biomed Data Sci 2020. [DOI: 10.1146/annurev-biodatasci-020320-093614] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pharmacogenomic information must be incorporated into electronic health records (EHRs) with clinical decision support in order to fully realize its potential to improve drug therapy. Supported by various clinical knowledge resources, pharmacogenomic workflows have been implemented in several healthcare systems. Little standardization exists across these efforts, however, which limits scalability both within and across clinical sites. Limitations in information standards, knowledge management, and the capabilities of modern EHRs remain challenges for the widespread use of pharmacogenomics in the clinic, but ongoing efforts are addressing these challenges. Although much work remains to use pharmacogenomic information more effectively within clinical systems, the experiences of pioneering sites and lessons learned from those programs may be instructive for other clinical areas beyond genomics. We present a vision of what can be achieved as informatics and data science converge to enable further adoption of pharmacogenomics in the clinic.
Collapse
Affiliation(s)
- James M. Hoffman
- Department of Pharmaceutical Sciences and the Office of Quality and Patient Care, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Allen J. Flynn
- Department of Learning Health Sciences, Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Justin E. Juskewitch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Robert R. Freimuth
- Division of Digital Health Sciences, Department of Health Sciences Research, Center for Individualized Medicine, and Information and Knowledge Management, Mayo Clinic, Rochester, Minnesota 55905, USA
| |
Collapse
|
76
|
Vasala A, Hytönen VP, Laitinen OH. Modern Tools for Rapid Diagnostics of Antimicrobial Resistance. Front Cell Infect Microbiol 2020; 10:308. [PMID: 32760676 PMCID: PMC7373752 DOI: 10.3389/fcimb.2020.00308] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/22/2020] [Indexed: 12/18/2022] Open
Abstract
Fast, robust, and affordable antimicrobial susceptibility testing (AST) is required, as roughly 50% of antibiotic treatments are started with wrong antibiotics and without a proper diagnosis of the pathogen. Validated growth-based AST according to EUCAST or CLSI (European Committee on Antimicrobial Susceptibility Testing, Clinical Laboratory Standards Institute) recommendations is currently suggested to guide the antimicrobial therapy. Any new AST should be validated against these standard methods. Many rapid diagnostic techniques can already provide pathogen identification. Some of them can additionally detect the presence of resistance genes or resistance proteins, but usually isolated pure cultures are needed for AST. We discuss the value of the technologies applying nucleic acid amplification, whole genome sequencing, and hybridization as well as immunodiagnostic and mass spectrometry-based methods and biosensor-based AST. Additionally, we evaluate the potential of integrated systems applying microfluidics to integrate cultivation, lysis, purification, and signal reading steps. We discuss technologies and commercial products with potential for Point-of-Care Testing (POCT) and their capability to analyze polymicrobial samples without pre-purification steps. The purpose of this critical review is to present the needs and drivers for AST development, to show the benefits and limitations of AST methods, to introduce promising new POCT-compatible technologies, and to discuss AST technologies that are likely to thrive in the future.
Collapse
Affiliation(s)
- Antti Vasala
- Protein Dynamics, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Vesa P. Hytönen
- Protein Dynamics, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories, Tampere, Finland
| | - Olli H. Laitinen
- Protein Dynamics, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
77
|
Mercer DK, Torres MDT, Duay SS, Lovie E, Simpson L, von Köckritz-Blickwede M, de la Fuente-Nunez C, O'Neil DA, Angeles-Boza AM. Antimicrobial Susceptibility Testing of Antimicrobial Peptides to Better Predict Efficacy. Front Cell Infect Microbiol 2020; 10:326. [PMID: 32733816 PMCID: PMC7358464 DOI: 10.3389/fcimb.2020.00326] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
During the development of antimicrobial peptides (AMP) as potential therapeutics, antimicrobial susceptibility testing (AST) stands as an essential part of the process in identification and optimisation of candidate AMP. Standard methods for AST, developed almost 60 years ago for testing conventional antibiotics, are not necessarily fit for purpose when it comes to determining the susceptibility of microorganisms to AMP. Without careful consideration of the parameters comprising AST there is a risk of failing to identify novel antimicrobials at a time when antimicrobial resistance (AMR) is leading the planet toward a post-antibiotic era. More physiologically/clinically relevant AST will allow better determination of the preclinical activity of drug candidates and allow the identification of lead compounds. An important consideration is the efficacy of AMP in biological matrices replicating sites of infection, e.g., blood/plasma/serum, lung bronchiolar lavage fluid/sputum, urine, biofilms, etc., as this will likely be more predictive of clinical efficacy. Additionally, specific AST for different target microorganisms may help to better predict efficacy of AMP in specific infections. In this manuscript, we describe what we believe are the key considerations for AST of AMP and hope that this information can better guide the preclinical development of AMP toward becoming a new generation of urgently needed antimicrobials.
Collapse
Affiliation(s)
| | - Marcelo D. T. Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Penn Institute for Computational Science, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Searle S. Duay
- Department of Chemistry, Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| | - Emma Lovie
- NovaBiotics Ltd, Aberdeen, United Kingdom
| | | | | | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Penn Institute for Computational Science, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Alfredo M. Angeles-Boza
- Department of Chemistry, Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
78
|
Label-Free Electrochemical Microfluidic Chip for the Antimicrobial Susceptibility Testing. Antibiotics (Basel) 2020; 9:antibiotics9060348. [PMID: 32575678 PMCID: PMC7344617 DOI: 10.3390/antibiotics9060348] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/17/2020] [Accepted: 06/17/2020] [Indexed: 01/15/2023] Open
Abstract
The emergence and spread of antibiotic-resistant bacteria is a global threat to human health. An accurate antibiotic susceptibility test (AST) before initiating the treatment is paramount in the treatment and bacterial resistance control. However, the current AST methods either are complex, use chemical and biological labels, lack multiplexing, are expensive, or are too slow to be used for routine screening. The primary objective of the current study is to develop an automated electrochemical microfluidic chip (EMC) for simple and rapid AST. The microfluidic channels and gold microelectrodes were designed for the automation of antibiotic mixing and distribution in multiple test chambers and for electrical signal measurements. The designed chip was tested for AST with E. coli samples, and the results were compared with conventional broth microdilution. The presented EMC provided rapid bacterial count and AST in 170 and 150 min, respectively, while the conventional broth microdilution evaluates in 450 and 240 min, respectively. The rapid AST capability of the EMC was further demonstrated with the artificial urine samples, and the results were obtained in 270 min, which was 90 min faster than the broth microdilution method. Additionally, the minimum inhibitory concentration (MIC) was evaluated on the EMC and compared with the results from an AlamarBlue assay. The experimental results indicate the sensitivity of the chip, minimum loss of antibiotics, and eventually, reduction in the evolution of antibiotic resistance. Cumulatively, we have developed an automated, label-free, economical, rapid, robust, and user-friendly EMC for the evaluation of AST in urine samples.
Collapse
|
79
|
Yang C, Hu F, Zhang X, Ren C, Huang F, Liu J, Zhang Y, Yang L, Gao Y, Liu B, Liu J. Combating bacterial infection by in situ self-assembly of AIEgen-peptide conjugate. Biomaterials 2020; 244:119972. [DOI: 10.1016/j.biomaterials.2020.119972] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 01/04/2023]
|
80
|
Iriya R, Jing W, Syal K, Mo M, Chen C, Yu H, Haydel SE, Wang S, Tao N. Rapid antibiotic susceptibility testing based on bacterial motion patterns with long short-term memory neural networks. IEEE SENSORS JOURNAL 2020; 20:4940-4950. [PMID: 32440258 PMCID: PMC7241544 DOI: 10.1109/jsen.2020.2967058] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Antibiotic resistance is an increasing public health threat. To combat it, a fast method to determine the antibiotic susceptibility of infecting pathogens is required. Here we present an optical imaging-based method to track the motion of single bacterial cells and generate a model to classify active and inactive cells based on the motion patterns of the individual cells. The model includes an image-processing algorithm to segment individual bacterial cells and track the motion of the cells over time, and a deep learning algorithm (Long Short-Term Memory network) to learn and determine if a bacterial cell is active or inactive. By applying the model to human urine specimens spiked with an Escherichia coli lab strain, we show that the method can accurately perform antibiotic susceptibility testing as fast as 30 minutes for five commonly used antibiotics.
Collapse
Affiliation(s)
- Rafael Iriya
- School of Electrical, Computer and Energy engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Wenwen Jing
- The Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ, 85287, USA
| | - Karan Syal
- The Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ, 85287, USA
| | - Manni Mo
- The Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ, 85287, USA
| | - Chao Chen
- The Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ, 85287, USA
| | - Hui Yu
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Shelley E Haydel
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Shaopeng Wang
- The Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, AZ, 85287, USA
| | - Nongjian Tao
- School of Electrical, Computer and Energy engineering, Arizona State University, Tempe, AZ, 85287, USA
| |
Collapse
|
81
|
Zhang K, Qin S, Wu S, Liang Y, Li J. Microfluidic systems for rapid antibiotic susceptibility tests (ASTs) at the single-cell level. Chem Sci 2020; 11:6352-6361. [PMID: 34094102 PMCID: PMC8159419 DOI: 10.1039/d0sc01353f] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/01/2020] [Indexed: 12/21/2022] Open
Abstract
Infectious diseases caused by multidrug resistant (MDR) bacterial pathogens are impending threats to global health. Since delays in identifying drug resistance would significantly increase mortality, fast and accurate antibiotic susceptibility tests (ASTs) are critical for addressing the antibiotic resistance issue. However, the conventional methods for ASTs are always labor-intensive, imprecise, complex and slow (taking 2-3 days). To address these issues, some advanced microfluidic systems have been designed for rapid phenotypic and genotypic analysis of antibiotic resistance. This review highlights the recent development of microfluidics-based ASTs at the single-cell or single-molecule level for guiding antibiotic treatment decisions and predicting therapeutic outcomes.
Collapse
Affiliation(s)
- Kaixiang Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University Zhengzhou 450001 China
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University Beijing 100084 China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University Zhengzhou 450001 China
| | - Sixuan Wu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University Zhengzhou 450001 China
| | - Yan Liang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University Zhengzhou 450001 China
| | - Jinghong Li
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University Beijing 100084 China
| |
Collapse
|
82
|
Integrating Bacterial Identification and Susceptibility Testing: A Simple and Rapid Approach to Reduce the Turnaround Time in the Management of Blood Cultures. BIOMED RESEARCH INTERNATIONAL 2020; 2019:8041746. [PMID: 31687399 PMCID: PMC6794959 DOI: 10.1155/2019/8041746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 08/15/2019] [Accepted: 09/16/2019] [Indexed: 11/18/2022]
Abstract
We evaluated a rapid bacterial identification (rID) and a rapid antimicrobial susceptibility testing by disk diffusion (rAST) from positive blood culture to overcome the limitations of the conventional methods and reduce the turnaround time in bloodstream infection diagnostics. The study included hemocultures flagged as positive by bacT/ALERT®, identification by MALDI-TOF MS, and rAST. The results were compared to identification and antimicrobial susceptibility testing (AST) results by current standard methods, after 24 h incubation. For rAST categorical agreement (CA), very major errors (VME), major errors (ME), and minor errors (mE) were calculated. A total of 524 bacterial samples isolated from blood cultures were obtained, including 246 Gram-negative (GN) and 278 Gram-positive (GP) aerobes. The overall concordance of rID was 88.6%, and it was highest among GN (96%). A total of 2196 and 1476 antimicrobial agent comparisons were obtained for GN and GP, respectively. Evaluation of rAST, CA, VME, ME, and mE disclosed 97.7, 0.7, 0.5, and 1.1% for GN and 98.0, 0.5, 0.7, and 0.8% for GP, respectively. Meropenem CA, VME, and ME were 98.3, 0.5, and 0.5%, respectively; mE was not observed. Oxacillin CA, ME, and mE were 97.4, 1.6, and 0.6%, respectively; VME was not observed. Overall, kappa scores of the results of the comparisons demonstrated the high agreement between rAST and the standard method. Identification and AST of aerobic bacteria from positive blood cultures after a short period of incubation on solid blood agar is a fast and reliable method that may improve the management of bloodstream infections.
Collapse
|
83
|
Monitoring of drug resistance towards reducing the toxicity of pharmaceutical compounds: Past, present and future. J Pharm Biomed Anal 2020; 186:113265. [PMID: 32283481 DOI: 10.1016/j.jpba.2020.113265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/15/2020] [Accepted: 03/16/2020] [Indexed: 12/12/2022]
Abstract
Drug resistance is worldwide health care crisis which decrease drug efficacy and developing toxicities. Effective resistance detection techniques could alleviate treatment cost and mortality associated with this crisis. In this review, the conventional and modern analysis methods for monitoring of drug resistance are presented. Also, various types of emerging rapid and sensitive techniques including electrochemical, electrical, optical and nano-based methods for the screening of drug resistance were discussed. Applications of various methods for the sensitive and rapid detection of drug resistance are investigated. The review outlines existing key issues in the determination which must be overcome before any of these techniques becomes a feasible method for the rapid detection of drug resistance. In this review, the roles of nanomaterials on development of novel methods for the monitoring of drug resistance were presented. Also, limitations and challenges of conventional and modern methods were discussed.
Collapse
|
84
|
Kumar N, Wang W, Ortiz-Marquez JC, Catalano M, Gray M, Biglari N, Hikari K, Ling X, Gao J, van Opijnen T, Burch KS. Dielectrophoresis assisted rapid, selective and single cell detection of antibiotic resistant bacteria with G-FETs. Biosens Bioelectron 2020; 156:112123. [PMID: 32174552 DOI: 10.1016/j.bios.2020.112123] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/07/2020] [Accepted: 02/21/2020] [Indexed: 01/19/2023]
Abstract
The rapid increase in antibiotic resistant pathogenic bacteria has become a global threat, which besides the development of new drugs, requires rapid, cheap, scalable, and accurate diagnostics. Label free biosensors relying on electrochemical, mechanical, and mass based detection of whole bacterial cells have attempted to meet these requirements. However, the trade-off between selectivity and sensitivity of such sensors remains a key challenge. In particular, point-of-care diagnostics that are able to reduce and/or prevent unneeded antibiotic prescriptions require highly specific probes with sensitive and accurate transducers that can be miniaturized and multiplexed, and that are easy to operate and cheap. Towards achieving this goal, we present a number of advances in the use of graphene field effect transistors (G-FET) including the first use of peptide probes to electrically detect antibiotic resistant bacteria in a highly specific manner. In addition, we dramatically reduce the needed concentration for detection by employing dielectrophoresis for the first time in a G-FET, allowing us to monitor changes in the Dirac point due to individual bacterial cells. Specifically, we realized rapid binding of bacterial cells to a G-FET by electrical field guiding to the device to realize an overall 3 orders of magnitude decrease in cell-concentration enabling a single-cell detection limit, and 9-fold reduction in needed time to 5 min. Utilizing our new biosensor and procedures, we demonstrate the first selective, electrical detection of the pathogenic bacterial species Staphylococcus aureus and antibiotic resistant Acinetobacter baumannii on a single platform.
Collapse
Affiliation(s)
- Narendra Kumar
- Department of Physics, Boston College, Chestnut Hill, MA, 02467, United States
| | - Wenjian Wang
- Department of Chemistry, Boston College, Chestnut Hill, MA, 02467, United States
| | | | - Matthew Catalano
- Department of Physics, Boston College, Chestnut Hill, MA, 02467, United States
| | - Mason Gray
- Department of Physics, Boston College, Chestnut Hill, MA, 02467, United States
| | - Nadia Biglari
- Department of Physics, Boston College, Chestnut Hill, MA, 02467, United States
| | - Kitadai Hikari
- Department of Chemistry, Boston University, Boston, MA, 02215, United States
| | - Xi Ling
- Department of Chemistry, Boston University, Boston, MA, 02215, United States; Division of Materials Science and Engineering, Boston University, Boston, MA, 02214, United States; The Photonics Center, Boston University, Boston, MA, 02214, United States
| | - Jianmin Gao
- Department of Chemistry, Boston College, Chestnut Hill, MA, 02467, United States.
| | - Tim van Opijnen
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, United States.
| | - Kenneth S Burch
- Department of Physics, Boston College, Chestnut Hill, MA, 02467, United States.
| |
Collapse
|
85
|
Rao R P, Sharma S, Mehrotra T, Das R, Kumar R, Singh R, Roy I, Basu T. Rapid Electrochemical Monitoring of Bacterial Respiration for Gram-Positive and Gram-Negative Microbes: Potential Application in Antimicrobial Susceptibility Testing. Anal Chem 2020; 92:4266-4274. [PMID: 32050756 DOI: 10.1021/acs.analchem.9b04810] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antimicrobial resistance is a grave threat to human life. Currently used time-consuming antibiotic susceptibility test (AST) methods limit physicians in selecting proper antibiotics. Hence, we developed a rapid AST using electroanalysis with a 15 min assay time, called EAST, which is live-monitored by time-lapse microscopy video. The present work reports systematical electrochemical analysis and standardization of protocol for EAST measurement. The proposed EAST is successfully applied for Gram-positive Bacillus subtilis and Gram-negative Escherichia coli as model organisms to monitor bacterial concentration, decay kinetics in the presence of various antibiotics (ciprofloxacin, cefixime, and amoxycillin), drug efficacy, and IC50. Bacterial decay kinetics in the presence of antibiotics were validated by the colony counting method, field emission scanning electron microscopy, and atomic force microscopy image analysis. The EAST predicts the antibiotic susceptibility of bacteria within 15 min, which is a significant advantage over existing techniques that consume hours to days. The EAST was explored further by using bacteria-friendly l-lysine-functionalized cerium oxide nanoparticle coated indium tin oxide as a working electrode to observe the enhanced electron-transfer rate in the EAST. The results are very significant for future miniaturization and automation. The proposed EAST has huge potential in the development of a rapid AST device for applications in the clinical and pharmaceutical industries.
Collapse
Affiliation(s)
| | - Shalini Sharma
- Department of Chemistry, University of Delhi, New Delhi, Delhi 110007, India
| | | | | | | | | | - Indrajit Roy
- Department of Chemistry, University of Delhi, New Delhi, Delhi 110007, India
| | | |
Collapse
|
86
|
Yi SY, Jeong J, Kim KE, Park K, Shin YB. Staphylococcus aureus Specific FRET Probe-Based Antibacterial Susceptibility Testing (SF-AST) by Detection of Micrococcal Nuclease Activity. ACS Infect Dis 2020; 6:215-223. [PMID: 31823600 DOI: 10.1021/acsinfecdis.9b00260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In this study, we describe a simple and rapid antibacterial susceptibility testing (AST) method for Staphylococcus aureus called S. aureus specific fluorescence resonance energy transfer (FRET) probe-based AST (SF-AST), which is based on an S. aureus specific FRET probe (SF probe) that detects micrococcal nuclease (MNase) activity secreted from S. aureus. The SF-AST was tested with an S. aureus quality control (QC) strain against six relevant antibiotics, and the minimum inhibitory concentration (MIC) values obtained with the broth microdilution (BMD) method were compared, as a gold standard AST. Results were obtained with high accuracy in 4-6 h. The MIC for the methicillin resistance using 20 clinical S. aureus isolates of SF-AST showed 100% sensitivity, specificity, positive predictive value, and negative predictive value, as compared to BMD. Thus, the SF-AST method is a simple, rapid, and useful antibiotic resistance test for S. aureus, and it provides a basis for clinical treatment in a short time.
Collapse
Affiliation(s)
- So Yeon Yi
- BioNano Health Guard Research Center, Daejeon 34141, Republic of Korea
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jinyoung Jeong
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Kyoon Eon Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Kyoungsook Park
- BioNano Health Guard Research Center, Daejeon 34141, Republic of Korea
| | - Yong Beom Shin
- BioNano Health Guard Research Center, Daejeon 34141, Republic of Korea
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| |
Collapse
|
87
|
Emerging Designs of Electronic Devices in Biomedicine. MICROMACHINES 2020; 11:mi11020123. [PMID: 31979030 PMCID: PMC7074089 DOI: 10.3390/mi11020123] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/15/2022]
Abstract
A long-standing goal of nanoelectronics is the development of integrated systems to be used in medicine as sensor, therapeutic, or theranostic devices. In this review, we examine the phenomena of transport and the interaction between electro-active charges and the material at the nanoscale. We then demonstrate how these mechanisms can be exploited to design and fabricate devices for applications in biomedicine and bioengineering. Specifically, we present and discuss electrochemical devices based on the interaction between ions and conductive polymers, such as organic electrochemical transistors (OFETs), electrolyte gated field-effect transistors (FETs), fin field-effect transistor (FinFETs), tunnelling field-effect transistors (TFETs), electrochemical lab-on-chips (LOCs). For these systems, we comment on their use in medicine.
Collapse
|
88
|
Prevalence and antibiotic resistance patterns of Vibrio parahaemolyticus isolated from different types of seafood in Selangor, Malaysia. Saudi J Biol Sci 2020; 27:1602-1608. [PMID: 32489301 PMCID: PMC7253911 DOI: 10.1016/j.sjbs.2020.01.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/05/2020] [Accepted: 01/06/2020] [Indexed: 01/07/2023] Open
Abstract
Vibrio parahaemolyticus is a foodborne bacterial pathogen that may cause gastroenteritis in humans through the consumption of seafood contaminated with this microorganism. The emergence of antimicrobial and multidrug-resistant bacteria is another serious public health threat worldwide. In this study, the prevalence and antibiotic susceptibility test of V. parahaemolyticus in blood clams, shrimps, surf clams, and squids were determined. The overall prevalence of V. parahaemolyticus in seafood was 85.71% (120/140), consisting of 91.43% (32/35) in blood clam, 88.57% (31/35) in shrimps, 82.86% (29/35) in surf clams, and 80% (28/35) in squids. The majority of V. parahaemolyticus isolates from the seafood samples were found to be susceptible to most antibiotics except ampicillin, cefazolin, and penicillin. The MAR indices of V. parahaemolyticus isolates ranged from 0.04 to 0.71 and about 90.83% of isolates were found resistant to more than one antibiotic. The high prevalence of V. parahaemolyticus in seafood and multidrug-resistant isolates detected in this study could pose a potential risk to human health and hence appropriate control methods should be in place to minimize the potential contamination and prevent the emergence of antibiotic resistance.
Collapse
|
89
|
Abstract
Antimicrobial susceptibility testing (AST) technologies help to accelerate the initiation of targeted antimicrobial therapy for patients with infections and could potentially extend the lifespan of current narrow-spectrum antimicrobials. Although conceptually new and rapid AST technologies have been described, including new phenotyping methods, digital imaging and genomic approaches, there is no single major, or broadly accepted, technological breakthrough that leads the field of rapid AST platform development. This might be owing to several barriers that prevent the timely development and implementation of novel and rapid AST platforms in health-care settings. In this Consensus Statement, we explore such barriers, which include the utility of new methods, the complex process of validating new technology against reference methods beyond the proof-of-concept phase, the legal and regulatory landscapes, costs, the uptake of new tools, reagent stability, optimization of target product profiles, difficulties conducting clinical trials and issues relating to quality and quality control, and present possible solutions. This Consensus Statement presents the barriers that currently prevent the timely development and implementation of novel and rapid antimicrobial susceptibility testing platforms, including the costs involved, uptake of new tools, legal and regulatory aspects, difficulties conducting clinical trials and quality control, and presents possible solutions.
Collapse
|
90
|
Ma L, Xie X, Liu H, Huang Y, Wu H, Jiang M, Xu P, Ye X, Zhou C. Potent antibacterial activity of MSI-1 derived from the magainin 2 peptide against drug-resistant bacteria. Theranostics 2020; 10:1373-1390. [PMID: 31938070 PMCID: PMC6956804 DOI: 10.7150/thno.39157] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 09/26/2019] [Indexed: 11/05/2022] Open
Abstract
The structural modification of existing AMPs is an effective strategy to develop antimicrobial agents with high-efficiency, low-cost and low-toxicity antimicrobial agents. Methods: Here, we truncated 14-amino-acids at the N-terminus of MSI-78 to obtain MSI and further modified MSI to obtain four peptide analogs: MSI-1, MSI-2, MSI-3 and MSI-4. These peptide mutants were evaluated regarding their antibacterial activity against various sensitive or resistant bacteria; toxicity against mammalian cells or mice; and stability against violent pH, temperature variations and high NaCl concentrations. Finally, we also elucidated the possible mechanisms underlying its mode of action. Results: The results showed that MSI-1 and MSI-3 displayed activity that was superior to that of MSI-78 with MICs of 4-16 μg/ml and MBCs of 8-64 μg/ml, respectively, especially against drug-resistant bacteria, due to the increase in percent helicity and amphiphilicity. However, MSI-3, with higher hydrophobicity and antibacterial activity, had a relatively higher hemolysis rate and toxicity than MSI-1. MSI-1 exerted rapid bactericidal activity and effectively improved the survival rate and wound closure in penicillin-resistant E. coli-infected mice by eliminating bacterial counts in mouse organs or subeschar, further inhibiting the systemic dissemination of bacteria. Additionally, MSI-1 displayed perfect stability against violent pH, temperature variations and high NaCl concentrations and has the ability to circumvent the development of drug resistance. In terms of the mode of action, we found that at the super-MIC level, MSI-1 exhibited direct antimicrobial activity by disrupting the integrity of the bacterial cell membrane, while at the sub-MIC level, it bound to bacterial DNA to inhibit DNA replication and protein expression and ultimately disrupted bacterial biological function. Conclusions: This novel peptide MSI-1 could be a potential candidate for drug development against infection induced by drug-resistant bacteria.
Collapse
|
91
|
Chen J, Shi X, Zhu Y, Chen Y, Gao M, Gao H, Liu L, Wang L, Mao C, Wang Y. On-demand storage and release of antimicrobial peptides using Pandora's box-like nanotubes gated with a bacterial infection-responsive polymer. Theranostics 2020; 10:109-122. [PMID: 31903109 PMCID: PMC6929614 DOI: 10.7150/thno.38388] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 09/03/2019] [Indexed: 01/08/2023] Open
Abstract
Background: Localized delivery of antimicrobial agents such as antimicrobial peptides (AMPs) by a biomaterial should be on-demand. Namely, AMPs should be latent and biocompatible in the absence of bacterial infection, but released in an amount enough to kill bacteria immediately in response to bacterial infection. Methods: To achieve the unmet goal of such on-demand delivery, here we turned a titanium implant with titania nanotubes (Ti-NTs) into a Pandora's box. The box was loaded with AMPs (HHC36 peptides, with a sequence of KRWWKWWRR) inside the nanotubes and "closed" (surface-modified) with a pH-responsive molecular gate, poly(methacrylic acid) (PMAA), which swelled under normal physiological conditions (pH 7.4) but collapsed under bacterial infection (pH ≤ 6.0). Thus, the PMAA-gated Ti-NTs behaved just like a Pandora's box. The box retarded the burst release of AMPs under physiological conditions because the gate swelled to block the nanotubes opening. However, it was opened to release AMPs to kill bacteria immediately when bacterial infection occurred to lowering the pH (and thus made the gate collapse). Results: We demonstrated such smart excellent bactericidal activity against a panel of four clinically important bacteria, including Staphylococcus aureus, Escherichia coli, Pseudomonas aeruginosa, methicillin-resistant Staphylococcus aureus. In addition, this box was biocompatible and could promote the osteogenic differentiation of human mesenchymal stem cells. Both in vitro and in vivo studies confirmed the smart "on-demand" bactericidal activity of the Pandora's box. The molecularly gated Pandora's box design represents a new strategy in smart drug delivery.
Collapse
Affiliation(s)
- Junjian Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- School of Biomedical Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Xuetao Shi
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510641, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| | - Ye Zhu
- Department of Chemistry and Biochemistry, University of Oklahoma, Stephenson Life Sciences Research Center Norman, OK, 73019, USA
| | - Yunhua Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| | - Meng Gao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510006, China
| | - Huichang Gao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- School of Biomedical Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Lei Liu
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510641, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510006, China
| | - Lin Wang
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510641, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510006, China
| | - Chuanbin Mao
- Department of Chemistry and Biochemistry, University of Oklahoma, Stephenson Life Sciences Research Center Norman, OK, 73019, USA
| | - Yingjun Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- School of Biomedical Science and Engineering, South China University of Technology, Guangzhou 510006, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510006, China
| |
Collapse
|
92
|
Mei L, Zhu S, Yin W, Chen C, Nie G, Gu Z, Zhao Y. Two-dimensional nanomaterials beyond graphene for antibacterial applications: current progress and future perspectives. Theranostics 2020; 10:757-781. [PMID: 31903149 PMCID: PMC6929992 DOI: 10.7150/thno.39701] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 09/21/2019] [Indexed: 12/12/2022] Open
Abstract
The marked augment of drug-resistance to traditional antibiotics underlines the crying need for novel replaceable antibacterials. Research advances have revealed the considerable sterilization potential of two-dimension graphene-based nanomaterials. Subsequently, two-dimensional nanomaterials beyond graphene (2D NBG) as novel antibacterials have also demonstrated their power for disinfection due to their unique physicochemical properties and good biocompatibility. Therefore, the exploration of antibacterial mechanisms of 2D NBG is vital to manipulate antibacterials for future applications. Herein, we summarize the recent research progress of 2D NBG-based antibacterial agents, starting with a detailed introduction of the relevant antibacterial mechanisms, including direct contact destruction, oxidative stress, photo-induced antibacterial, control drug/metallic ions releasing, and the multi-mode synergistic antibacterial. Then, the effect of the physicochemical properties of 2D NBG on their antibacterial activities is also discussed. Additionally, a summary of the different kinds of 2D NBG is given, such as transition-metal dichalcogenides/oxides, metal-based compounds, nitride-based nanomaterials, black phosphorus, transition metal carbides, and nitrides. Finally, we rationally analyze the current challenges and new perspectives for future study of more effective antibacterial agents. This review not only can help researchers grasp the current status of 2D NBG antibacterials, but also may catalyze breakthroughs in this fast-growing field.
Collapse
Affiliation(s)
- Linqiang Mei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Wenyan Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Chunying Chen
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100190, China
| | - Guangjun Nie
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuliang Zhao
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100190, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
93
|
Guliy OI, Zaitsev BD, Borodina IA. New approach for determination of antimicrobial susceptibility to antibiotics by an acoustic sensor. Appl Microbiol Biotechnol 2019; 104:1283-1290. [PMID: 31865437 DOI: 10.1007/s00253-019-10295-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 12/19/2022]
Abstract
For the first time, a rapid method was proposed to determine the susceptibility of Escherichia coli cells to antibiotics by the example of ampicillin by using a biological sensor based on a slot mode in an acoustic delay line. It has been established that an indicator of the antibiotic activity to microbial cells is the difference between the recorded sensor's signal before and after exposure cells with antibiotic. The depth and frequency of the peaks of resonant absorption in the frequency dependence of the insertion loss of sensor varied after adding an antibiotic with different concentrations to the microbial cells. By using the acoustic sensor based on slot-mode a criterion of E. coli sensitivity to ampicillin was established. The advantages of this method are the ability to carry out the analysis directly in the liquid, the short analysis time (within 10-15 min), and the possibility to reusable sensor.
Collapse
Affiliation(s)
- O I Guliy
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, Saratov, 410049, Russia.
| | - B D Zaitsev
- Kotelnikov Institute of Radio Engineering and Electronics, Russian Academy of Sciences, Saratov Branch, Saratov, 410019, Russia
| | - I A Borodina
- Kotelnikov Institute of Radio Engineering and Electronics, Russian Academy of Sciences, Saratov Branch, Saratov, 410019, Russia
| |
Collapse
|
94
|
Highly specific Electrochemical Sensing of Pseudomonas aeruginosa in patients suffering from corneal ulcers: A comparative study. Sci Rep 2019; 9:18320. [PMID: 31797959 PMCID: PMC6892848 DOI: 10.1038/s41598-019-54667-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 11/08/2019] [Indexed: 01/12/2023] Open
Abstract
Pseudomonas aeruginosa is the most common pathogenic gram-negative bacteria causing corneal ulcers globally. In severe cases, often after trauma and eye injury, corneal destruction progresses rapidly and may be completed within 24–48 h causing blindness. In our preliminary work, we have established an ultrasensitive polyaniline (PANI)/gold nanoparticles (Au NPs)/indium tin oxide (ITO) modified sensor for rapid detection of pyocyanin (PYO) in P. aeruginosa infections with a linear range from 238 μM to 1.9 μM and a detection limit of 500 nM. In the present study, we evaluated the efficiency of the established modified electrochemical sensor in the diagnosis of P. aeruginosa in 50 samples collected from patients suffering from corneal ulcers. The obtained results were compared with the results gained by the screen-printed electrode, conventional techniques, automated identification method, and the amplification of the 16 s rRNA gene by PCR as a gold standard test for P. aeruginosa identification. We have found that the electrochemical detection of PYO by square wave voltammetry technique using PANI/Au NPs modified ITO electrode was the only technique showing 100% agreement with the molecular method in sensitivity, specificity, positive and negative predictive values when compared with the SPE, conventional and automated methods.
Collapse
|
95
|
Hossain SMZ, Mansour N. Biosensors for on-line water quality monitoring – a review. ARAB JOURNAL OF BASIC AND APPLIED SCIENCES 2019. [DOI: 10.1080/25765299.2019.1691434] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- S. M. Zakir Hossain
- Department of Chemical Engineering, University of Bahrain, Isa Town, Kingdom of Bahrain
| | - Noureddine Mansour
- Department of Chemical Engineering, University of Bahrain, Isa Town, Kingdom of Bahrain
| |
Collapse
|
96
|
García-Álvarez L, Busto H, Oteo JA. Proton nuclear magnetic resonance for antimicrobial drug susceptibility studies: why has progress been slow? Future Microbiol 2019; 14:1175-1177. [PMID: 31625445 DOI: 10.2217/fmb-2019-0216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Lara García-Álvarez
- Department of Infectious Diseases, Center for Biomedical Research of La Rioja (CIBIR), San Pedro University Hospital. Logroño, 26006, Spain
| | - Héctor Busto
- Department of Chemistry, Center for Research in Chemical Synthesis (CISQ), University of La Rioja. Logroño, 26006, Spain
| | - José A Oteo
- Department of Infectious Diseases, Center for Biomedical Research of La Rioja (CIBIR), San Pedro University Hospital. Logroño, 26006, Spain
| |
Collapse
|
97
|
Miller LM, Silver CD, Herman R, Duhme-Klair AK, Thomas GH, Krauss TF, Johnson SD. Surface-Bound Antibiotic for the Detection of β-Lactamases. ACS APPLIED MATERIALS & INTERFACES 2019; 11:32599-32604. [PMID: 31449379 PMCID: PMC7007045 DOI: 10.1021/acsami.9b05793] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/25/2019] [Indexed: 06/10/2023]
Abstract
Antimicrobial resistance (AMR) has been identified as a major threat to public health worldwide. To ensure appropriate use of existing antibiotics, rapid and reliable tests of AMR are necessary. One of the most common and clinically important forms of bacterial resistance is to β-lactam antibiotics (e.g., penicillin). This resistance is often caused by β-lactamases, which hydrolyze β-lactam drugs, rendering them ineffective. Current methods for detecting these enzymes require either time-consuming growth assays or antibiotic mimics such as nitrocefin. Here, we report the development of a surface-bound, clinically relevant β-lactam drug that can be used to detect β-lactamases and that is compatible with a range of high-sensitivity, low-cost, and label-free analytical techniques currently being developed for point-of-care-diagnostics. Furthermore, we demonstrate the use of these functionalized surfaces to selectively detect β-lactamases in complex biological media, such as urine.
Collapse
Affiliation(s)
- Lisa M. Miller
- Department
of Chemistry, Department of Electronic Engineering, Department of Biology, and Department of
Physics, University of York, Heslington YO10 5DD, U.K.
| | - Callum D. Silver
- Department
of Chemistry, Department of Electronic Engineering, Department of Biology, and Department of
Physics, University of York, Heslington YO10 5DD, U.K.
| | - Reyme Herman
- Department
of Chemistry, Department of Electronic Engineering, Department of Biology, and Department of
Physics, University of York, Heslington YO10 5DD, U.K.
| | - Anne-Kathrin Duhme-Klair
- Department
of Chemistry, Department of Electronic Engineering, Department of Biology, and Department of
Physics, University of York, Heslington YO10 5DD, U.K.
| | - Gavin H. Thomas
- Department
of Chemistry, Department of Electronic Engineering, Department of Biology, and Department of
Physics, University of York, Heslington YO10 5DD, U.K.
| | - Thomas F. Krauss
- Department
of Chemistry, Department of Electronic Engineering, Department of Biology, and Department of
Physics, University of York, Heslington YO10 5DD, U.K.
| | - Steven D. Johnson
- Department
of Chemistry, Department of Electronic Engineering, Department of Biology, and Department of
Physics, University of York, Heslington YO10 5DD, U.K.
| |
Collapse
|
98
|
Abstract
Advanced microbiology technologies are rapidly changing our ability to diagnose infections, improve patient care, and enhance clinical workflow. These tools are increasing the breadth, depth, and speed of diagnostic data generated per patient, and testing is being moved closer to the patient through rapid diagnostic technologies, including point-of-care (POC) technologies. Advanced microbiology technologies are rapidly changing our ability to diagnose infections, improve patient care, and enhance clinical workflow. These tools are increasing the breadth, depth, and speed of diagnostic data generated per patient, and testing is being moved closer to the patient through rapid diagnostic technologies, including point-of-care (POC) technologies. While select stakeholders have an appreciation of the value/importance of improvements in the microbial diagnostic field, there remains a disconnect between clinicians and some payers and hospital administrators in terms of understanding the potential clinical utility of these novel technologies. Therefore, a key challenge for the clinical microbiology community is to clearly articulate the value proposition of these technologies to encourage payers to cover and hospitals to adopt advanced microbiology tests. Specific guidance on how to define and demonstrate clinical utility would be valuable. Addressing this challenge will require alignment on this topic, not just by microbiologists but also by primary care and emergency room (ER) physicians, infectious disease specialists, pharmacists, hospital administrators, and government entities with an interest in public health. In this article, we discuss how to best conduct clinical studies to demonstrate and communicate clinical utility to payers and to set reasonable expectations for what diagnostic manufacturers should be required to demonstrate to support reimbursement from commercial payers and utilization by hospital systems.
Collapse
|
99
|
Lee WB, Chien CC, You HL, Kuo FC, Lee MS, Lee GB. An integrated microfluidic system for antimicrobial susceptibility testing with antibiotic combination. LAB ON A CHIP 2019; 19:2699-2708. [PMID: 31328212 DOI: 10.1039/c9lc00585d] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Polypharmacy is routinely administered to fight severe infections, though it has led to rampant multi-drug resistance in many bacterial strains. Preferably, antimicrobial susceptibility testing (AST) would be carried out prior to antibiotic prescription, though it is generally thought to be too complex and labor-intensive. In order to assist clinicians with better antibiotic administration for the effective treatment of bacterial infections, an integrated microfluidic system (IMS) capable of automating AST for 1-2 antibiotics against clinical bacterial pathogens was developed herein. Accurate determination of the minimum and fractional inhibitory concentrations of vancomycin, gentamicin, and linezolid were determined by assaying growth of two clinical methicillin-resistant Staphylococcus aureus isolates via a colorimetric assay on-chip. By applying various antibiotic combinations against a single pathogen in multiple chambers, the IMS could identify the optimal drug combination and the minimum effective dosage by evaluating the fractional inhibitory concentration index. This IMS possessed several advantages over conventional methods, including (1) a 50% reduction in bacterial sample and reagent volume (<50 μL per well), (2) less potential for human error due to its automatic nature, (3) faster liquid manipulation time by integrating the microfluidic components rather than labor-intensive process, and (4) straightforward result interpretation via colorimetric change instead of turbidity degree. Personalized medicine for treatment of bacterial infections may therefore be realized using this IMS.
Collapse
Affiliation(s)
- Wen-Bin Lee
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Chun-Chih Chien
- Laboratory Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, Kaohsiung 83301, Taiwan
| | - Huey-Ling You
- Laboratory Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, Kaohsiung 83301, Taiwan
| | - Feng-Chih Kuo
- Department of Orthopaedic Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, Kaohsiung 83301, Taiwan.
| | - Mel S Lee
- Department of Orthopaedic Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, Kaohsiung 83301, Taiwan.
| | - Gwo-Bin Lee
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan. and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan and Institute of NanoEngineering and Microsystems, National Tsing Hua University, Hsinchu, 30013 Taiwan
| |
Collapse
|
100
|
Behera B, Anil Vishnu GK, Chatterjee S, Sitaramgupta V VSN, Sreekumar N, Nagabhushan A, Rajendran N, Prathik BH, Pandya HJ. Emerging technologies for antibiotic susceptibility testing. Biosens Bioelectron 2019; 142:111552. [PMID: 31421358 DOI: 10.1016/j.bios.2019.111552] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/27/2019] [Accepted: 07/29/2019] [Indexed: 12/22/2022]
Abstract
Superbugs such as infectious bacteria pose a great threat to humanity due to an increase in bacterial mortality leading to clinical treatment failure, lengthy hospital stay, intravenous therapy and accretion of bacteraemia. These disease-causing bacteria gain resistance to drugs over time which further complicates the treatment. Monitoring of antibiotic resistance is therefore necessary so that bacterial infectious diseases can be diagnosed rapidly. Antimicrobial susceptibility testing (AST) provides valuable information on the efficacy of antibiotic agents and their dosages for treatment against bacterial infections. In clinical laboratories, most widely used AST methods are disk diffusion, gradient diffusion, broth dilution, or commercially available semi-automated systems. Though these methods are cost-effective and accurate, they are time-consuming, labour-intensive, and require skilled manpower. Recently much attention has been on developing rapid AST techniques to avoid misuse of antibiotics and provide effective treatment. In this review, we have discussed emerging engineering AST techniques with special emphasis on phenotypic AST. These techniques include fluorescence imaging along with computational image processing, surface plasmon resonance, Raman spectra, and laser tweezer as well as micro/nanotechnology-based device such as microfluidics, microdroplets, and microchamber. The mechanical and electrical behaviour of single bacterial cell and bacterial suspension for the study of AST is also discussed.
Collapse
Affiliation(s)
- Bhagaban Behera
- Biomedical and Electronic (10(-6)-10(-9)) Engineering Systems Laboratory, Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, India
| | - G K Anil Vishnu
- Biomedical and Electronic (10(-6)-10(-9)) Engineering Systems Laboratory, Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, India; Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Suman Chatterjee
- Biomedical and Electronic (10(-6)-10(-9)) Engineering Systems Laboratory, Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, India
| | - V S N Sitaramgupta V
- Biomedical and Electronic (10(-6)-10(-9)) Engineering Systems Laboratory, Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, India
| | - Niranjana Sreekumar
- Biomedical and Electronic (10(-6)-10(-9)) Engineering Systems Laboratory, Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, India
| | - Apoorva Nagabhushan
- Biomedical and Electronic (10(-6)-10(-9)) Engineering Systems Laboratory, Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, India
| | | | - B H Prathik
- Indira Gandhi Institute of Child Health, Bangalore, India
| | - Hardik J Pandya
- Biomedical and Electronic (10(-6)-10(-9)) Engineering Systems Laboratory, Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore, India.
| |
Collapse
|