1
|
Park JH, Kwon S, Kim MJ, Song Z, Bae HE, Kang MJ, Pyun JC. Dual-mode capacitive and localized surface plasmon resonance biosensor based on high-density Au nanoislands. Biosens Bioelectron 2025; 277:117274. [PMID: 39987657 DOI: 10.1016/j.bios.2025.117274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 01/14/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
A capacitive-localized plasmon surface resonance (LSPR) dual-mode biosensor was developed using gold (Au) nanoislands modified on an Au interdigitated electrode (IDE). Au nanoislands were deposited through repeated thermal dewetting to increase their packing density and enhance sensor sensitivity. The response of the capacitive sensor to antibody-antigen interactions was optimized at 0.5 Hz in phosphate-buffered saline. Modification with Au nanoislands significantly reduced the effective electrode gap of the IDE, thereby enhancing the capacitive sensitivity, as evidenced by charge-transfer resistance and electric field analysis. Computer simulations confirmed that the effective electrode gap of a 5 μm gap Au IDE with an 88.1% packing density of Au nanoislands decreased to 525.9 nm. The influence of Au nanoislands on LSPR was assessed through parameters such as λmax, full width at half maximum, Q factor, and figure of merit. Additionally, the electric field enhancement factor, which indicates LSPR sensitivity, was calculated relative to the packing density of the Au nanoislands. The dual-mode sensor demonstrated efficacy in detecting Salmonella typhimurium, and the capacitive and LSPR sensor results showed a statistically significant correlation.
Collapse
Affiliation(s)
- Jun-Hee Park
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, 03722, Seoul, South Korea
| | - Soonil Kwon
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, 03722, Seoul, South Korea
| | - Moon-Ju Kim
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, 03722, Seoul, South Korea
| | - Zhiquan Song
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, 03722, Seoul, South Korea
| | - Hyung Eun Bae
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, 03722, Seoul, South Korea
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), 02456, Seoul, South Korea
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, 03722, Seoul, South Korea.
| |
Collapse
|
2
|
Kim MJ, Bae HE, Kwon S, Park MK, Yong D, Kang MJ, Pyun JC. Phage-targeting bimetallic nanoplasmonic biochip functionalized with bacterial outer membranes as a biorecognition element. Biosens Bioelectron 2023; 238:115598. [PMID: 37597282 DOI: 10.1016/j.bios.2023.115598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/03/2023] [Accepted: 08/12/2023] [Indexed: 08/21/2023]
Abstract
The use of phages-a natural predator of bacteria-has emerged as a therapeutic strategy for treating multidrug-resistant bacterial infections; thus, the isolation and detection of phages from the environment is crucial for advancing phage therapy. Herein, for the first time, we propose a nanoplasmonic-based biodetection platform for phages that utilizes bacterial outer membranes (OMs) as a biorecognition element. Conventional biosensors based on phage-bacteria interactions encounter multiple challenges due to the bacteriolytic phages and potentially toxic bacteria, resulting in instability and risk in the measurement. Therefore, instead of whole living bacteria, we employ a safe biochemical OMs fraction presenting phage-specific receptors, allowing the robust and reliable phage detection. In addition, the biochip is constructed on bimetallic nanoplasmonic islands through solid-state dewetting for synergy between Au and Ag, whereby sensitive detection of phage-OMs interactions is achieved by monitoring the absorption peak shift. For high detection performance, the nanoplasmonic chip is optimized by systematically investigating the morphological features, e.g., size and packing density of the nanoislands. Using our optimized device, phages are detected with high sensitivity (≥∼104 plaques), specificity (little cross-reactivity), and affinity (stronger binding to the host OMs than anti-bacterial antibodies), further exhibiting the cell-killing activities.
Collapse
Affiliation(s)
- Moon-Ju Kim
- Department of Materials and Science and Engineering, Yonsei University, Seoul, 03722, South Korea
| | - Hyung Eun Bae
- Department of Materials and Science and Engineering, Yonsei University, Seoul, 03722, South Korea
| | - Soonil Kwon
- Department of Materials and Science and Engineering, Yonsei University, Seoul, 03722, South Korea
| | - Mi-Kyung Park
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, South Korea
| | - Dongeun Yong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Min-Jung Kang
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, 02792, South Korea
| | - Jae-Chul Pyun
- Department of Materials and Science and Engineering, Yonsei University, Seoul, 03722, South Korea.
| |
Collapse
|
3
|
Jung J, Bong JH, Sung JS, Park JH, Kim TH, Kwon S, Kang MJ, Jose J, Pyun JC. Immunoaffinity biosensors for the detection of SARS-CoV-1 using screened Fv-antibodies from an autodisplayed Fv-antibody library. Biosens Bioelectron 2023; 237:115439. [PMID: 37301177 PMCID: PMC10223632 DOI: 10.1016/j.bios.2023.115439] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/21/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023]
Abstract
The detection of severe acute respiratory syndrome coronavirus (SARS-CoV-1) was demonstrated using screened Fv-antibodies for SPR biosensor and impedance spectrometry. The Fv-antibody library was first prepared on the outer membrane of E. coli using autodisplay technology and the Fv-variants (clones) with a specific affinity toward the SARS-CoV-1 spike protein (SP) were screened using magnetic beads immobilized with the SP. Upon screening the Fv-antibody library, two target Fv-variants (clones) with a specific binding affinity toward the SARS-CoV-1 SP were determined and the Fv-antibodies on two clones were named "Anti-SP1" (with CDR3 amino acid sequence: 1GRTTG5NDRPD11Y) and "Anti-SP2" (with CDR3 amino acid sequence: 1CLRQA5GTADD11V). The binding affinities of the two screened Fv-variants (clones) were analyzed using flow cytometry and the binding constants (KD) were estimated to be 80.5 ± 3.6 nM for Anti-SP1 and 45.6 ± 8.9 nM for Anti-SP2 (n = 3). In addition, the Fv-antibody including three CDR regions (CDR1, CDR2, and CDR3) and frame regions (FRs) between the CDR regions was expressed as a fusion protein (Mw. 40.6 kDa) with a green fluorescent protein (GFP) and the KD values of the expressed Fv-antibodies toward the SP estimated to be 15.3 ± 1.5 nM for Anti-SP1 (n = 3) and 16.3 ± 1.7 nM for Anti-SP2 (n = 3). Finally, the expressed Fv-antibodies screened against SARS-CoV-1 SP (Anti-SP1 and Anti-SP2) were applied for the detection of SARS-CoV-1. Consequently, the detection of SARS-CoV-1 was demonstrated to be feasible using the SPR biosensor and impedance spectrometry utilizing the immobilized Fv-antibodies against the SARS-CoV-1 SP.
Collapse
Affiliation(s)
- Jaeyong Jung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Ji-Hong Bong
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Jeong Soo Sung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Jun-Hee Park
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Tae-Hun Kim
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Soonil Kwon
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Joachim Jose
- Institute of Pharmaceutical and Medical Chemistry, Westphalian Wilhelms-University Münster, Münster, 48149, Germany
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
4
|
Dong T, Zhang X, Yuan J, Lin Z, Yin P, Yu H, Wang M, Liu A. Sensitive Lateral Flow Immunoassay Based on Specific Peptide and Superior Oxidase Mimics with a Universal Dual-Mode Significant Signal Amplification. Anal Chem 2023; 95:12532-12540. [PMID: 37553756 DOI: 10.1021/acs.analchem.3c02821] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Rapid and sensitive antigen detection using a lateral flow immunoassay (LFIA) is crucial for diagnosing infectious diseases due to its simplicity, speed, and user-friendly features. However, it remains a critical issue to explore specific biorecognition elements and powerful signal amplification. In this study, taking SARS-CoV-2 as a proof of concept, a specific peptide, WFLNDSELIML, binding to the SARS-CoV-2 spike (S) antigen was identified by a nonamplified biopanning method, which exhibited high affinity to the target, with a dissociation constant of 9.29 ± 1.55 nM. Molecular docking analysis reveals that this peptide binds to the N-terminal domain of the SARS-CoV-2 S antigen. Then, using this peptide as a capture probe and angiotensin-converting enzyme 2 as a detection probe, a peptide-based lateral flow immunoassay (pLFIA) for the sensitive detection of the SARS-CoV-2 S antigen without any antibody was developed, for which a polydopamine nanosphere (PDA)@MnO2 nanocomposite with excellent oxidase-like activity was used as a colorimetric label, exhibiting dual-mode remarkable signal amplification of natural melanin and on-demand nanozyme catalytic enhancement. The PDA@MnO2-based pLFIA is capable of detecting the SARS-CoV-2 S antigen with a limit of detection of 8.01 pg/mL, which is 18.7 times lower than that of a conventional pLFIA tagged with gold nanoparticles. Additionally, the as-proposed PDA@MnO2-based pLFIA can detect up to 150 transduction units/mL SARS-CoV-2 pseudoviruses spiked in saliva samples. Given the outstanding analytical performance, the proposed PDA@MnO2-based pLFIA may offer a reliable option for the rapid diagnosis of SARS-CoV-2.
Collapse
Affiliation(s)
- Tao Dong
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
- School of Pharmacy, Medical College, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Xin Zhang
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Jinlong Yuan
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Ziting Lin
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
- Qingdao Hightop Biotech Co., Ltd., 369 Hedong Road, Hi-tech Industrial Development Zone, Qingdao 266112, China
| | - Pengxue Yin
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Haipeng Yu
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Mingyang Wang
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Aihua Liu
- Institute for Chemical Biology & Biosensing, College of Life Sciences, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| |
Collapse
|
5
|
Singpant P, Tubsuwan A, Sakdee S, Ketterman AJ, Jearawiriyapaisarn N, Kurita R, Nakamura Y, Songdej D, Tangprasittipap A, Bhukhai K, Chiangjong W, Hongeng S, Saisawang C. Recombinant Cas9 protein production in an endotoxin-free system and evaluation with editing the BCL11A gene in human cells. Protein Expr Purif 2023:106313. [PMID: 37276914 DOI: 10.1016/j.pep.2023.106313] [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: 04/20/2023] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 06/07/2023]
Abstract
Many therapeutic proteins are expressed in Escherichia coli bacteria for the low cost and high yield obtained. However, these gram-negative bacteria also generate undesirable endotoxin byproducts such as lipopolysaccharides (LPS). These endotoxins can induce a human immune response and cause severe inflammation. To mitigate this problem, we have employed the ClearColi BL21 (DE3) endotoxin-free cells as an expression host for Cas9 protein production. Cas9 is an endonuclease enzyme that plays a key role in the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR associated protein 9 (CRISPR/Cas9) genome editing technique. This technology is very promising for use in diagnostics as well as treatment of diseases, especially for genetic diseases such as thalassemia. The potential uses for this technology thus generate a considerable interest for Cas9 utilization as a therapeutic protein in clinical treatment. Therefore, special care in protein production should be a major concern. Accordingly, we expressed the Cas9 protein in endotoxin-free bacterial cells achieving 99% purity with activity comparable to commercially available Cas9. Our protocol therefore yields a cost-effective product suitable for invitro experiments with stem cells.
Collapse
Affiliation(s)
- Passanan Singpant
- Molecular Medical Biosciences Cluster, Institute of Molecular Biosciences, Mahidol University, 25/25 Putthamonthol Road 4, Salaya, Nakhon Pathom, 73170, Thailand
| | - Alisa Tubsuwan
- Molecular Medical Biosciences Cluster, Institute of Molecular Biosciences, Mahidol University, 25/25 Putthamonthol Road 4, Salaya, Nakhon Pathom, 73170, Thailand
| | - Somsri Sakdee
- Molecular Medical Biosciences Cluster, Institute of Molecular Biosciences, Mahidol University, 25/25 Putthamonthol Road 4, Salaya, Nakhon Pathom, 73170, Thailand
| | - Albert J Ketterman
- Molecular Medical Biosciences Cluster, Institute of Molecular Biosciences, Mahidol University, 25/25 Putthamonthol Road 4, Salaya, Nakhon Pathom, 73170, Thailand
| | - Natee Jearawiriyapaisarn
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Ryo Kurita
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Duantida Songdej
- Pediatric Hematology-Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Amornrat Tangprasittipap
- Office of Research, Academic Affairs and Innovations, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Kanit Bhukhai
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Wararat Chiangjong
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suradej Hongeng
- Pediatric Hematology-Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Chonticha Saisawang
- Molecular Medical Biosciences Cluster, Institute of Molecular Biosciences, Mahidol University, 25/25 Putthamonthol Road 4, Salaya, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
6
|
One-step immunoassay based on filtration for detection of food poisoning-related bacteria. Talanta 2023; 255:124203. [PMID: 36565526 DOI: 10.1016/j.talanta.2022.124203] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
A one-step immunoassay based on filtration was presented, which used microbeads for target analyte detection and filters with appropriate pore sizes to distinguish the complexity of target analyte and microbeads. For effective bacterial detection, the microbead size and the filter's pore size must be optimized. The optimal concentrations of the enzyme (urease) and antibody were determined at the maximum absorbance change, that is, the maximum pH change. The pH change was measured using a field-effect transistor (FET). The correlation between pH change and threshold voltage was estimated to be 21.7 mV/pH, and the correlation between pH change and the source-drain current was estimated to be -379 nA/pH. For the one-step immunoassay, antibodies against target bacteria were isolated from horse serum by filtration, and these antibodies were estimated to have a sufficiently high specificity to overcome cross-reactivity among five types of food poisoning-related bacteria: Escherichia coli O157, Salmonella typhimurium, Listeria monocytogenes, Bacillus cereus, and Staphylococcus aureus. Finally, the FET-based one-step immunoassay was demonstrated for five types of food poisoning-related bacteria in human serum.
Collapse
|
7
|
Hu Q, Feng W, Liang Y, Liang Z, Cao X, Li S, Luo Y, Wan J, Ma Y, Han D, Niu L. Boronate Affinity-Amplified Electrochemical Aptasensing of Lipopolysaccharide. Anal Chem 2022; 94:17733-17738. [PMID: 36475636 DOI: 10.1021/acs.analchem.2c05004] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
As lipopolysaccharide (LPS) is closely associated with sepsis and other life-threatening conditions, the point-of-care (POC) detection of LPS is of significant importance to human health. In this work, we illustrate an electrochemical aptasensor for the POC detection of low-abundance LPS by utilizing boronate affinity (BA) as a simple, efficient, and cost-effective amplification strategy. Briefly, the BA-amplified electrochemical aptasensing of LPS involves the tethering of the aptamer receptors and the BA-mediated direct decoration of LPS with redox signal tags. As the polysaccharide chain of LPS contains hundreds of cis-diol sites, the covalent crosslinking between the phenylboronic acid group and cis-diol sites can be harnessed for the site-specific decoration of each LPS with hundreds of redox signal tags, thereby enabling amplified detection. As it involves only a single-step operation (∼15 min), the BA-mediated signal amplification holds the significant advantages of unrivaled simplicity, rapidness, and cost-effectiveness over the conventional nanomaterial- and enzyme-based strategies. The BA-amplified electrochemical aptasensor has been successfully applied to specifically detect LPS within 45 min, with a detection limit of 0.34 pg/mL. Moreover, the clinical utility has been validated based on LPS detection in complex serum samples. As a proof of concept, a portable device has been developed to showcase the potential applicability of the BA-amplified electrochemical LPS aptasensor in the POC testing. In view of its simplicity, rapidness, and cost-effectiveness, the BA-amplified electrochemical LPS aptasensor holds broad application prospects in the POC testing.
Collapse
Affiliation(s)
- Qiong Hu
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Wenxing Feng
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Yiyi Liang
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Zhiwen Liang
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Xiaojing Cao
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Shiqi Li
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Yilin Luo
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Jianwen Wan
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Yingming Ma
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Dongxue Han
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Li Niu
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| |
Collapse
|
8
|
Sheng A, Yang J, Cheng L, Zhang J. Boronic Ester-Mediated Dual Recognition Coupled with a CRISPR/Cas12a System for Lipopolysaccharide Analysis. Anal Chem 2022; 94:12523-12530. [PMID: 36040369 DOI: 10.1021/acs.analchem.2c02776] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this work, boronic ester-mediated dual recognition has been coupled with a CRISPR/Cas12a system; thus, a new method for highly specific and sensitive detection of lipopolysaccharide (LPS) is proposed via the simultaneous recognition of boronic acid and an LPS aptamer (LPSA) as well as signal amplification by CRISPR/Cas12a. Specifically, boronic acid-modified magnetic beads (MB@APBA) and aptamers are employed for the simultaneous dual recognition of LPS, while polymerase isotherm amplification is further utilized to induce LPS cycling and form a double strand, which can activate the CRISPR/Cas12a system so as to amplify the signal. Consequently, a linear detection range can be obtained from 0.05 to 5000 ng/mL, with the lowest detection limit of 44.86 pg/mL. The capturing of MB@APBA on 1, 2- and 1, 3-cis dihydroxyl-containing substances can not only eliminate the interference of other molecules but also enhance the highly specific recognition of LPSA on LPS. Moreover, MB@APBA can be reused by adjusting the pH value of the reaction system. The method can be developed as a universal platform for the analytical detection of other carbohydrates.
Collapse
Affiliation(s)
- Anzhi Sheng
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, P. R. China.,Research Center of Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Jingyi Yang
- Research Center of Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Liangfen Cheng
- Research Center of Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Juan Zhang
- Research Center of Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
9
|
Zhang Y, Wu T, Liu D, Xu R, Ma H, Wei Q, Zhang Y. Photoelectrochemical immunosensor for the sensitive detection of neuron-specific enolase based on the effect of Z-scheme WO 3/NiCo 2O 4 nanoarrays p-n heterojunction. Biosens Bioelectron 2022; 213:114452. [PMID: 35679647 DOI: 10.1016/j.bios.2022.114452] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/20/2022] [Accepted: 06/01/2022] [Indexed: 11/26/2022]
Abstract
In this study, a signal-on type PEC immunosensor was constructed to detect neuron-specific enolase (NSE) via Z-scheme WO3/NiCo2O4 p-n heterojunction with cactus-like structure used as photoactive materials and MnxCd1-xS⊃Au NPs (MCS⊃Au NPs) as signal labels. Firstly, Z-scheme WO3/NiCo2O4 heterojunction could accelerate the separation efficiency of carriers and well-matched photoactive materials may promote charge migration, which resulted in WO3/NiCo2O4 generating strong and stable current. In addition, Z-scheme WO3/NiCo2O4 heterojunction directly grown on the surface of FTO via hydrothermal method facilitated the preparation of PEC immunosensor with outstanding stability. Secondly, an efficient signal amplification strategy was proposed by MnxCd1-xS⊃Au NPs incubating with signal antibody (Ab2). On the one hand, the well-matched energy levels of MnxCd1-xS with WO3/NiCo2O4 boosted the photo-generated electrons transferred to the electrode; on the other hand, the LSPR effect of Au may convert thermion to photocurrent to achieve signal amplification. Based on the above strategies, a PEC immunosensor with outstanding reproducibility and stability was obtained for sensitive detection of NSE. Under the optimum experimental conditions, current response range of the constructed signal amplification PEC sensor to NSE was 0.1 pg/mL ∼50 ng/mL and the detection limit was 0.07 pg/mL (S/N = 3). After the application tests in the detection of actual samples, the feasibility of the prepared PEC immunosensor with excellent selectivity, high sensitivity and satisfactory reproducibility was verified and the satisfactory results were obtained.
Collapse
Affiliation(s)
- Yingying Zhang
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, Shandong, China; Provincial Key Laboratory of Rural Energy Engineering in Yunnan, School of Energy and Environment Science, Yunnan Normal University, Kunming, 650500, China
| | - Tingting Wu
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, Shandong, China
| | - Deling Liu
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, Shandong, China
| | - Rui Xu
- Provincial Key Laboratory of Rural Energy Engineering in Yunnan, School of Energy and Environment Science, Yunnan Normal University, Kunming, 650500, China
| | - Hongmin Ma
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, Shandong, China.
| | - Qin Wei
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, Shandong, China
| | - Yong Zhang
- Provincial Key Laboratory of Rural Energy Engineering in Yunnan, School of Energy and Environment Science, Yunnan Normal University, Kunming, 650500, China.
| |
Collapse
|
10
|
Thorel M, Mateos-Hernandez L, Mulot B, Azzouni MN, Hodžić A, Gaillot H, Ruel Y, Desoubeaux G, Delaye JB, Obregon D, Wu-Chuang A, de la Fuente J, Bermúdez-Humarán LG, Risco-Castillo V, Leclerc A, Cabezas-Cruz A. Assessment of the Safety and Efficacy of an Oral Probiotic-Based Vaccine Against Aspergillus Infection in Captive-Bred Humboldt Penguins ( Spheniscus humboldti). Front Immunol 2022; 13:897223. [PMID: 35634323 PMCID: PMC9137413 DOI: 10.3389/fimmu.2022.897223] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Aspergillosis is a fungal infection caused mainly by Aspergillus fumigatus that often results in respiratory disease in birds. Aspergillosis is a major cause of morbidity and mortality in captive-bred penguin species. Currently, there is no registered vaccine to prevent aspergillosis. Recent research demonstrated that oral administration of gram-negative bacteria expressing high levels of galactose-α-1,3-galactose (α-Gal) modulates anti-α-Gal immunity and protects turkeys from clinical aspergillosis caused by experimental A. fumigatus infection. The role of anti-α-Gal immunity in penguins has not been studied. Here, we tested the distribution of α-1,3-galactosyltransferase (α1,3GT) genes in the fecal microbiome of Humboldt penguins (Spheniscus humboldti). The occurrence of natural anti-α-Gal antibodies (Abs) in sera and eggs of healthy Humboldt penguins was also assessed. A trial was then conducted to test whether oral administration of Escherichia coli Nissle, expressing high α-Gal levels, modulates anti-α-Gal immunity in a colony of Humboldt penguins. Animals in the vaccination and placebo groups were evaluated before the trial and followed for one year for aspergillosis detection using a diagnostic panel including computed tomography scans, capillary zone electrophoresis, 3-hydroxybutyrate levels, and anti-A. fumigatus Abs. Anti-α-Gal Abs were detected in sera (IgM and IgY) and eggs (IgY) of healthy penguins. Microbiota analysis and functional predictions revealed the presence of α1,3GT genes in the microbiota of Humboldt penguins and other penguin species. A strong decrease in anti-α-Gal IgM levels was observed in all animals in the placebo group three months after vaccination protocol. This decrease was not observed in E. coli Nissle-treated penguins. After the vaccination protocol, we found a positive correlation between anti-E. coli IgY and anti-α-Gal IgY in the E. coli Nissle group, suggesting a correlation between the presence of the bacteria and these Abs. During the study period, three penguins exhibited respiratory signs consistent with aspergillosis. Two were from the placebo group whose symptoms resolved with specific treatments, while a single vaccinated individual developed fatal respiratory aspergillosis eight months after the trial. We conclude that E. coli Nissle represents a safe potential probiotic with a protective effect against aspergillosis in Humboldt penguins that deserves to be further explored for therapeutic uses in these animals.
Collapse
Affiliation(s)
- Milan Thorel
- ZooParc de Beauval and Beauval Nature, Saint-Aignan-sur-Cher, France
| | - Lourdes Mateos-Hernandez
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Baptiste Mulot
- ZooParc de Beauval and Beauval Nature, Saint-Aignan-sur-Cher, France
| | - Mouna Naila Azzouni
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Adnan Hodžić
- Department of Pathobiology, Institute of Parasitology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Hugues Gaillot
- ADVETIA Veterinary Hospital Center, Vélizy-Villacoublay, France
| | - Yannick Ruel
- ADVETIA Veterinary Hospital Center, Vélizy-Villacoublay, France
| | - Guillaume Desoubeaux
- CHU de Tours, Service de Parasitologie, Mycologie, Médecine Tropicale, Tours, France
- Université de Tours, Inserm U1100 – Centre d’Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France
| | - Jean-Baptiste Delaye
- CHU de Tours, Pôle de Biologie médicale, Laboratoire de Médecine Nucléaire In Vitro – Centre Régional de Dépistage Néonatal, Tours, France
| | - Dasiel Obregon
- School of Environmental Sciences, University of Guelph, Guelph, ON, Canada
| | - Alejandra Wu-Chuang
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, United States
| | | | - Veronica Risco-Castillo
- EA 7380 Dynamyc, UPEC, USC, ANSES, Ecole nationale vétérinaire d’Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Antoine Leclerc
- ZooParc de Beauval and Beauval Nature, Saint-Aignan-sur-Cher, France
| | - Alejandro Cabezas-Cruz
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| |
Collapse
|
11
|
Richter R, Kamal MAM, Koch M, Niebuur B, Huber A, Goes A, Volz C, Vergalli J, Kraus T, Müller R, Schneider‐Daum N, Fuhrmann G, Pagès J, Lehr C. An Outer Membrane Vesicle-Based Permeation Assay (OMPA) for Assessing Bacterial Bioavailability. Adv Healthc Mater 2022; 11:e2101180. [PMID: 34614289 PMCID: PMC11468809 DOI: 10.1002/adhm.202101180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/14/2021] [Indexed: 11/11/2022]
Abstract
When searching for new antibiotics against Gram-negative bacterial infections, a better understanding of the permeability across the cell envelope and tools to discriminate high from low bacterial bioavailability compounds are urgently needed. Inspired by the phospholipid vesicle-based permeation assay (PVPA), which is designed to predict non-facilitated permeation across phospholipid membranes, outer membrane vesicles (OMVs) of Escherichia coli either enriched or deficient of porins are employed to coat filter supports for predicting drug uptake across the complex cell envelope. OMVs and the obtained in vitro model are structurally and functionally characterized using cryo-TEM, SEM, CLSM, SAXS, and light scattering techniques. In vitro permeability, obtained from the membrane model for a set of nine antibiotics, correlates with reported in bacterio accumulation data and allows to discriminate high from low accumulating antibiotics. In contrast, the correlation of the same data set generated by liposome-based comparator membranes is poor. This better correlation of the OMV-derived membranes points to the importance of hydrophilic membrane components, such as lipopolysaccharides and porins, since those features are lacking in liposomal comparator membranes. This approach can offer in the future a high throughput screening tool with high predictive capacity or can help to identify compound- and bacteria-specific passive uptake pathways.
Collapse
Affiliation(s)
- Robert Richter
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
| | - Mohamed A. M. Kamal
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| | - Marcus Koch
- INM – Leibniz Institute for New MaterialsCampus D2.2Saarbrücken66123Germany
| | - Bart‐Jan Niebuur
- INM – Leibniz Institute for New MaterialsCampus D2.2Saarbrücken66123Germany
| | - Anna‐Lena Huber
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| | - Adriely Goes
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| | - Carsten Volz
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
| | - Julia Vergalli
- UMR_MD1U‐1261Aix‐Marseille UniversitéINSERMIRBAMCTFaculté de Pharmacie27 Boulevard Jean MoulinMarseille13005France
| | - Tobias Kraus
- INM – Leibniz Institute for New MaterialsCampus D2.2Saarbrücken66123Germany
- Colloid and Interface ChemistrySaarland UniversityCampus D2.2Saarbrücken66123Germany
| | - Rolf Müller
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| | - Nicole Schneider‐Daum
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
| | - Gregor Fuhrmann
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| | - Jean‐Marie Pagès
- UMR_MD1U‐1261Aix‐Marseille UniversitéINSERMIRBAMCTFaculté de Pharmacie27 Boulevard Jean MoulinMarseille13005France
| | - Claus‐Michael Lehr
- Helmholtz Centre for Infection ResearchHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Saarland UniversityDepartment of PharmacyCampus E8.1Saarbrücken66123Germany
| |
Collapse
|
12
|
Park JH, Song Z, Bong JH, Kim HR, Kim MJ, Choi KH, Shin SS, Kang MJ, Lee DY, Pyun JC. Electrochemical One-Step Immunoassay Based on Switching Peptides and Pyrolyzed Carbon Electrodes. ACS Sens 2022; 7:215-224. [PMID: 34984905 DOI: 10.1021/acssensors.1c01998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Switching peptides were designed to bind reversibly to the binding pocket of antibodies (IgG) by interacting with frame regions (FRs). These peptides can be quantitatively released when antigens bind to IgG. As FRs have conserved amino acid sequences, switching peptides can be used as antibodies for different antigens and different source animals. In this study, an electrochemical one-step immunoassay was conducted using switching peptides labeled with ferrocene for the quantitative measurement of analytes. For the effective amperometry of the switching peptides labeled with ferrocene, a pyrolyzed carbon electrode was prepared by pyrolysis of the parylene-C film. The feasibility of the pyrolyzed carbon electrode for the electrochemical one-step immunoassay was determined by analyzing its electrochemical properties, such as its low double-layer capacitance (Cdl), high electron transfer rate (kapp), and wide electrochemical window. In addition, the factors influencing the amperometry of switching peptides labeled with ferrocene were analyzed according to the hydrodynamic radius, the number of intrahydrogen bonds, dipole moments, and diffusion coefficients. Finally, the applicability of the electrochemical one-step immunoassay for the medical diagnosis of the human hepatitis B surface antigen (hHBsAg) was assessed.
Collapse
Affiliation(s)
- Jun-Hee Park
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Zhiquan Song
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ji-Hong Bong
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hong-Rae Kim
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Moon-Ju Kim
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Kyung-Hak Choi
- OPTOLANE Technologies Inc., 20 Pangyoyeok-ro 241beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13494, Republic of Korea
| | - Seung-Shick Shin
- OPTOLANE Technologies Inc., 20 Pangyoyeok-ro 241beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13494, Republic of Korea
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Do Young Lee
- OPTOLANE Technologies Inc., 20 Pangyoyeok-ro 241beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13494, Republic of Korea
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
13
|
ClearColi as a platform for untagged pneumococcal surface protein A production: cultivation strategy, bioreactor culture, and purification. Appl Microbiol Biotechnol 2022; 106:1011-1029. [PMID: 35024919 PMCID: PMC8755982 DOI: 10.1007/s00253-022-11758-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 11/27/2022]
Abstract
Abstract
Several studies have searched for new antigens to produce pneumococcal vaccines that are more effective and could provide broader coverage, given the great number of serotypes causing pneumococcal diseases. One of the promising subunit vaccine candidates is untagged recombinant pneumococcal surface protein A (PspA4Pro), obtainable in high quantities using recombinant Escherichia coli as a microbial factory. However, lipopolysaccharides (LPS) present in E. coli cell extracts must be removed, in order to obtain the target protein at the required purity, which makes the downstream process more complex and expensive. Endotoxin-free E. coli strains, which synthesize a nontoxic mutant LPS, may offer a cost-effective alternative way to produce recombinant proteins for application as therapeutics. This paper presents an investigation of PspA4Pro production employing the endotoxin-free recombinant strain ClearColi® BL21(DE3) with different media (defined, auto-induction, and other complex media), temperatures (27, 32, and 37 °C), and inducers. In comparison to conventional E. coli cells in a defined medium, ClearColi presented similar PspA4Pro yields, with lower productivities. Complex medium formulations supplemented with salts favored PspA4Pro yields, titers, and ClearColi growth rates. Induction with isopropyl-β-d-thiogalactopyranoside (0.5 mM) and lactose (2.5 g/L) together in a defined medium at 32 °C, which appeared to be a promising cultivation strategy, was reproduced in 5 L bioreactor culture, leading to a yield of 146.0 mg PspA4Pro/g dry cell weight. After purification, the cell extract generated from ClearColi led to 98% purity PspA4Pro, which maintained secondary structure and biological function. ClearColi is a potential host for industrial recombinant protein production. Key points • ClearColi can produce as much PspA4Pro as conventional E. coli BL21(DE3) cells. • 10.5 g PspA4Pro produced in ClearColi bioreactor culture using a defined medium. • Functional PspA4Pro (98% of purity) was obtained in ClearColi bioreactor culture.Graphical abstract ![]() Supplementary Information The online version contains supplementary material available at 10.1007/s00253-022-11758-9.
Collapse
|
14
|
Capacitive biosensor based on vertically paired electrodes for the detection of SARS-CoV-2. Biosens Bioelectron 2022; 202:113975. [PMID: 35042131 PMCID: PMC8741629 DOI: 10.1016/j.bios.2022.113975] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/31/2021] [Accepted: 01/06/2022] [Indexed: 12/21/2022]
Abstract
Vertically paired electrodes (VPEs) with multiple electrode pairs were developed for the enhancement of capacitive measurements by optimizing the electrode gap and number of electrode pairs. The electrode was fabricated using a conductive polymer layer of PEDOT:PSS instead of Ag and Pt metal electrodes to increase the VPE fabrication yield because the PEDOT:PSS layer could be effectively etched using a reactive dry etching process. In this study, sensitivity enhancement was realized by decreasing the electrode gap and increasing the number of VPE electrode pairs. Such an increase in sensitivity according to the electrode gap and the number of electrode pairs was estimated using a model analyte for an immunoassay. Additionally, a computer simulation was performed using VPEs with different electrode gaps and numbers of VPE electrode pairs. Finally, VPEs with multiple electrode pairs were applied for SARS-CoV-2 nucleoprotein (NP) detection. The capacitive biosensor based on the VPE with immobilized anti-SARS-CoV-2 NP was applied for the specific detection of SARS-CoV-2 in viral cultures. Using viral cultures of SARS-CoV-2, SARS-CoV, MERS-CoV, and CoV-strain 229E, the limit of detection (LOD) was estimated to satisfy the cutoff value (dilution factor of 1/800) for the medical diagnosis of COVID-19, and the assay results from the capacitive biosensor were compared with commercial rapid kit based on a lateral flow immunoassay.
Collapse
|
15
|
Shao Y, Zhou H, Wu Q, Xiong Y, Wang J, Ding Y. Recent advances in enzyme-enhanced immunosensors. Biotechnol Adv 2021; 53:107867. [PMID: 34774928 DOI: 10.1016/j.biotechadv.2021.107867] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/31/2021] [Accepted: 11/05/2021] [Indexed: 12/19/2022]
Abstract
Among the products for rapid detection in different fields, enzyme-based immunosensors have received considerable attention. Recently, great efforts have been devoted to enhancing the output signals of enzymes through different strategies that can significantly improve the sensitivity of enzyme-based immunosensors for the need of practical applications. In this manuscript, the significance of enzyme-based signal transduction patterns in immunoassay and the central role of enzymes in achieving precise control of reaction systems are systematically described. In view of the rapid development of this field, we classify these strategies based on the combination of immune recognition and enzyme amplification into three categories, namely enzyme-based enhancement strategies, combination of the catalytic amplification of enzymes with other signal amplification methods, and substrate-based enhancement strategies. The current focus and future direction of enzyme-based immunoassays are also discussed. This article is not exhaustive, but focuses on the latest advances in different signal generation methods based on enzyme-initiated catalytic reactions and their applications in the detection field, which could provide an accessible introduction of enzyme-based immunosensors for the community with a view to further improving its application efficiency.
Collapse
Affiliation(s)
- Yanna Shao
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Huan Zhou
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qingping Wu
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yonghua Xiong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou 510432, China
| | - Yu Ding
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
16
|
Bong JH, Park JH, Sung JS, Lee CK, Lee GY, Kang MJ, Kim HO, Pyun JC. Rapid Analysis of Bacterial Contamination in Platelets without Pre-Enrichment Using Pig Serum-Derived Antibodies. ACS APPLIED BIO MATERIALS 2021; 4:7779-7789. [DOI: 10.1021/acsabm.1c00538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Ji-Hong Bong
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Korea
| | - Jun-Hee Park
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Korea
| | - Jeong Soo Sung
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Korea
| | - Chang Kyu Lee
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Korea
| | - Ga-Yeon Lee
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Korea
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Hyun Ok Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Korea
| |
Collapse
|
17
|
Low endotoxin E. coli strain-derived plasmids reduce rAAV vector-mediated immune responses both in vitro and in vivo. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 22:293-303. [PMID: 34485612 PMCID: PMC8403685 DOI: 10.1016/j.omtm.2021.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/10/2021] [Indexed: 11/24/2022]
Abstract
The major challenge of recombinant adeno-associated virus (rAAV) vectors is host immunological barriers. Compared to the neutralizing antibody and the cytotoxic T lymphocyte response, the host immune responses induced by unsatisfactory rAAV manufacturing were largely ignored previously. rAAV vector production usually requires large amounts of plasmid DNAs. The DNA are commonly isolated from the DH5α bacterial strain, which contains lipopolysaccharide (LPS) contamination. LPS, also named endotoxin, in plasmid DNA is intractable, and residual endotoxin in the subsequent rAAV vectors may result in substantial host immune response. Recently, a ClearColi K12 bacterial strain is commercially available, with genetically modified LPS that does not trigger endotoxic response in mammalian cells. Here, we produced rAAV-DJ vectors by plasmids yielded from either DH5α or ClearColi K12 bacterial strains. Our data indicated that the ClearColi K12 strain had satisfactory protection for the rAAV inverted terminal repeat (ITR) sequence. As expected, the ClearColi K12-derived rAAV-DJ vectors had lower endotoxin levels. The physical and biological equivalency of the purified viral stocks were confirmed by electron micrographs, Coomassie blue staining, and transduction assays. Most importantly, the ClearColi K12-derived rAAV-DJ vectors triggered reduced nuclear factor-kappa B (NF-κB) signaling pathway both in cell cultures in vitro and in C57BL/6 mice retinas in vivo. We believe that the use of the ClearColi K12 bacterial strain could eliminate the LPS in the purified vector stock at the source. Our data indicate its promising use in future clinical development.
Collapse
|
18
|
Suhito IR, Angeline N, Lee KH, Kim H, Park CG, Luo Z, Kim TH. A Spheroid-Forming Hybrid Gold Nanostructure Platform That Electrochemically Detects Anticancer Effects of Curcumin in a Multicellular Brain Cancer Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2002436. [PMID: 32954643 DOI: 10.1002/smll.202002436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/27/2020] [Indexed: 05/26/2023]
Abstract
In this study, a multifunctional platform that enables the highly efficient formation of 3D multicellular cancer spheroids and precise real-time assessments of the anticancer effects of curcumin in a brain tumor coculture model is reported. A highly conductive gold nanostructure (HCGN) is fabricated to facilitate cancer spheroid formation without using anti-cell adhesion molecules. A neuroblastoma (SH-SY5Y) and glioblastoma (U-87MG) coculture model is generated on HCGN with a specific cell-to-cell ratio (SH-SY5Y: U-87MG = 1:1), and their redox behaviors are successfully measured without destroying the distinct 3D structure of the multicellular spheroids. Using electrochemical signals as an indicator of spheroid viability, the effects of potential anticancer compounds on cocultured spheroids are further assessed. Remarkably, decreased cell viability in 3D spheroids caused by a low concentration of curcumin (30 µM) is detectable using the electrochemical method (29.4%) but not with a conventional colorimetric assay (CCK-8). The detection is repeated more than ten times for both short- (63 h) and long-term cultivation (144 h) without damaging the spheroids, enabling real-time, non-destructive pharmacokinetic analysis of various drug candidates. Therefore, it can be concluded that the hybrid platform is a highly promising, precise, and high-throughput drug screening tool based on 3D cell cultivation.
Collapse
Affiliation(s)
- Intan Rosalina Suhito
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Novi Angeline
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Kwang-Ho Lee
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Huijung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Zhengtang Luo
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Kowloon, Hong Kong, 999077, China
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
- Integrative Research Center for Two-Dimensional Functional Materials, Institute of Interdisciplinary Convergence Research, Chung-Ang University, Seoul, 06974, Republic of Korea
| |
Collapse
|
19
|
Anti-SARS-CoV-2 Nucleoprotein Antibodies Derived from Pig Serum with a Controlled Specificity. BIOCHIP JOURNAL 2021. [DOI: 10.1007/s13206-021-00019-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
20
|
Microbial biosensor for Salmonella using anti-bacterial antibodies isolated from human serum. Enzyme Microb Technol 2020; 144:109721. [PMID: 33541568 DOI: 10.1016/j.enzmictec.2020.109721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 11/22/2022]
Abstract
In this work, we present a novel microbial biosensor for Salmonella based on impedance spectrometry by using isolated antibodies against a specific bacterial strain from human serum. Anti-Salmonella (or BL21(DE3)) antibodies were isolated from human serum using S. enteritidis (or BL21(DE3)) and the mutant strain ClearColi. After the purification steps, the purification yield of the antibodies was calculated to be 0.2 %. From the FACS analysis, the isolated anti-Salmonella antibodies were estimated to have more than 6-fold higher binding affinity for S. enteritidis compared to antibodies against other kinds of Gram-negative bacterial strains, including HB101, ClearColi, JM110, DH5α, and BL21(DE3). Finally, the anti-Salmonella antibodies isolated herein were used for bacterial detection using electrochemical biosensors based on impedance spectrometry and the Rct value of the antibodies was estimated for S. enteritidis from the Nyquist plot. The limit of detection of the isolated anti-Salmonella antibodies was estimated to be 1.0 × 103 cells/mL for S. enteritidis and 1.0 × 106 cells/mL for BL21(DE3), respectively.
Collapse
|
21
|
Pig Sera-derived Anti-SARS-CoV-2 Antibodies in Surface Plasmon Resonance Biosensors. BIOCHIP JOURNAL 2020; 14:358-368. [PMID: 33133395 PMCID: PMC7590559 DOI: 10.1007/s13206-020-4404-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 09/18/2020] [Indexed: 12/30/2022]
Abstract
Anti-coronavirusdisease-2019 (COVID-19; anti-severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2)) antibodies against nucleoprotein (NP) were purified from pig sera. Following the separation of the antibody fraction using a protein-A column, the final yield of the purified antibodies against SARS-CoV-2 NPs was estimated to be 0.26 ± 0.05 % (absolute amount of 143.4 ± 25.2 ng, n=5) from 1 mL of pig sera. The binding activities of the isolated antibodies were confirmed using immunoassay and immunostaining. Based on the specific binding activity to NPs, a quantitative assay was performed using a surface plasmon resonance (SPR) biosensor. From the doseresponse curve, the binding constant (Kd) was calculated to be 185 pM and the limit of detection was estimated to be 1.02 pM. The SPR biosensor with the isolated antibodies against SARS-CoV-2 NPs was applied for the detection of SARS-CoV-2, MERS-CoV, and CoV strain 229E in culture fluid.
Collapse
|
22
|
Bong JH, Kim HR, Yoo JW, Kang MJ, Shin MG, Lee JS, Shim WB, Lee SD, Pyun JC. One-step immunoassay without washing steps for influenza A virus detection using ISFET. Biosens Bioelectron 2020; 165:112341. [DOI: 10.1016/j.bios.2020.112341] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 01/07/2023]
|
23
|
Kim HR, Bong JH, Jung J, Sung JS, Kang MJ, Park JG, Pyun JC. An On-chip Chemiluminescent Immunoassay for Bacterial Detection using in Situ-synthesized Cadmium Sulfide Nanowires with Passivation Layers. BIOCHIP JOURNAL 2020. [DOI: 10.1007/s13206-020-4305-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
24
|
Lee GY, Bong JH, Jung J, Kang MJ, Jose J, Pyun JC. Application of a thermophoretic immunoassay in the diagnosis of lupus using outer membrane particles from E. coli. Biosens Bioelectron 2020; 156:112110. [PMID: 32174550 DOI: 10.1016/j.bios.2020.112110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/02/2020] [Accepted: 02/18/2020] [Indexed: 10/24/2022]
Abstract
Thermophoresis is the physical diffusion of molecules from hot to cold induced by a thermal gradient. Thermophoresis has been used to evaluate the interaction of biomolecules in solution. In this study, the outer membrane from E. coli was isolated and used to produce OM particles with a diameter of approximately 100 nm. These prepared OM particles were applied in a thermophoretic immunoassay. First, outer membrane (OM) particles with lipopolysaccharides (LPS) and anti-LPS antibodies were used as a model to demonstrate proof of concept and the difference in E. coli thermophoresis was explained by the changes in the molecular surface area (A) and effective charge (σeff). The hydrodynamic size of the molecules was measured as a changing parameter, molecular surface area (A), by dynamic laser scattering (DLS), and the zeta potential was measured as a changing parameter of effective charge (σeff) and then evaluated by the Soret equation. Using the hydrodynamic size and zeta potential values, the interaction between the antigen (OM particle with LPS) and antibody (anti-LPS antibodies) could be monitored and the results were fitted to the thermophoretic immunoassay using the Soret coefficient and equation. Finally, this OM-based immunoassay was applied to the medical diagnosis of systemic lupus erythematosus. Here, OM particles with Ro and La proteins were used to analyze the autoantibodies in patient and control sera. Thermophoretic immunoassay results were also compared to the fitted analysis using hydrodynamic size and zeta potential values and the Soret coefficient and equation.
Collapse
Affiliation(s)
- Ga-Yeon Lee
- Department of Materials Science and Engineering, Yonsei University, 134 Shinchon-dong, Seodaemun-gu, Seoul, 120-749, South Korea
| | - Ji-Hong Bong
- Department of Materials Science and Engineering, Yonsei University, 134 Shinchon-dong, Seodaemun-gu, Seoul, 120-749, South Korea
| | - Jaeyong Jung
- Department of Materials Science and Engineering, Yonsei University, 134 Shinchon-dong, Seodaemun-gu, Seoul, 120-749, South Korea
| | - Min-Jung Kang
- Korea Institute of Science and Technology, Seoul, South Korea
| | - Joachim Jose
- Institute of Pharmaceutical and Medical Chemistry, University of Münster, Münster, Germany
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 134 Shinchon-dong, Seodaemun-gu, Seoul, 120-749, South Korea.
| |
Collapse
|