1
|
Lyu Y, Zhang B, Chai Y, Zhang J, Wang L, Xiao Y, Cheng B, Qian C, Yang H, Li H, Tan X. A Quantitative First Passage Time Model for Tubular Microfluidic Immunoassays. ACS Sens 2025; 10:1387-1397. [PMID: 39883891 DOI: 10.1021/acssensors.4c03336] [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] [Indexed: 02/01/2025]
Abstract
Solid-phase immunosorbent reactions, such as ELISA, are widely used for detecting, identifying, and quantifying protein markers. However, traditional centimeter scale well-based immunoreactors suffer from low surface-to-volume (S/V) ratios, leading to large sample consumption and a long assay time. Microfluidic technologies, particularly tubular microfluidic immunoreactors, have emerged as promising alternatives due to their high S/V ratios. Despite experimental advancements, multifactor theoretical studies on tubular microfluidic systems are limited. In this study, we present a theoretical model based on the first passage time method to analyze diffusion-controlled reaction kinetics in tubular microfluidic immunoreactors. We focus on key parameters including binding kinetics, reactor size, and solution viscosity. To validate the model, controlled laboratory experiments were conducted using our in-house developed tip optofluidic immunoassay (TOI). These experimental results confirmed the reliability of theoretical models in the behavior prediction of tubular microfluidic systems under real-world conditions. Our model revealed that accurate and rapid protein biomarker quantification requires not only the development of microscale bioreactors but also the design of next-generation probes with extraordinary binding affinity and specificity. This work offers insights into optimizing critical design parameters in future microfluidic immunoassay development, paving ways for next generation microliter-sized biomolecular analysis.
Collapse
Affiliation(s)
- Yingkai Lyu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- National Innovation Center for Advanced Medical Devices, Shenzhen 518110, China
| | - Binmao Zhang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Department of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Yujuan Chai
- Department of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Jie Zhang
- Beijing Key Laboratory of Monoclonal Antibody Research and Development, Sino Biological Inc., Beijing 100176, China
| | - Li Wang
- Beijing Key Laboratory of Monoclonal Antibody Research and Development, Sino Biological Inc., Beijing 100176, China
| | - Yujin Xiao
- Shenzhen YHLO Biotech Co., Shenzhen 518116, China
| | | | - Chungen Qian
- Shenzhen YHLO Biotech Co., Shenzhen 518116, China
| | - Hui Yang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- State Key Laboratory of Biomedical Imaging Science and System, Shenzhen 518055, China
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hao Li
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- National Innovation Center for Advanced Medical Devices, Shenzhen 518110, China
| | - Xiaotian Tan
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- State Key Laboratory of Biomedical Imaging Science and System, Shenzhen 518055, China
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
2
|
Wang R, Xue J, Wei G, Zhang Y, Wang C, Li J, Geng X, Ostovan A, Chen L, Song Z. Fast and Sensitive Detection of Anti-SARS-CoV-2 IgG Using SiO 2@Au@CDs Nanoparticle-Based Lateral Flow Immunoassay Strip Coupled with Miniaturized Fluorimeter. Biomolecules 2024; 14:1568. [PMID: 39766275 PMCID: PMC11673715 DOI: 10.3390/biom14121568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
The development of a novel strategy for the measurement of SARS-CoV-2 IgG antibodies is of vital significance for COVID-19 diagnosis and effect of vaccination evaluation. In this investigation, an SiO2@Au@CDs nanoparticle (NP)-based lateral flow immunoassay (LFIA) strip was fabricated and coupled with a miniaturized fluorimeter. The morphology features and particle sizes of the SiO2@Au@CDs NPs were characterized carefully, and the results indicated that the materials possess monodisperse, uniform, and spherical structures. Finally, this system was employed for SARS-CoV-2 IgG antibody test. In this work, the strategy for the SARS-CoV-2 IgG antibody test possesses several merits, such as speed (less than 15 min), high sensitivity (1.2 × 10-7 mg/mL), broad linearity range (7.4 × 10-7~7.4 × 10-4 mg/mL), accurate results, high selectivity, good stability, and low cost. Additionally, future trends in LFAs using quantum dot-based diagnostics are envisioned.
Collapse
Affiliation(s)
- Rui Wang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China; (R.W.); (J.X.); (G.W.); (Y.Z.)
| | - Junping Xue
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China; (R.W.); (J.X.); (G.W.); (Y.Z.)
| | - Guo Wei
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China; (R.W.); (J.X.); (G.W.); (Y.Z.)
| | - Yimeng Zhang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China; (R.W.); (J.X.); (G.W.); (Y.Z.)
| | - Chuanliang Wang
- Department of Instrumentation & Analytical Chemistry, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Key Laboratory of Deep-Sea Composition Detection Technology of Liaoning Province, Dalian Institute of Chemical Physics, CAS, 457 Zhongshan Road, Dalian 116023, China; (C.W.); (X.G.)
| | - Jinhua Li
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Research Center for Coastal Environmental Engineering and Technology, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; (J.L.); (A.O.); (L.C.)
| | - Xuhui Geng
- Department of Instrumentation & Analytical Chemistry, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Key Laboratory of Deep-Sea Composition Detection Technology of Liaoning Province, Dalian Institute of Chemical Physics, CAS, 457 Zhongshan Road, Dalian 116023, China; (C.W.); (X.G.)
| | - Abbas Ostovan
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Research Center for Coastal Environmental Engineering and Technology, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; (J.L.); (A.O.); (L.C.)
| | - Lingxin Chen
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Research Center for Coastal Environmental Engineering and Technology, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; (J.L.); (A.O.); (L.C.)
| | - Zhihua Song
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, China; (R.W.); (J.X.); (G.W.); (Y.Z.)
| |
Collapse
|
3
|
Lv H, Shi F, Yin H, Jiao Y, Wei P. Development of a double-antibody sandwich ELISA for detection of SARS-CoV-2 variants based on nucleocapsid protein-specific antibodies. Microbiol Immunol 2024; 68:393-398. [PMID: 39287179 DOI: 10.1111/1348-0421.13173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
The COVID-19 pandemic, driven by the SARS-CoV-2 virus, has posed a severe threat to global public health. Rapid, reliable, and easy-to-use detection methods for SARS-CoV-2 variants are critical for effective epidemic prevention and control. The N protein of SARS-CoV-2 serves as an ideal target for antigen detection. In this study, we achieved soluble expression of the recombinant SARS-CoV-2 N protein using an Escherichia coli expression system and generated specific monoclonal antibodies by immunizing BALB/c mice. We successfully developed 10 monoclonal antibodies against the N protein, designated 5B7, 5F2-C11, 5E2-E8, 6C3-D8, 7C8, 9F2-E9, 12H5-D11, 13G2-C10, 14E9-F6, and 15H3-E10. Using these antibodies, we established a sandwich ELISA with 6C3-D8 as the capture antibody and 5F2-C11 as the detection antibody. The assay demonstrated a sensitivity of 0.78 ng/mL and showed no cross-reactivity with MERS-CoV, HCoV-OC43, HCoV-NL63, and HCoV-229E. Furthermore, this method successfully detected both wild-type SARS-CoV-2 and its variants, including Alpha, Beta, Delta, and Omicron. These findings indicate that our sandwich ELISA exhibits excellent sensitivity, specificity, and broad-spectrum applicability, providing a robust tool for detecting SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Hai Lv
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Fengjuan Shi
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Province Center for Disease Control and Prevention, Nanjing, China
| | - Huimin Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Yongjun Jiao
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Province Center for Disease Control and Prevention, Nanjing, China
| | - Pingmin Wei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
4
|
Ye XF, Huang ZP, Li MM, Liu SF, Huang WL, Hamud AMS, Ye LC, Li LY, Wu SJ, Zhuang JL, Chen YH, Chen XR, Lin S, Wei XF, Chen CN. Update on aquaporin-4 antibody detection: the early diagnosis of neuromyelitis optica spectrum disorders. Mult Scler Relat Disord 2024; 90:105803. [PMID: 39128164 DOI: 10.1016/j.msard.2024.105803] [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: 03/17/2024] [Revised: 07/06/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune-mediated primary inflammatory myelinopathy of the central nervous system that primarily affects the optic nerve and spinal cord. The aquaporin 4 antibody (AQP4-Ab) is a specific autoantibody marker for NMOSD. Most patients with NMOSD are seropositive for AQP4-Ab, thus aiding physicians in identifying ways to treat NMOSD. AQP4-Ab has been tested in many clinical and laboratory studies, demonstrating effectiveness in diagnosing NMOSD. Recently, novel assays have been developed for the rapid and accurate detection of AQP4-Ab, providing further guidance for the diagnosis and treatment of NMOSD. This article summarizes the importance of rapid and accurate diagnosis for treating NMOSD based on a review of the latest relevant literature. We discussed current challenges and methods for improvement to offer new ideas for exploring rapid and accurate AQP4-Ab detection methods, aiming for early diagnosis of NMOSD.
Collapse
Affiliation(s)
- Xiao-Fang Ye
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China; The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000Fujian Province, China
| | - Zheng-Ping Huang
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China; The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000Fujian Province, China
| | - Mi-Mi Li
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China; The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000Fujian Province, China
| | - Shu-Fen Liu
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China; The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000Fujian Province, China
| | - Wan-Li Huang
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China; The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000Fujian Province, China
| | - Abdullahi Mukhtar Sheik Hamud
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China; The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000Fujian Province, China
| | - Li-Chao Ye
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China; The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000Fujian Province, China
| | - Lin-Yi Li
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China; The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000Fujian Province, China
| | - Shu-Juan Wu
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China; The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000Fujian Province, China
| | - Jian-Long Zhuang
- Prenatal Diagnosis Centre, Quanzhou Women's and Children's Hospital, Quanzhou 362000, Fujian China
| | - Yan-Hong Chen
- Department of Neurology, Shishi General Hospital, Quanzhou 362000, Fujian Province, China
| | - Xiang-Rong Chen
- The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000Fujian Province, China; Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian Province, China; Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia.
| | - Xiao-Feng Wei
- College of Materials Science and Engineering, Huaqiao University, Xiamen 361021, Fujian Province, China.
| | - Chun-Nuan Chen
- Department of Neurology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China; The Second Clinical Medical College of Fujian Medical University, Quanzhou 362000Fujian Province, China.
| |
Collapse
|
5
|
Wang Y, Chen J, Zhang Y, Yang Z, Zhang K, Zhang D, Zheng L. Advancing Microfluidic Immunity Testing Systems: New Trends for Microbial Pathogen Detection. Molecules 2024; 29:3322. [PMID: 39064900 PMCID: PMC11279515 DOI: 10.3390/molecules29143322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Pathogenic microorganisms play a crucial role in the global disease burden due to their ability to cause various diseases and spread through multiple transmission routes. Immunity tests identify antigens related to these pathogens, thereby confirming past infections and monitoring the host's immune response. Traditional pathogen detection methods, including enzyme-linked immunosorbent assays (ELISAs) and chemiluminescent immunoassays (CLIAs), are often labor-intensive, slow, and reliant on sophisticated equipment and skilled personnel, which can be limiting in resource-poor settings. In contrast, the development of microfluidic technologies presents a promising alternative, offering automation, miniaturization, and cost efficiency. These advanced methods are poised to replace traditional assays by streamlining processes and enabling rapid, high-throughput immunity testing for pathogens. This review highlights the latest advancements in microfluidic systems designed for rapid and high-throughput immunity testing, incorporating immunosensors, single molecule arrays (Simoas), a lateral flow assay (LFA), and smartphone integration. It focuses on key pathogenic microorganisms such as SARS-CoV-2, influenza, and the ZIKA virus (ZIKV). Additionally, the review discusses the challenges, commercialization prospects, and future directions to advance microfluidic systems for infectious disease detection.
Collapse
Affiliation(s)
- Yiran Wang
- Engineering Research Center of Optical Instrument and System, The Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jingwei Chen
- Engineering Research Center of Optical Instrument and System, The Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yule Zhang
- Engineering Research Center of Optical Instrument and System, The Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Zhijin Yang
- Engineering Research Center of Optical Instrument and System, The Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Kaihuan Zhang
- 2020 X-Lab, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China
| | - Dawei Zhang
- Engineering Research Center of Optical Instrument and System, The Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Engineering Research Center of Environmental Biosafety Instruments and Equipment, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Institute of Intelligent Science and Technology, Tongji University, Shanghai 200092, China
| | - Lulu Zheng
- Engineering Research Center of Optical Instrument and System, The Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
- Shanghai Engineering Research Center of Environmental Biosafety Instruments and Equipment, University of Shanghai for Science and Technology, Shanghai 200093, China
| |
Collapse
|
6
|
Chen C, Porter R, Zhou X, Snozek CL, Yang EH, Wang S. Microfluidic Digital Immunoassay for Point-of-Care Detection of NT-proBNP from Whole Blood. Anal Chem 2024; 96:10569-10576. [PMID: 38877973 DOI: 10.1021/acs.analchem.4c01046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
The high prevalence and economic burden of heart failure remain a challenge to global health. This lifelong disease leads to a buildup of permanent heart damage, making early detection and frequent monitoring crucial for effective treatment. N-terminal proBNP (NT-proBNP) is an important biomarker for monitoring the disease state, but current commercial and research NT-proBNP assays require phlebotomy and bulky equipment or do not satisfy clinical requirements such as sensitivity and detection thresholds. Here, we report a point-of-care (POC) compatible microfluidic digital immunoassay that can quantify the NT-proBNP concentration in a small volume of whole blood. Our automated microfluidic device takes whole blood samples mixed with biotinylated detection antibodies and passes through a plasma filter to react with a capture antibody-functionalized sensor surface. Streptavidin-coated gold nanoparticles (GNPs) are then released to mark the surface-bound single NT-proBNP immunocomplexes and recorded with bright-field microscopy. NT-proBNP concentrations in the sample are quantified via a hybrid digital/analog calibration curve. Digital counts of bound GNPs are used as readout signal at low concentrations for high sensitivity detection, and GNP pixel occupancies are used at high concentrations for extended dynamic range. With this approach, we detected NT-proBNP in the range of 8.24-10 000 pg/mL from 7 μL of whole blood in 10 min, with a limit of detection of 0.94 pg/mL. Finally, the method was validated with 15 clinical serum samples, showing excellent linear correlation (r = 0.998) with Roche's Elecsys proBNP II assay. This evidence indicates that this method holds promise for decentralized monitoring of heart failure.
Collapse
Affiliation(s)
- Chao Chen
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
- Center for Biosensors and Bioelectronics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Ryan Porter
- Center for Biosensors and Bioelectronics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
- School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Xiaoyan Zhou
- Center for Biosensors and Bioelectronics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
- School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Christine Lh Snozek
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Phoenix, Arizona 85054, United States
| | - Eric H Yang
- Department of Cardiovascular Disease, Mayo Clinic Arizona, Phoenix, Arizona 85054, United States
| | - Shaopeng Wang
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
- Center for Biosensors and Bioelectronics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
7
|
Mortelmans T, Marty B, Kazazis D, Padeste C, Li X, Ekinci Y. Three-Dimensional Microfluidic Capillary Device for Rapid and Multiplexed Immunoassays in Whole Blood. ACS Sens 2024; 9:2455-2464. [PMID: 38687557 PMCID: PMC11129352 DOI: 10.1021/acssensors.4c00153] [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: 01/21/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
In this study, we demonstrate whole blood immunoassays using a microfluidic device optimized for conducting rapid and multiplexed fluorescence-linked immunoassays. The device is capable of handling whole blood samples without any preparatory treatment. The three-dimensional channels in poly(methyl methacrylate) are designed to passively load bodily fluids and, due to their linearly tapered profile, facilitate size-dependent immobilization of biofunctionalized particles. The channel geometry is optimized to allow for the unimpeded flow of cellular constituents such as red blood cells (RBCs). Additionally, to make the device easier to operate, the biofunctionalized particles are pretrapped in a first step, and the channel is dried under vacuum, after which it can be loaded with the biological sample. This novel approach and design eliminated the need for traditionally laborious steps such as filtering, incubation, and washing steps, thereby substantially simplifying the immunoassay procedures. Moreover, by leveraging the shallow device dimensions, we show that sample loading to read-out is possible within 5 min. Our results also show that the presence of RBCs does not compromise the sensitivity of the assays when compared to those performed in a pure buffer solution. This highlights the practical adaptability of the device for simple and rapid whole-blood assays. Lastly, we demonstrate the device's multiplexing capability by pretrapping particles of different sizes, each functionalized with a different antigen, thus enabling the performance of multiplexed on-chip whole-blood immunoassays, showcasing the device's versatility and effectiveness toward low-cost, simple, and multiplexed sensing of biomarkers and pathogens directly in whole blood.
Collapse
Affiliation(s)
- Thomas Mortelmans
- Laboratory for X-ray Nanoscience and Technologies, 5232 Villigen, Switzerland
- Swiss Nanoscience Institute, University of Basel, 4056 Basel, Switzerland
| | - Balz Marty
- Laboratory for X-ray Nanoscience and Technologies, 5232 Villigen, Switzerland
| | - Dimitrios Kazazis
- Laboratory for X-ray Nanoscience and Technologies, 5232 Villigen, Switzerland
| | - Celestino Padeste
- Laboratory of Nanoscale Biology, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Xiaodan Li
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Yasin Ekinci
- Laboratory for X-ray Nanoscience and Technologies, 5232 Villigen, Switzerland
| |
Collapse
|
8
|
Huang Y, Chen J, Chen S, Huang C, Li B, Li J, Jin Z, Zhang Q, Pan P, Du W, Liu L, Liu Z. Molecular characterization of SARS-CoV-2 nucleocapsid protein. Front Cell Infect Microbiol 2024; 14:1415885. [PMID: 38846351 PMCID: PMC11153676 DOI: 10.3389/fcimb.2024.1415885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 06/09/2024] Open
Abstract
Corona Virus Disease 2019 (COVID-19) is a highly prevalent and potent infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Until now, the world is still endeavoring to develop new ways to diagnose and treat COVID-19. At present, the clinical prevention and treatment of COVID-19 mainly targets the spike protein on the surface of SRAS-CoV-2. However, with the continuous emergence of SARS-CoV-2 Variants of concern (VOC), targeting the spike protein therapy shows a high degree of limitation. The Nucleocapsid Protein (N protein) of SARS-CoV-2 is highly conserved in virus evolution and is involved in the key process of viral infection and assembly. It is the most expressed viral structural protein after SARS-CoV-2 infection in humans and has high immunogenicity. Therefore, N protein as the key factor of virus infection and replication in basic research and clinical application has great potential research value. This article reviews the research progress on the structure and biological function of SARS-CoV-2 N protein, the diagnosis and drug research of targeting N protein, in order to promote researchers' further understanding of SARS-CoV-2 N protein, and lay a theoretical foundation for the possible outbreak of new and sudden coronavirus infectious diseases in the future.
Collapse
Affiliation(s)
- Yanping Huang
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Junkai Chen
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Siwei Chen
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Congcong Huang
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Bei Li
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Jian Li
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Zhixiong Jin
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Qiwei Zhang
- Central Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Pan Pan
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Weixing Du
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Long Liu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Zhixin Liu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
9
|
Shen Q, Hossain F, Fang C, Shu T, Zhang X, Law JLM, Logan M, Houghton M, Tyrrell DL, Joyce MA, Serpe MJ. Bovine Serum Albumin-Protected Gold Nanoclusters for Sensing of SARS-CoV-2 Antibodies and Virus. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37314985 DOI: 10.1021/acsami.3c03705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
An approach to assess severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection (and past infection) was developed. For virus detection, the SARS-CoV-2 virus nucleocapsid protein (NP) was targeted. To detect the NP, antibodies were immobilized on magnetic beads to capture the NPs, which were subsequently detected using rabbit anti-SARS-CoV-2 nucleocapsid antibodies and alkaline phosphatase (AP)-conjugated anti-rabbit antibodies. A similar approach was used to assess SARS-CoV-2-neutralizing antibody levels by capturing spike receptor-binding domain (RBD)-specific antibodies utilizing RBD protein-modified magnetic beads and detecting them using AP-conjugated anti-human IgG antibodies. The sensing mechanism for both assays is based on cysteamine etching-induced fluorescence quenching of bovine serum albumin-protected gold nanoclusters where cysteamine is generated in proportion to the amount of either SARS-CoV-2 virus or anti-SARS-CoV-2 receptor-binding domain-specific immunoglobulin antibodies (anti-RBD IgG antibodies). High sensitivity can be achieved in 5 h 15 min for the anti-RBD IgG antibody detection and 6 h 15 min for virus detection, although the assay can be run in "rapid" mode, which takes 1 h 45 min for the anti-RBD IgG antibody detection and 3 h 15 min for the virus. By spiking the anti-RBD IgG antibodies and virus in serum and saliva, we demonstrate that the assay can detect the anti-RBD IgG antibodies with a limit of detection (LOD) of 4.0 and 2.0 ng/mL in serum and saliva, respectively. For the virus, we can achieve an LOD of 8.5 × 105 RNA copies/mL and 8.8 × 105 RNA copies/mL in serum and saliva, respectively. Interestingly, this assay can be easily modified to detect myriad analytes of interest.
Collapse
Affiliation(s)
- Qiming Shen
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Faisal Hossain
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
- Department of Chemistry, Faculty of Science, University of Chittagong, Chattogram 4331, Bangladesh
| | - Changhao Fang
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Tong Shu
- Guangdong Laboratory of Artificial Intelligence and Digital Economy (SZ), Shenzhen Key Laboratory for Nano-Biosensing Technology, Research Center for Biosensor and Nanotheranostic, International Health Science Innovation Center, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518060, P. R. China
| | - Xueji Zhang
- Guangdong Laboratory of Artificial Intelligence and Digital Economy (SZ), Shenzhen Key Laboratory for Nano-Biosensing Technology, Research Center for Biosensor and Nanotheranostic, International Health Science Innovation Center, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518060, P. R. China
| | - John Lok Man Law
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Michael Logan
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Michael Houghton
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - D Lorne Tyrrell
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Michael A Joyce
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Michael J Serpe
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| |
Collapse
|
10
|
Zhu L, Ma J, Jin D, Zhang Y, Wu S, Xu A, Gu Y, An Y, Miao Y. Flower-like WSe 2 used as bio-matrix in ultrasensitive label-free electrochemical immunosensor for human immunoglobulin G determination. ANAL SCI 2023:10.1007/s44211-023-00351-3. [PMID: 37227625 DOI: 10.1007/s44211-023-00351-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 04/16/2023] [Indexed: 05/26/2023]
Abstract
The abnormal concentrations of human immunoglobulin G (hIgG) refers to many kinds of diseases. Analytical methods with the characteristics of rapid response, easy operation and high sensitivity should be designed to accurately determinate the hIgG levels in human serum. In this work, a label-free electrochemical immunosensor based on WSe2/rGO was developed to sensitively detect human immunoglobulin G. First, the flower-like transition metal dichalcogenides (TMDCs) Tungsten Diselenide (WSe2) with large effective specific surface area and porous structure was synthesized by hydrothermal synthesis. As a bio-matrix, the flower-like WSe2 efficiently increased the active sites for loading antibodies. Meanwhile, reduced graphene oxide (rGO) obtained by tannic acid reduction was used to improve the current response of the sensing interface. WSe2 was combined with rGO and the electrochemical active surface area (ECSA) of the sensing interface was enlarged to 2.1 times that of GCE. Finally, the combination of flower-like WSe2 and rGO broadened the detection range and reduced the detection limit of the sensing platform. The immunosensor exhibited a high sensitivity with a wide linear range of 0.01-1000 ng/mL and low detection limit of 4.72 pg/mL. The real sample analysis of hIgG were conducted under optimal conditions, and the spiked recovery rates were between 95.5 and 104.1%. Moreover, satisfactory results were obtained by testing the stability, specificity and reproducibility of the immunosensor. Therefore, it can be concluded that the as-proposed immunosensor has the application potential of clinical analyze of hIgG in human serum.
Collapse
Affiliation(s)
- Leijing Zhu
- Institute of Bismuth and Rhenium, School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jing Ma
- Department of Pharmacy, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Danli Jin
- Institute of Bismuth and Rhenium, School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yongjian Zhang
- Institute of Bismuth and Rhenium, School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Siyu Wu
- Institute of Bismuth and Rhenium, School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Ajing Xu
- Department of Pharmacy, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yingying Gu
- Institute of Bismuth and Rhenium, School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yarui An
- Institute of Bismuth and Rhenium, School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, China.
| | - Yuqing Miao
- Institute of Bismuth and Rhenium, School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, China
| |
Collapse
|
11
|
Yang J, Phan VM, Heo CK, Nguyen HV, Lim WH, Cho EW, Poo H, Seo TS. Development of nucleocapsid-specific monoclonal antibodies for SARS-CoV-2 and their ELISA diagnostics on an automatic microfluidic device. SENSORS AND ACTUATORS. B, CHEMICAL 2023; 380:133331. [PMID: 36644652 PMCID: PMC9826540 DOI: 10.1016/j.snb.2023.133331] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
Coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has threatened public health globally, and the emergence of viral variants has exacerbated an already precarious situation. To prevent further spread of the virus and determine government action required for virus control, accurate and rapid immunoassays for SARS-CoV-2 diagnosis are urgently needed. In this study, we generated monoclonal antibodies (mAbs) against the SARS-CoV-2 nucleocapsid protein (NP), compared their reactivity using an enzyme-linked immunosorbent assay (ELISA), and selected four mAbs designated 1G6, 3E10, 3F10, and 5B6 which have higher reactivity to NP and viral lysates of SARS-CoV-2 than other mAbs. Using an epitope mapping assay, we identified that 1G6 detected the C-terminal domain of SARS-CoV-2 NP (residues 248-364), while 3E10 and 3F10 bound to the N-terminal domain (residues 47-174) and 3F10 detected the N-arm region (residues 1-46) of SARS-CoV-2 NP. Based on the epitope study and sandwich ELISA, we selected the 1G6 and 3E10 Abs as an optimal Ab pair and applied them for a microfluidics-based point-of-care (POC) ELISA assay to detect the NPs of SARS-CoV-2 and its variants. The integrated and automatic microfluidic system could operate the serial injection of the sample, the washing solution, the HRP-conjugate antibody, and the TMB substrate solution simply by controlling air purge via a single syringe. The proposed Ab pair-equipped microsystem effectively detected the NPs of SARS-CoV-2 variants as well as in clinical samples. Collectively, our proposed platform provides an advanced protein-based diagnostic tool for detecting SARS-CoV-2.
Collapse
Affiliation(s)
- Jihyun Yang
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Vu Minh Phan
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin 17104, South Korea
| | - Chang-Kyu Heo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Hau Van Nguyen
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin 17104, South Korea
| | - Won-Hee Lim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Eun-Wie Cho
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Haryoung Poo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Tae Seok Seo
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin 17104, South Korea
| |
Collapse
|
12
|
Tarim EA, Anil Inevi M, Ozkan I, Kecili S, Bilgi E, Baslar MS, Ozcivici E, Oksel Karakus C, Tekin HC. Microfluidic-based technologies for diagnosis, prevention, and treatment of COVID-19: recent advances and future directions. Biomed Microdevices 2023; 25:10. [PMID: 36913137 PMCID: PMC10009869 DOI: 10.1007/s10544-023-00649-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/14/2023]
Abstract
The COVID-19 pandemic has posed significant challenges to existing healthcare systems around the world. The urgent need for the development of diagnostic and therapeutic strategies for COVID-19 has boomed the demand for new technologies that can improve current healthcare approaches, moving towards more advanced, digitalized, personalized, and patient-oriented systems. Microfluidic-based technologies involve the miniaturization of large-scale devices and laboratory-based procedures, enabling complex chemical and biological operations that are conventionally performed at the macro-scale to be carried out on the microscale or less. The advantages microfluidic systems offer such as rapid, low-cost, accurate, and on-site solutions make these tools extremely useful and effective in the fight against COVID-19. In particular, microfluidic-assisted systems are of great interest in different COVID-19-related domains, varying from direct and indirect detection of COVID-19 infections to drug and vaccine discovery and their targeted delivery. Here, we review recent advances in the use of microfluidic platforms to diagnose, treat or prevent COVID-19. We start by summarizing recent microfluidic-based diagnostic solutions applicable to COVID-19. We then highlight the key roles microfluidics play in developing COVID-19 vaccines and testing how vaccine candidates perform, with a focus on RNA-delivery technologies and nano-carriers. Next, microfluidic-based efforts devoted to assessing the efficacy of potential COVID-19 drugs, either repurposed or new, and their targeted delivery to infected sites are summarized. We conclude by providing future perspectives and research directions that are critical to effectively prevent or respond to future pandemics.
Collapse
Affiliation(s)
- E Alperay Tarim
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Muge Anil Inevi
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Ilayda Ozkan
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Seren Kecili
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Eyup Bilgi
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - M Semih Baslar
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Engin Ozcivici
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | | | - H Cumhur Tekin
- Department of Bioengineering, Izmir Institute of Technology, Izmir, Turkey.
- METU MEMS Center, Ankara, Turkey.
| |
Collapse
|
13
|
Li F, Hong J, Guan C, Chen K, Xie Y, Wu Q, Chen J, Deng B, Shen J, Liu X, Hu R, Zhang Y, Chen Y, Zhu J. Affinity Exploration of SARS-CoV-2 RBD Variants to mAb-Functionalized Plasmonic Metasurfaces for Label-Free Immunoassay Boosting. ACS NANO 2023; 17:3383-3393. [PMID: 36630157 PMCID: PMC9847236 DOI: 10.1021/acsnano.2c08153] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/03/2023] [Indexed: 06/09/2023]
Abstract
Plasmonic metasurfaces (PMs) functionalized with the monoclonal antibody (mAb) are promising biophotonic sensors for biomolecular interaction analysis and convenient immunoassay of various biomarkers, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. Previous PM biosensing suffers from the slow affinity detection rate and lack of sufficient immunoassay studies on various SARS-CoV-2 variants. Here, we develop a high-efficiency affinity testing method based on label-free PM sensors with mAbs and demonstrate their binding characteristics to 12 spike receptor binding domain (RBD) variants of SARS-CoV-2. In addition to the research of plasmonic near-field influence on surface biomolecule sensing, we provide a comprehensive report about the Langmuir binding equilibrium of molecular kinetics between 12 SARS-CoV-2 RBD variants and mAb-functionalized PMs, which plays a crucial role in label-free immunosensing. A high-affinity mAb can be combined with the highly sensitive propagating plasmonic mode to boost the detection of SARS-CoV-2 variants. Owing to a better understanding of molecular dynamics on PMs, we develop an ultrasensitive biosensor of the SARS-CoV-2 Omicron variant. The experiments show great distinguishment of P < 0.0001 from respiratory diseases induced by other viruses, and the limit of detection is 2 orders smaller than the commercial colloidal gold immunoassay. Our study shows the label-free biosensing by low-cost wafer-scale PMs, which will provide essential information on biomolecular interaction and facilitate high-precision point-of-care testing for emerging SARS-CoV-2 variants in the future.
Collapse
Affiliation(s)
- Fajun Li
- Institute of Electromagnetics and Acoustics and Key
Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen
University, Xiamen361005, China
| | - Junping Hong
- State Key Laboratory of Molecular Vaccinology and
Molecular Diagnostics and National Institute of Diagnostics and Vaccine Development in
Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen
University, Xiamen361005, China
| | - Chaoheng Guan
- Institute of Electromagnetics and Acoustics and Key
Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen
University, Xiamen361005, China
| | - Kaiyun Chen
- State Key Laboratory of Molecular Vaccinology and
Molecular Diagnostics and National Institute of Diagnostics and Vaccine Development in
Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen
University, Xiamen361005, China
| | - Yinong Xie
- Institute of Electromagnetics and Acoustics and Key
Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen
University, Xiamen361005, China
| | - Qian Wu
- State Key Laboratory of Molecular Vaccinology and
Molecular Diagnostics and National Institute of Diagnostics and Vaccine Development in
Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen
University, Xiamen361005, China
| | - Junjie Chen
- Analysis and Measurement Center, School of
Pharmaceutical Science, Xiamen University, Xiamen361003,
China
| | - Baichang Deng
- Institute of Electromagnetics and Acoustics and Key
Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen
University, Xiamen361005, China
| | - Jiaqing Shen
- Institute of Electromagnetics and Acoustics and Key
Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen
University, Xiamen361005, China
| | - Xueying Liu
- Institute of Electromagnetics and Acoustics and Key
Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen
University, Xiamen361005, China
| | - Rongsheng Hu
- Institute of Electromagnetics and Acoustics and Key
Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen
University, Xiamen361005, China
| | - Yulong Zhang
- Pen-Tung Sah Institute of Micro-Nano Science and
Technology, Xiamen University, Xiamen361005,
China
| | - Yixin Chen
- State Key Laboratory of Molecular Vaccinology and
Molecular Diagnostics and National Institute of Diagnostics and Vaccine Development in
Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen
University, Xiamen361005, China
| | - Jinfeng Zhu
- Institute of Electromagnetics and Acoustics and Key
Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen
University, Xiamen361005, China
| |
Collapse
|
14
|
Lin Z, Zou Z, Pu Z, Wu M, Zhang Y. Application of microfluidic technologies on COVID-19 diagnosis and drug discovery. Acta Pharm Sin B 2023; 13:S2211-3835(23)00061-8. [PMID: 36855672 PMCID: PMC9951611 DOI: 10.1016/j.apsb.2023.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/02/2023] [Accepted: 02/15/2023] [Indexed: 02/26/2023] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic has boosted the development of antiviral research. Microfluidic technologies offer powerful platforms for diagnosis and drug discovery for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) diagnosis and drug discovery. In this review, we introduce the structure of SARS-CoV-2 and the basic knowledge of microfluidic design. We discuss the application of microfluidic devices in SARS-CoV-2 diagnosis based on detecting viral nucleic acid, antibodies, and antigens. We highlight the contribution of lab-on-a-chip to manufacturing point-of-care equipment of accurate, sensitive, low-cost, and user-friendly virus-detection devices. We then investigate the efforts in organ-on-a-chip and lipid nanoparticles (LNPs) synthesizing chips in antiviral drug screening and mRNA vaccine preparation. Microfluidic technologies contribute to the ongoing SARS-CoV-2 research efforts and provide tools for future viral outbreaks.
Collapse
Affiliation(s)
- Zhun Lin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhengyu Zou
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhe Pu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Minhao Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
15
|
Wu W, Cheng Y, Zhou H, Sun C, Zhang S. The SARS-CoV-2 nucleocapsid protein: its role in the viral life cycle, structure and functions, and use as a potential target in the development of vaccines and diagnostics. Virol J 2023; 20:6. [PMID: 36627683 PMCID: PMC9831023 DOI: 10.1186/s12985-023-01968-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) continues to take a heavy toll on personal health, healthcare systems, and economies around the globe. Scientists are expending tremendous effort to develop diagnostic technologies for detecting positive infections within the shortest possible time, and vaccines and drugs specifically for the prevention and treatment of COVID-19 disease. At the same time, emerging novel variants have raised serious concerns about vaccine efficacy. The SARS-CoV-2 nucleocapsid (N) protein plays an important role in the coronavirus life cycle, and participates in various vital activities after virus invasion. It has attracted a large amount of attention for vaccine and drug development. Here, we summarize the latest research of the N protein, including its role in the SARS-CoV-2 life cycle, structure and function, and post-translational modifications in addition to its involvement in liquid-liquid phase separation (LLPS) and use as a basis for the development of vaccines and diagnostic techniques.
Collapse
Affiliation(s)
- Wenbing Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Ying Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Hong Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Changzhen Sun
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Shujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
16
|
Szczerska M, Wityk P, Listewnik P. The SARS-CoV-2 specific IgG antibodies biophotonic sensor. JOURNAL OF BIOPHOTONICS 2023; 16:e202200172. [PMID: 36222282 PMCID: PMC9874777 DOI: 10.1002/jbio.202200172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
In this paper, we present the design and the principle of operation of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) specific immunoglobulin G (IgG) biophotonic sensor, which is based on the single-mode telecommunication fiber. We fabricated the sensor head at the face of the single mode fiber-28. Due to the process of bio-functionalization, our sensor has the ability to selectively detect the SARS-CoV-2 specific IgG antibodies. The results of preliminary tests allowed us to correctly determine the presence of antibodies in less than 1 min in 5 μl in a volume sample of concentration of 10 μg/ml, which according to studies, corresponds to the concentration of IgG antibodies in human serum. Additionally, the tested sample can be smaller than 5 μl in volume.
Collapse
Affiliation(s)
- Małgorzata Szczerska
- Department of Metrology and Optoelectronics, Faculty of Electronics, Telecommunications and InformaticsGdańsk University of TechnologyGdańskPoland
| | - Paweł Wityk
- Department of Biopharmaceutics and PharmacodynamicsMedical University of GdańskGdańskPoland
| | - Paulina Listewnik
- Department of Metrology and Optoelectronics, Faculty of Electronics, Telecommunications and InformaticsGdańsk University of TechnologyGdańskPoland
| |
Collapse
|
17
|
Li H, Wen X, Ding Y, Wang G, Zhu H, Liu J, Zhao H, Hong X. Photoluminescent and multi-phonon resonance Raman scattering dual-mode immunoassays based on CdS nanoparticles for HIgG detection. Mikrochim Acta 2022; 189:477. [DOI: 10.1007/s00604-022-05530-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/07/2022] [Indexed: 11/28/2022]
|
18
|
Yuan H, Chen P, Wan C, Li Y, Liu BF. Merging microfluidics with luminescence immunoassays for urgent point-of-care diagnostics of COVID-19. Trends Analyt Chem 2022; 157:116814. [PMID: 36373139 PMCID: PMC9637550 DOI: 10.1016/j.trac.2022.116814] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/29/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
The Coronavirus disease 2019 (COVID-19) outbreak has urged the establishment of a global-wide rapid diagnostic system. Current widely-used tests for COVID-19 include nucleic acid assays, immunoassays, and radiological imaging. Immunoassays play an irreplaceable role in rapidly diagnosing COVID-19 and monitoring the patients for the assessment of their severity, risks of the immune storm, and prediction of treatment outcomes. Despite of the enormous needs for immunoassays, the widespread use of traditional immunoassay platforms is still limited by high cost and low automation, which are currently not suitable for point-of-care tests (POCTs). Microfluidic chips with the features of low consumption, high throughput, and integration, provide the potential to enable immunoassays for POCTs, especially in remote areas. Meanwhile, luminescence detection can be merged with immunoassays on microfluidic platforms for their good performance in quantification, sensitivity, and specificity. This review introduces both homogenous and heterogenous luminescence immunoassays with various microfluidic platforms. We also summarize the strengths and weaknesses of the categorized methods, highlighting their recent typical progress. Additionally, different microfluidic platforms are described for comparison. The latest advances in combining luminescence immunoassays with microfluidic platforms for POCTs of COVID-19 are further explained with antigens, antibodies, and related cytokines. Finally, challenges and future perspectives were discussed.
Collapse
Affiliation(s)
- Huijuan Yuan
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chao Wan
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
19
|
Everitt ML, Boegner DJ, White IM. Sample-to-Answer Immuno-Magnetic Assay Using Thermally Responsive Alkane Partitions. BIOSENSORS 2022; 12:1030. [PMID: 36421148 PMCID: PMC9688217 DOI: 10.3390/bios12111030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 06/16/2023]
Abstract
To combat pandemics, there is a need for rapid point-of-care diagnostics to identify infected patients and to track the spread of the disease. While recent progress has been made in response to COVID-19, there continues to be a need for point-of-care diagnostics capable of detecting biomarkers-such as antibodies-in whole blood. We have recently reported the development of thermally responsive alkane partitions (TRAPs) for the automation of point-of-care immuno-magnetic assays. Here, we demonstrate the use of TRAPs to enable sample-to-answer detection of antibodies against the SARS-CoV-2 virus in whole blood samples. We report a limit of detection of 84 pg/mL, well below the clinically relevant threshold. We anticipate that the TRAP-enabled sample-to-answer immunoassay can be used to track the progression of future pandemics, leading to a more informed and robust clinical and societal response.
Collapse
|
20
|
Fox T, Geppert J, Dinnes J, Scandrett K, Bigio J, Sulis G, Hettiarachchi D, Mathangasinghe Y, Weeratunga P, Wickramasinghe D, Bergman H, Buckley BS, Probyn K, Sguassero Y, Davenport C, Cunningham J, Dittrich S, Emperador D, Hooft L, Leeflang MM, McInnes MD, Spijker R, Struyf T, Van den Bruel A, Verbakel JY, Takwoingi Y, Taylor-Phillips S, Deeks JJ. Antibody tests for identification of current and past infection with SARS-CoV-2. Cochrane Database Syst Rev 2022; 11:CD013652. [PMID: 36394900 PMCID: PMC9671206 DOI: 10.1002/14651858.cd013652.pub2] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND The diagnostic challenges associated with the COVID-19 pandemic resulted in rapid development of diagnostic test methods for detecting SARS-CoV-2 infection. Serology tests to detect the presence of antibodies to SARS-CoV-2 enable detection of past infection and may detect cases of SARS-CoV-2 infection that were missed by earlier diagnostic tests. Understanding the diagnostic accuracy of serology tests for SARS-CoV-2 infection may enable development of effective diagnostic and management pathways, inform public health management decisions and understanding of SARS-CoV-2 epidemiology. OBJECTIVES To assess the accuracy of antibody tests, firstly, to determine if a person presenting in the community, or in primary or secondary care has current SARS-CoV-2 infection according to time after onset of infection and, secondly, to determine if a person has previously been infected with SARS-CoV-2. Sources of heterogeneity investigated included: timing of test, test method, SARS-CoV-2 antigen used, test brand, and reference standard for non-SARS-CoV-2 cases. SEARCH METHODS The COVID-19 Open Access Project living evidence database from the University of Bern (which includes daily updates from PubMed and Embase and preprints from medRxiv and bioRxiv) was searched on 30 September 2020. We included additional publications from the Evidence for Policy and Practice Information and Co-ordinating Centre (EPPI-Centre) 'COVID-19: Living map of the evidence' and the Norwegian Institute of Public Health 'NIPH systematic and living map on COVID-19 evidence'. We did not apply language restrictions. SELECTION CRITERIA We included test accuracy studies of any design that evaluated commercially produced serology tests, targeting IgG, IgM, IgA alone, or in combination. Studies must have provided data for sensitivity, that could be allocated to a predefined time period after onset of symptoms, or after a positive RT-PCR test. Small studies with fewer than 25 SARS-CoV-2 infection cases were excluded. We included any reference standard to define the presence or absence of SARS-CoV-2 (including reverse transcription polymerase chain reaction tests (RT-PCR), clinical diagnostic criteria, and pre-pandemic samples). DATA COLLECTION AND ANALYSIS We use standard screening procedures with three reviewers. Quality assessment (using the QUADAS-2 tool) and numeric study results were extracted independently by two people. Other study characteristics were extracted by one reviewer and checked by a second. We present sensitivity and specificity with 95% confidence intervals (CIs) for each test and, for meta-analysis, we fitted univariate random-effects logistic regression models for sensitivity by eligible time period and for specificity by reference standard group. Heterogeneity was investigated by including indicator variables in the random-effects logistic regression models. We tabulated results by test manufacturer and summarised results for tests that were evaluated in 200 or more samples and that met a modification of UK Medicines and Healthcare products Regulatory Agency (MHRA) target performance criteria. MAIN RESULTS We included 178 separate studies (described in 177 study reports, with 45 as pre-prints) providing 527 test evaluations. The studies included 64,688 samples including 25,724 from people with confirmed SARS-CoV-2; most compared the accuracy of two or more assays (102/178, 57%). Participants with confirmed SARS-CoV-2 infection were most commonly hospital inpatients (78/178, 44%), and pre-pandemic samples were used by 45% (81/178) to estimate specificity. Over two-thirds of studies recruited participants based on known SARS-CoV-2 infection status (123/178, 69%). All studies were conducted prior to the introduction of SARS-CoV-2 vaccines and present data for naturally acquired antibody responses. Seventy-nine percent (141/178) of studies reported sensitivity by week after symptom onset and 66% (117/178) for convalescent phase infection. Studies evaluated enzyme-linked immunosorbent assays (ELISA) (165/527; 31%), chemiluminescent assays (CLIA) (167/527; 32%) or lateral flow assays (LFA) (188/527; 36%). Risk of bias was high because of participant selection (172, 97%); application and interpretation of the index test (35, 20%); weaknesses in the reference standard (38, 21%); and issues related to participant flow and timing (148, 82%). We judged that there were high concerns about the applicability of the evidence related to participants in 170 (96%) studies, and about the applicability of the reference standard in 162 (91%) studies. Average sensitivities for current SARS-CoV-2 infection increased by week after onset for all target antibodies. Average sensitivity for the combination of either IgG or IgM was 41.1% in week one (95% CI 38.1 to 44.2; 103 evaluations; 3881 samples, 1593 cases), 74.9% in week two (95% CI 72.4 to 77.3; 96 evaluations, 3948 samples, 2904 cases) and 88.0% by week three after onset of symptoms (95% CI 86.3 to 89.5; 103 evaluations, 2929 samples, 2571 cases). Average sensitivity during the convalescent phase of infection (up to a maximum of 100 days since onset of symptoms, where reported) was 89.8% for IgG (95% CI 88.5 to 90.9; 253 evaluations, 16,846 samples, 14,183 cases), 92.9% for IgG or IgM combined (95% CI 91.0 to 94.4; 108 evaluations, 3571 samples, 3206 cases) and 94.3% for total antibodies (95% CI 92.8 to 95.5; 58 evaluations, 7063 samples, 6652 cases). Average sensitivities for IgM alone followed a similar pattern but were of a lower test accuracy in every time slot. Average specificities were consistently high and precise, particularly for pre-pandemic samples which provide the least biased estimates of specificity (ranging from 98.6% for IgM to 99.8% for total antibodies). Subgroup analyses suggested small differences in sensitivity and specificity by test technology however heterogeneity in study results, timing of sample collection, and smaller sample numbers in some groups made comparisons difficult. For IgG, CLIAs were the most sensitive (convalescent-phase infection) and specific (pre-pandemic samples) compared to both ELISAs and LFAs (P < 0.001 for differences across test methods). The antigen(s) used (whether from the Spike-protein or nucleocapsid) appeared to have some effect on average sensitivity in the first weeks after onset but there was no clear evidence of an effect during convalescent-phase infection. Investigations of test performance by brand showed considerable variation in sensitivity between tests, and in results between studies evaluating the same test. For tests that were evaluated in 200 or more samples, the lower bound of the 95% CI for sensitivity was 90% or more for only a small number of tests (IgG, n = 5; IgG or IgM, n = 1; total antibodies, n = 4). More test brands met the MHRA minimum criteria for specificity of 98% or above (IgG, n = 16; IgG or IgM, n = 5; total antibodies, n = 7). Seven assays met the specified criteria for both sensitivity and specificity. In a low-prevalence (2%) setting, where antibody testing is used to diagnose COVID-19 in people with symptoms but who have had a negative PCR test, we would anticipate that 1 (1 to 2) case would be missed and 8 (5 to 15) would be falsely positive in 1000 people undergoing IgG or IgM testing in week three after onset of SARS-CoV-2 infection. In a seroprevalence survey, where prevalence of prior infection is 50%, we would anticipate that 51 (46 to 58) cases would be missed and 6 (5 to 7) would be falsely positive in 1000 people having IgG tests during the convalescent phase (21 to 100 days post-symptom onset or post-positive PCR) of SARS-CoV-2 infection. AUTHORS' CONCLUSIONS Some antibody tests could be a useful diagnostic tool for those in whom molecular- or antigen-based tests have failed to detect the SARS-CoV-2 virus, including in those with ongoing symptoms of acute infection (from week three onwards) or those presenting with post-acute sequelae of COVID-19. However, antibody tests have an increasing likelihood of detecting an immune response to infection as time since onset of infection progresses and have demonstrated adequate performance for detection of prior infection for sero-epidemiological purposes. The applicability of results for detection of vaccination-induced antibodies is uncertain.
Collapse
Affiliation(s)
- Tilly Fox
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Julia Geppert
- Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Jacqueline Dinnes
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Katie Scandrett
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Jacob Bigio
- Research Institute of the McGill University Health Centre, Montreal, Canada
- McGill International TB Centre, Montreal, Canada
| | - Giorgia Sulis
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
| | - Dineshani Hettiarachchi
- Department of Anatomy Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Yasith Mathangasinghe
- Department of Anatomy Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Praveen Weeratunga
- Department of Clinical Medicine, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | | | | | - Brian S Buckley
- Cochrane Response, Cochrane, London, UK
- Department of Surgery, University of the Philippines, Manila, Philippines
| | | | | | - Clare Davenport
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Jane Cunningham
- Global Malaria Programme, World Health Organization, Geneva, Switzerland
| | | | | | - Lotty Hooft
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht , Netherlands
| | - Mariska Mg Leeflang
- Epidemiology and Data Science, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Public Health, Amsterdam, Netherlands
| | | | - René Spijker
- Medical Library, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health, Amsterdam, Netherlands
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Thomas Struyf
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Ann Van den Bruel
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Jan Y Verbakel
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Yemisi Takwoingi
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Sian Taylor-Phillips
- Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, UK
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Jonathan J Deeks
- Test Evaluation Research Group, Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| |
Collapse
|
21
|
Alyafei K, Ahmed R, Abir FF, Chowdhury MEH, Naji KK. A comprehensive review of COVID-19 detection techniques: From laboratory systems to wearable devices. Comput Biol Med 2022; 149:106070. [PMID: 36099862 PMCID: PMC9433350 DOI: 10.1016/j.compbiomed.2022.106070] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 08/03/2022] [Accepted: 08/27/2022] [Indexed: 11/30/2022]
Abstract
Screening of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) among symptomatic and asymptomatic patients offers unique opportunities for curtailing the transmission of novel coronavirus disease 2019, commonly known as COVID-19. Molecular diagnostic techniques, namely reverse transcription loop-mediated isothermal amplification (RT-LAMP), reverse transcription-polymerase chain reaction (RT-PCR), and immunoassays, have been frequently used to identify COVID-19 infection. Although these techniques are robust and accurate, mass testing of potentially infected individuals has shown difficulty due to the resources, manpower, and costs it entails. Moreover, as these techniques are typically used to test symptomatic patients, healthcare systems have failed to screen asymptomatic patients, whereas the spread of COVID-19 by these asymptomatic individuals has turned into a crucial problem. Besides, respiratory infections or cardiovascular conditions generally demonstrate changes in physiological parameters, namely body temperature, blood pressure, and breathing rate, which signifies the onset of diseases. Such vitals monitoring systems have shown promising results employing artificial intelligence (AI). Therefore, the potential use of wearable devices for monitoring asymptomatic COVID-19 individuals has recently been explored. This work summarizes the efforts that have been made in the domains from laboratory-based testing to asymptomatic patient monitoring via wearable systems.
Collapse
Affiliation(s)
- Khalid Alyafei
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, 2713, Qatar
| | - Rashid Ahmed
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, 2713, Qatar; Department of Biotechnology, Mirpur University of Science and Technology (MUST), Mirpur, 10250, AJK, Pakistan
| | - Farhan Fuad Abir
- Department of Electrical Engineering, Qatar University, Doha, 2713, Qatar
| | | | | |
Collapse
|
22
|
Wu F, Mao M, Cai L, Lin Q, Guan X, Shi X, Ma L. Platinum-Decorated Gold Nanoparticle-Based Microfluidic Chip Immunoassay for Ultrasensitive Colorimetric Detection of SARS-CoV-2 Nucleocapsid Protein. ACS Biomater Sci Eng 2022; 8:3924-3932. [PMID: 35929757 PMCID: PMC9380870 DOI: 10.1021/acsbiomaterials.2c00600] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022]
Abstract
Gold nanoparticle-based point-of-care tests (POCT) are one of the most widely used diagnostic tools for SARS-CoV-2 screening. However, the limitation of their insufficient sensitivity often leads to false negative results in early disease diagnostics. The ongoing pandemic of COVID-19 makes diagnostic tools that are more accurate, sensitive, simple, and affordable in high demand. In this work, we develop a platinum-decorated gold nanoparticle (Au@Pt NP)-based microfluidic chip immunoassay with a sensitivity surpassing that of paper-based detection of nucleocapsid (N) protein, one of the most conserved biomarkers of COVID-19. The synthesized Au@Pt NPs show high stability and catalytic activity in complex environments. The catalytic amplification of Au@Pt NPs enables naked-eye detection of N protein in the low femtogram range (ca. 0.1 pg/mL) and the detection of throat swab samples in under 40 min. This microfluidic chip immunoassay is easy for operation and readout without instrument assistance, making it more suitable for on-site detection and future pathogen surveillance.
Collapse
Affiliation(s)
- Feng Wu
- School of Life Sciences, Tsinghua
University, Beijing 100084, China
- Institute of Biopharmaceutical and Health Engineering,
Tsinghua Shenzhen International Graduate School, Tsinghua
University, Shenzhen 518055, China
| | - Mao Mao
- School of Life Sciences, Tsinghua
University, Beijing 100084, China
- Institute of Biopharmaceutical and Health Engineering,
Tsinghua Shenzhen International Graduate School, Tsinghua
University, Shenzhen 518055, China
| | - Liangyu Cai
- School of Life Sciences, Tsinghua
University, Beijing 100084, China
- Institute of Biopharmaceutical and Health Engineering,
Tsinghua Shenzhen International Graduate School, Tsinghua
University, Shenzhen 518055, China
| | - Qianyu Lin
- Tsinghua-Berkeley Shenzhen Institute,
Tsinghua University, Shenzhen 518055,
China
| | - Xuejiao Guan
- School of Life Sciences, Tsinghua
University, Beijing 100084, China
- Institute of Biopharmaceutical and Health Engineering,
Tsinghua Shenzhen International Graduate School, Tsinghua
University, Shenzhen 518055, China
| | - Xueying Shi
- Institute of Biopharmaceutical and Health Engineering,
Tsinghua Shenzhen International Graduate School, Tsinghua
University, Shenzhen 518055, China
| | - Lan Ma
- Institute of Biopharmaceutical and Health Engineering,
Tsinghua Shenzhen International Graduate School, Tsinghua
University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute,
Tsinghua University, Shenzhen 518055,
China
- State Key Laboratory of Chemical Oncogenomics,
Tsinghua Shenzhen International Graduate School, Tsinghua
University, Shenzhen 518055, China
- Institute of Biomedical Health Technology
and Engineering, Shenzhen Bay Laboratory, Shenzhen 518055,
China
| |
Collapse
|
23
|
Bao L, Park J, Qin B, Kim B. Anti-SARS-CoV-2 IgM/IgG antibodies detection using a patch sensor containing porous microneedles and a paper-based immunoassay. Sci Rep 2022; 12:10693. [PMID: 35778408 PMCID: PMC9249772 DOI: 10.1038/s41598-022-14725-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/10/2022] [Indexed: 12/21/2022] Open
Abstract
Infectious diseases are among the leading causes of mortality worldwide. A new coronavirus named severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) was identified in Wuhan, China in 2019, and the World Health Organization (WHO) declared its outbreak, coronavirus disease 2019 (COVID-19), as a global pandemic in 2020. COVID-19 can spread quickly from person to person. One of the most challenging issues is to identify the infected individuals and prevent potential spread of SARS-CoV-2. Recently, anti-SARS-CoV-2 immunoglobulin M (IgM) and immunoglobulin G (IgG) antibody tests using immunochromatographic methods have been used as a complement to current detection methods and have provided information of the approximate course of COVID-19 infection. However, blood sampling causes pain and poses risks of infection at the needle puncture site. In this study, a novel patch sensor integrating porous microneedles and an immunochromatographic assay (PMNIA) was developed for the rapid detection of anti-SARS-CoV-2 IgM/IgG in dermal interstitial fluid (ISF), which is a rich source of protein biomarkers, such as antibodies. Biodegradable porous microneedles (MNs) made of polylactic acid were fabricated to extract ISF from human skin by capillary effect. The extracted ISF was vertically transported and flowed into the affixed immunoassay biosensor, where specific antibodies could be detected colorimetrically on-site. Anti-SARS-CoV-2 IgM/IgG antibodies were simultaneously detected within 3 min in vitro. Moreover, the limit of detection of anti-SARS-CoV-2 IgM and IgG concentrations was as low as 3 and 7 ng/mL, respectively. The developed device integrating porous MNs and immunochromatographic biosensors is expected to enable minimally invasive, simple, and rapid anti-SARS-CoV-2 IgM/IgG antibody testing. Furthermore, the compact size of the MN and biosensor-integrated device is advantageous for its widespread use. The proposed device has great potential for rapid screening of various infectious diseases in addition to COVID-19 as an effective complementary method with other diagnostic tests.
Collapse
Affiliation(s)
- Leilei Bao
- Institute of Industrial Science, The Univeristy of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Jongho Park
- Institute of Industrial Science, The Univeristy of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Boyu Qin
- Institute of Industrial Science, The Univeristy of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Beomjoon Kim
- Institute of Industrial Science, The Univeristy of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan.
| |
Collapse
|
24
|
Zeng J, Duarte PA, Ma Y, Savchenko O, Shoute L, Khaniani Y, Babiuk S, Zhuo R, Abdelrasoul GN, Charlton C, Kanji JN, Babiuk L, Edward C, Chen J. An impedimetric biosensor for COVID-19 serology test and modification of sensor performance via dielectrophoresis force. Biosens Bioelectron 2022; 213:114476. [PMID: 35716642 PMCID: PMC9186431 DOI: 10.1016/j.bios.2022.114476] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 11/29/2022]
Abstract
Coronavirus disease 2019 (COVID-19) has caused significant global morbidity and mortality. The serology test that detects antibodies against the disease causative agent, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has often neglected value in supporting immunization policies and therapeutic decision-making. The ELISA-based antibody test is time-consuming and bulky. This work described a gold micro-interdigitated electrodes (IDE) biosensor for COVID antibody detection based on Electrochemical Impedance Spectroscopy (EIS) responses. The IDE architecture allows easy surface modification with the viral structure protein, Spike (S) protein, in the gap of the electrode digits to selectively capture anti-S antibodies in buffer solutions or human sera. Two strategies were employed to resolve the low sensitivity issue of non-faradic impedimetric sensors and the sensor fouling phenomenon when using the serum. One uses secondary antibody-gold nanoparticle (AuNP) conjugates to further distinguish anti-S antibodies from the non-specific binding and obtain a more significant impedance change. The second strategy consists of increasing the concentration of target antibodies in the gap of IDEs by inducing an AC electrokinetic effect such as dielectrophoresis (DEP). AuNP and DEP methods reached a limit of detection of 200 ng/mL and 2 μg/mL, respectively using purified antibodies in buffer, while the DEP method achieved a faster testing time of only 30 min. Both strategies could qualitatively distinguish COVID-19 antibody-positive and -negative sera. Our work, especially the impedimetric detection of COVID-19 antibodies under the assistance of the DEP force presents a promising path toward rapid, point-of-care solutions for COVID-19 serology tests.
Collapse
Affiliation(s)
- Jie Zeng
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, Canada
| | - Pedro A Duarte
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, Canada
| | - Yuhao Ma
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, Canada
| | - Oleksandra Savchenko
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, Canada
| | - Lian Shoute
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, Canada
| | - Yeganeh Khaniani
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - Shawn Babiuk
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, Canada
| | - Ran Zhuo
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, T6G 2B7, Canada
| | - Gaser N Abdelrasoul
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, Canada
| | - Carmen Charlton
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, T6G 2B7, Canada; Public Health Laboratory, Alberta Precision Laboratories, Calgary, AB, Canada; Li Ka Shing Institute for Virology, University of Alberta, Edmonton, AB, Canada
| | - Jamil N Kanji
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, T6G 2B7, Canada; Public Health Laboratory, Alberta Precision Laboratories, Calgary, AB, Canada; Division of Infectious Diseases, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Pathology & Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lorne Babiuk
- Vaccine and Infectious Disease Organization, University of Alberta, Edmonton, AB, Canada
| | - Cole Edward
- Public Health Laboratory, Alberta Precision Laboratories, Calgary, AB, Canada
| | - Jie Chen
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, Canada; Department of Biomedical Engineering, University of Alberta, Edmonton, Canada.
| |
Collapse
|
25
|
Jamiruddin MR, Meghla BA, Islam DZ, Tisha TA, Khandker SS, Khondoker MU, Haq MA, Adnan N, Haque M. Microfluidics Technology in SARS-CoV-2 Diagnosis and Beyond: A Systematic Review. Life (Basel) 2022; 12:649. [PMID: 35629317 PMCID: PMC9146058 DOI: 10.3390/life12050649] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/22/2022] Open
Abstract
With the progression of the COVID-19 pandemic, new technologies are being implemented for more rapid, scalable, and sensitive diagnostics. The implementation of microfluidic techniques and their amalgamation with different detection techniques has led to innovative diagnostics kits to detect SARS-CoV-2 antibodies, antigens, and nucleic acids. In this review, we explore the different microfluidic-based diagnostics kits and how their amalgamation with the various detection techniques has spearheaded their availability throughout the world. Three other online databases, PubMed, ScienceDirect, and Google Scholar, were referred for articles. One thousand one hundred sixty-four articles were determined with the search algorithm of microfluidics followed by diagnostics and SARS-CoV-2. We found that most of the materials used to produce microfluidics devices were the polymer materials such as PDMS, PMMA, and others. Centrifugal force is the most commonly used fluid manipulation technique, followed by electrochemical pumping, capillary action, and isotachophoresis. The implementation of the detection technique varied. In the case of antibody detection, spectrometer-based detection was most common, followed by fluorescence-based as well as colorimetry-based. In contrast, antigen detection implemented electrochemical-based detection followed by fluorescence-based detection, and spectrometer-based detection were most common. Finally, nucleic acid detection exclusively implements fluorescence-based detection with a few colorimetry-based detections. It has been further observed that the sensitivity and specificity of most devices varied with implementing the detection-based technique alongside the fluid manipulation technique. Most microfluidics devices are simple and incorporate the detection-based system within the device. This simplifies the deployment of such devices in a wide range of environments. They can play a significant role in increasing the rate of infection detection and facilitating better health services.
Collapse
Affiliation(s)
| | - Bushra Ayat Meghla
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (B.A.M.); (D.Z.I.); (T.A.T.)
| | - Dewan Zubaer Islam
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (B.A.M.); (D.Z.I.); (T.A.T.)
| | - Taslima Akter Tisha
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (B.A.M.); (D.Z.I.); (T.A.T.)
| | - Shahad Saif Khandker
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.A.H.)
| | - Mohib Ullah Khondoker
- Department of Community Medicine, Gonoshasthaya Samaj Vittik Medical College, Savar, Dhaka 1344, Bangladesh;
| | - Md. Ahsanul Haq
- Gonoshasthaya-RNA Molecular Diagnostic & Research Center, Dhanmondi, Dhaka 1205, Bangladesh; (S.S.K.); (M.A.H.)
| | - Nihad Adnan
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (B.A.M.); (D.Z.I.); (T.A.T.)
| | - Mainul Haque
- The Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sugai Besi, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
26
|
Jiang M, Dong T, Han C, Liu L, Zhang T, Kang Q, Wang P, Zhou F. Regenerable and high-throughput surface plasmon resonance assay for rapid screening of anti-SARS-CoV-2 antibody in serum samples. Anal Chim Acta 2022; 1208:339830. [PMID: 35525598 PMCID: PMC9006689 DOI: 10.1016/j.aca.2022.339830] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/30/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022]
|
27
|
Luo J, Brakel A, Krizsan A, Ludwig T, Mötzing M, Volke D, Lakowa N, Grünewald T, Lehmann C, Wolf J, Borte S, Milkovska-Stamenova S, Gabert J, Fingas F, Scholz M, Hoffmann R. Sensitive and specific serological ELISA for the detection of SARS-CoV-2 infections. Virol J 2022; 19:50. [PMID: 35305688 PMCID: PMC8934124 DOI: 10.1186/s12985-022-01768-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/24/2022] [Indexed: 12/28/2022] Open
Abstract
Background The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has triggered the worldwide coronavirus disease 2019 (COVID-19) pandemic. Serological assays for the detection of SARS-CoV-2 infections are important to understand the immune response in patients and to obtain epidemiological data about the number of infected people, especially to identify asymptomatic persons not aware of a past infection. Methods We recombinantly produced SARS-CoV-2 nucleocapsid (N)-protein in Escherichia coli. We used the purified protein to develop an indirect enzyme-linked immunosorbent assay (ELISA) for the detection of SARS-CoV-2 specific antibodies. This ELISA method was optimized and validated with serum samples collected from 113 patients with RT-PCR-confirmed SARS-CoV-2 infections including hospitalized COVID-19 patients and 1500 control sera mostly collected before 2015 with different clinical background. Results The optimized N-protein-ELISA provided a sensitivity of 89.7% (n = 68) for samples collected from patients with confirmed SARS-CoV-2 infections and mild to severe symptoms more than 14 days after symptom onset or a positive PCR test. The antibody levels remained low for serum samples collected in the first six days (n = 23) and increased in the second week (n = 22) post symptom onset or PCR confirmation. At this early phase, the ELISA provided a sensitivity of 39.1% and 86.4%, respectively, reflecting the time of an IgG immune response against pathogens. The assay specificity was 99.3% (n = 1500; 95% CI 0.995–0.999). Serum samples from persons with confirmed antibody titers against human immunodeficiency viruses 1/2, parvovirus B19, hepatitis A/B virus, cytomegalovirus, Epstein Barr virus, and herpes simplex virus were tested negative. Conclusions We conclude that the N-protein-based ELISA developed here is well suited for the sensitive and specific serological detection of SARS-CoV-2 specific IgG antibodies in human serum for symptomatic infections. It may also prove useful to identify previous SARS-CoV-2 infections in vaccinated people, as all currently approved vaccines rely on the SARS-CoV-2 spike (S-) protein. Supplementary Information The online version contains supplementary material available at 10.1186/s12985-022-01768-4.
Collapse
|
28
|
Flores-Contreras EA, González-González RB, Rodríguez-Sánchez IP, Yee-de León JF, Iqbal HMN, González-González E. Microfluidics-Based Biosensing Platforms: Emerging Frontiers in Point-of-Care Testing SARS-CoV-2 and Seroprevalence. BIOSENSORS 2022; 12:179. [PMID: 35323449 PMCID: PMC8946853 DOI: 10.3390/bios12030179] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the ongoing COVID-19 (coronavirus disease-2019) outbreak and has unprecedentedly impacted the public health and economic sector. The pandemic has forced researchers to focus on the accurate and early detection of SARS-CoV-2, developing novel diagnostic tests. Among these, microfluidic-based tests stand out for their multiple benefits, such as their portability, low cost, and minimal reagents used. This review discusses the different microfluidic platforms applied in detecting SARS-CoV-2 and seroprevalence, classified into three sections according to the molecules to be detected, i.e., (1) nucleic acid, (2) antigens, and (3) anti-SARS-CoV-2 antibodies. Moreover, commercially available alternatives based on microfluidic platforms are described. Timely and accurate results allow healthcare professionals to perform efficient treatments and make appropriate decisions for infection control; therefore, novel developments that integrate microfluidic technology may provide solutions in the form of massive diagnostics to control the spread of infectious diseases.
Collapse
Affiliation(s)
- Elda A. Flores-Contreras
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Nuevo León, Mexico; (E.A.F.-C.); (R.B.G.-G.)
| | | | - Iram P. Rodríguez-Sánchez
- Laboratorio de Fisiología Molecular y Estructural, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, Nuevo León, Mexico;
| | | | - Hafiz M. N. Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Nuevo León, Mexico; (E.A.F.-C.); (R.B.G.-G.)
| | - Everardo González-González
- Laboratorio de Fisiología Molecular y Estructural, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, Nuevo León, Mexico;
| |
Collapse
|
29
|
Wu W, Tan X, Zupancic J, Schardt JS, Desai AA, Smith MD, Zhang J, Xie L, Oo MK, Tessier PM, Fan X. Rapid and Quantitative In Vitro Evaluation of SARS-CoV-2 Neutralizing Antibodies and Nanobodies. Anal Chem 2022; 94:4504-4512. [PMID: 35238533 PMCID: PMC9356539 DOI: 10.1021/acs.analchem.2c00062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Neutralizing monoclonal antibodies and nanobodies have shown promising results as potential therapeutic agents for COVID-19. Identifying such antibodies and nanobodies requires evaluating the neutralization activity of a large number of lead molecules via biological assays, such as the virus neutralization test (VNT). These assays are typically time-consuming and demanding on-lab facilities. Here, we present a rapid and quantitative assay that evaluates the neutralizing efficacy of an antibody or nanobody within 1.5 h, does not require BSL-2 facilities, and consumes only 8 μL of a low concentration (ng/mL) sample for each assay run. We tested the human angiotensin-converting enzyme 2 (ACE2) binding inhibition efficacy of seven antibodies and eight nanobodies and verified that the IC50 values of our assay are comparable with those from SARS-CoV-2 pseudovirus neutralization tests. We also found that our assay could evaluate the neutralizing efficacy against three widespread SARS-CoV-2 variants. We observed increased affinity of these variants for ACE2, including the β and γ variants. Finally, we demonstrated that our assay enables the rapid identification of an immune-evasive mutation of the SARS-CoV-2 spike protein, utilizing a set of nanobodies with known binding epitopes.
Collapse
Affiliation(s)
- Weishu Wu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Xiaotian Tan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jennifer Zupancic
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - John S Schardt
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Alec A Desai
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Matthew D Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jie Zhang
- Beijing Key Laboratory of Monoclonal Antibody Research and Development, Sino Biological Inc., Beijing 100176, China
| | - Liangzhi Xie
- Beijing Key Laboratory of Monoclonal Antibody Research and Development, Sino Biological Inc., Beijing 100176, China.,Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China
| | - Maung Khaing Oo
- Optofluidic Bioassay, LLC, Ann Arbor, Michigan 48103, United States
| | - Peter M Tessier
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States.,Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Xudong Fan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
30
|
Peng P, Liu C, Li Z, Xue Z, Mao P, Hu J, Xu F, Yao C, You M. Emerging ELISA Derived Technologies for in vitro Diagnostics. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Yang ZW, Pham TTH, Hsu CC, Lien CH, Phan QH. Single-Layer-Graphene-Coated and Gold-Film-Based Surface Plasmon Resonance Prism Coupler Sensor for Immunoglobulin G Detection. SENSORS 2022; 22:s22041362. [PMID: 35214258 PMCID: PMC8962983 DOI: 10.3390/s22041362] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 02/07/2023]
Abstract
A graphene-based surface plasmon resonance (SPR) prism coupler sensor is proposed for the rapid detection of immunoglobulin G (IgG) antibodies. The feasibility of the proposed sensor is demonstrated by measuring the IgG concentration in phantom mouse and human serum solutions over the range of 0–250 ng/mL. The results show that the circular dichroism and principal fast axis angle of linear birefringence increase in line with increases in IgG concentration over the considered range. Moreover, the proposed device has a resolution of 5–10 ng/mL and a response time of less than three minutes. In general, the sensor provides a promising approach for IgG detection and has significant potential for rapid infectious viral disease testing applications.
Collapse
Affiliation(s)
- Zhe-Wei Yang
- Department of Mechanical Engineering, National United University, Miaoli 36063, Taiwan; (Z.-W.Y.); (C.-C.H.)
| | - Thi-Thu-Hien Pham
- Department of Biomedical Engineering, International University-Vietnam National University, Ho Chi Minh City 700000, Vietnam;
| | - Chin-Chi Hsu
- Department of Mechanical Engineering, National United University, Miaoli 36063, Taiwan; (Z.-W.Y.); (C.-C.H.)
| | - Chi-Hsiang Lien
- Department of Mechanical Engineering, National United University, Miaoli 36063, Taiwan; (Z.-W.Y.); (C.-C.H.)
- Correspondence: (C.-H.L.); (Q.-H.P.)
| | - Quoc-Hung Phan
- Department of Mechanical Engineering, National United University, Miaoli 36063, Taiwan; (Z.-W.Y.); (C.-C.H.)
- Correspondence: (C.-H.L.); (Q.-H.P.)
| |
Collapse
|
32
|
Bian S, Shang M, Sawan M. Rapid biosensing SARS-CoV-2 antibodies in vaccinated healthy donors. Biosens Bioelectron 2022; 204:114054. [PMID: 35151002 PMCID: PMC8810518 DOI: 10.1016/j.bios.2022.114054] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/23/2021] [Accepted: 01/27/2022] [Indexed: 01/06/2023]
Abstract
In this study, we report two fiber optic-biolayer interferometry (FO-BLI)-based biosensors for the rapid detection of SARS-CoV-2 neutralizing antibodies (NAbs) and binding antibodies (BAbs) in human serum. The use of signal enhancer 3,3′-diaminobenzidine enabled the detection of NAbs, anti-receptor binding domain (anti-RBD) BAbs, and anti-extracellular domain of spike protein (anti-S-ECD) BAbs up to as low as 10 ng/mL in both buffer and 100-fold diluted serum. NAbs and BAbs could be detected individually over 7.5 and 13 min, respectively, or simultaneously by prolonging the detection time of the former. The protocol for the detection of BAbs could be utilized for detection of the RBD-N501Y variant with equal sensitivity and speed. Results of the NAbs and the anti-RBD BAbs biosensors correlated well with those of the corresponding commercial assay kit. Clinical utility of the two FO-BLI biosensors were further validated using a small cohort of samples randomly taken from 16 enrolled healthy participants who received inactivated vaccines. Two potent serum antibodies were identified, which showed high neutralizing capacities toward RBD and pseudovirus. Overall, the rapid automated biosensors can be used for an individual sample measurement of NAbs and BAbs as well as for high-throughput analysis. The findings of this study would be useful in COVID-19 related studies in vaccine trials, research on dynamics of the immune response, and epidemiology studies.
Collapse
Affiliation(s)
- Sumin Bian
- CenBRAIN, School of Engineering, Westlake University, China
| | - Min Shang
- Dept. of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, China
| | - Mohamad Sawan
- CenBRAIN, School of Engineering, Westlake University, China.
| |
Collapse
|
33
|
Peng R, Pan Y, Li Z, Qin Z, Rini JM, Liu X. SPEEDS: A portable serological testing platform for rapid electrochemical detection of SARS-CoV-2 antibodies. Biosens Bioelectron 2022; 197:113762. [PMID: 34773750 PMCID: PMC8558107 DOI: 10.1016/j.bios.2021.113762] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/27/2021] [Accepted: 10/30/2021] [Indexed: 12/23/2022]
Abstract
The COVID-19 pandemic has resulted in a worldwide health crisis. Rapid diagnosis, new therapeutics and effective vaccines will all be required to stop the spread of COVID-19. Quantitative evaluation of serum antibody levels against the SARS-CoV-2 virus provides a means of monitoring a patient's immune response to a natural viral infection or vaccination, as well as evidence of a prior infection. In this paper, a portable and low-cost electrochemical immunosensor is developed for the rapid and accurate quantification of SARS-CoV-2 serum antibodies. The immunosensor is capable of quantifying the concentrations of immunoglobulin G (IgG) and immunoglobulin M (IgM) antibodies against the SARS-CoV-2 spike protein in human serum. For IgG and IgM, it provides measurements in the range of 10.1 ng/mL - 60 μg/mL and 1.64 ng/mL - 50 μg/mL, respectively, both with an assay time of 13 min. We also developed device stabilization and storage strategies to achieve stable performance of the immunosensor over 24-week storage at room temperature. We evaluated the performance of the immunosensor using COVID-19 patient serum samples collected at different time points after symptom onset. The rapid and sensitive detection of IgG and IgM provided by our immunosensor fulfills the need of rapid COVID-19 serological testing for both point-of-care diagnosis and population immunity screening.
Collapse
Affiliation(s)
- Ran Peng
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, Ontario, M5S 3G8, Canada
| | - Yueyue Pan
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, Ontario, M5S 3G8, Canada
| | - Zhijie Li
- Department of Molecular Genetics, University of Toronto, 361 University Ave, Toronto, Ontario, M5G 1M1, Canada
| | - Zhen Qin
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, Ontario, M5S 3G8, Canada
| | - James M Rini
- Department of Molecular Genetics, University of Toronto, 361 University Ave, Toronto, Ontario, M5G 1M1, Canada; Department of Biochemistry, University of Toronto, 361 University Ave, Toronto, Ontario, M5G 1M1, Canada
| | - Xinyu Liu
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, Ontario, M5S 3G8, Canada; Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario, M5S 3G9, Canada.
| |
Collapse
|
34
|
Rapid and quantitative detection of SARS-CoV-2 IgG antibody in serum using optofluidic point-of-care testing fluorescence biosensor. Talanta 2021; 235:122800. [PMID: 34517658 PMCID: PMC8363510 DOI: 10.1016/j.talanta.2021.122800] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/23/2021] [Accepted: 08/11/2021] [Indexed: 12/25/2022]
Abstract
The COVID-19 pandemic brings unprecedented crisis for public health and economics in the world. Detecting specific antibodies to SARS-CoV-2 is a powerful supplement for the diagnosis of COVID-19 and is important for epidemiological studies and vaccine validations. Herein, a rapid and quantitative detection method of anti-SARS-CoV-2 IgG antibody was built based on the optofluidic point-of-care testing fluorescence biosensor. Without complicated steps needed, the portable system is suitable for on-site sensitive determination of anti-SARS-CoV-2 IgG antibody in serum. Under the optimal conditions, the whole detection procedure is about 25 min with a detection limit of 12.5 ng/mL that can well meet the diagnostic requirements. The method was not obviously affected by IgM and serum matrix and demonstrated to have good stability and reliability in real sample analysis. Compared to ELISA test results, the proposed method exhibits several advantages including wider measurement range and easier operation. The method provides a universal platform for rapid and quantitative analysis of other related biomarkers, which is of significance for the prevention and control of COVID-19 pandemic.
Collapse
|
35
|
Asif M, Xu Y, Xiao F, Sun Y. Diagnosis of COVID-19, vitality of emerging technologies and preventive measures. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2021; 423:130189. [PMID: 33994842 PMCID: PMC8103773 DOI: 10.1016/j.cej.2021.130189] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/22/2021] [Accepted: 05/02/2021] [Indexed: 05/18/2023]
Abstract
Coronavirus diseases-2019 (COVID-19) is becoming increasing serious and major threat to public health concerns. As a matter of fact, timely testing enhances the life-saving judgments on treatment and isolation of COVID-19 infected individuals at possible earliest stage which ultimately suppresses spread of infectious diseases. Many government and private research institutes and manufacturing companies are striving to develop reliable tests for prompt quantification of SARS-CoV-2. In this review, we summarize existing diagnostic methods as manual laboratory-based nucleic acid assays for COVID-19 and their limitations. Moreover, vitality of rapid and point of care serological tests together with emerging biosensing technologies has been discussed in details. Point of care tests with characteristics of rapidity, accurateness, portability, low cost and requiring non-specific devices possess great suitability in COVID-19 diagnosis and detection. Besides, this review also sheds light on several preventive measures to track and manage disease spread in current and future outbreaks of diseases.
Collapse
Affiliation(s)
- Muhammad Asif
- Hubei Key Laboratory of Plasma Chemistry and Advanced Materials, School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Yun Xu
- Hubei Key Laboratory of Material Chemistry and Service Failure, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430205, China
| | - Fei Xiao
- Hubei Key Laboratory of Material Chemistry and Service Failure, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430205, China
| | - Yimin Sun
- Hubei Key Laboratory of Plasma Chemistry and Advanced Materials, School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| |
Collapse
|
36
|
Liu H, Yang A, Song J, Wang N, Lam P, Li Y, Law HKW, Yan F. Ultrafast, sensitive, and portable detection of COVID-19 IgG using flexible organic electrochemical transistors. SCIENCE ADVANCES 2021; 7:eabg8387. [PMID: 34524851 PMCID: PMC8443172 DOI: 10.1126/sciadv.abg8387] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The outbreak of COVID-19 and its continued spread have seriously threatened public health. Antibody testing is essential for infection diagnosis, seroepidemiological analysis, and vaccine evaluation. However, convenient, fast, and accurate antibody detection remains a challenge in this protracted battle. Here, we report an ultrafast, low-cost, label-free, and portable SARS-CoV-2 immunoglobulin G (IgG) detection platform based on organic electrochemical transistors (OECTs), which can be remotely controlled by a mobile phone. To enable faster detection, voltage pulses are applied on the gate electrode of the OECT to accelerate binding between the antibody and antigen. By optimizing ion concentrations and pH values of test solutions, we realize specific detection of SARS-CoV-2 IgG in several minutes with a detectable region from 10 fM to 100 nM, which encompasses the range of serum SARS-CoV-2 IgG levels in humans. These portable sensors show promise for use in diagnosis and prognosis of COVID-19.
Collapse
Affiliation(s)
- Hong Liu
- Department of Applied Physics, The Hong Kong Polytechnic University, Hung Hom, 999077 Kowloon, Hong Kong
| | - Anneng Yang
- Department of Applied Physics, The Hong Kong Polytechnic University, Hung Hom, 999077 Kowloon, Hong Kong
| | - Jiajun Song
- Department of Applied Physics, The Hong Kong Polytechnic University, Hung Hom, 999077 Kowloon, Hong Kong
| | - Naixiang Wang
- Department of Applied Physics, The Hong Kong Polytechnic University, Hung Hom, 999077 Kowloon, Hong Kong
| | - Puiyiu Lam
- Department of Applied Physics, The Hong Kong Polytechnic University, Hung Hom, 999077 Kowloon, Hong Kong
| | - Yuenling Li
- Department of Applied Physics, The Hong Kong Polytechnic University, Hung Hom, 999077 Kowloon, Hong Kong
| | - Helen Ka-wai Law
- Department of Health Technology and Informatics Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, 999077 Kowloon, Hong Kong
| | - Feng Yan
- Department of Applied Physics, The Hong Kong Polytechnic University, Hung Hom, 999077 Kowloon, Hong Kong
- Corresponding author.
| |
Collapse
|
37
|
Escobar A, Chiu P, Qu J, Zhang Y, Xu CQ. Integrated Microfluidic-Based Platforms for On-Site Detection and Quantification of Infectious Pathogens: Towards On-Site Medical Translation of SARS-CoV-2 Diagnostic Platforms. MICROMACHINES 2021; 12:1079. [PMID: 34577722 PMCID: PMC8470930 DOI: 10.3390/mi12091079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/18/2022]
Abstract
The rapid detection and quantification of infectious pathogens is an essential component to the control of potentially lethal outbreaks among human populations worldwide. Several of these highly infectious pathogens, such as Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), have been cemented in human history as causing epidemics or pandemics due to their lethality and contagiousness. SARS-CoV-2 is an example of these highly infectious pathogens that have recently become one of the leading causes of globally reported deaths, creating one of the worst economic downturns and health crises in the last century. As a result, the necessity for highly accurate and increasingly rapid on-site diagnostic platforms for highly infectious pathogens, such as SARS-CoV-2, has grown dramatically over the last two years. Current conventional non-microfluidic diagnostic techniques have limitations in their effectiveness as on-site devices due to their large turnaround times, operational costs and the need for laboratory equipment. In this review, we first present criteria, both novel and previously determined, as a foundation for the development of effective and viable on-site microfluidic diagnostic platforms for several notable pathogens, including SARS-CoV-2. This list of criteria includes standards that were set out by the WHO, as well as our own "seven pillars" for effective microfluidic integration. We then evaluate the use of microfluidic integration to improve upon currently, and previously, existing platforms for the detection of infectious pathogens. Finally, we discuss a stage-wise means to translate our findings into a fundamental framework towards the development of more effective on-site SARS-CoV-2 microfluidic-integrated platforms that may facilitate future pandemic diagnostic and research endeavors. Through microfluidic integration, many limitations in currently existing infectious pathogen diagnostic platforms can be eliminated or improved upon.
Collapse
Affiliation(s)
- Andres Escobar
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada; (A.E.); (J.Q.); (Y.Z.)
| | - Phyllis Chiu
- Department of Engineering Physics, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada;
| | - Jianxi Qu
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada; (A.E.); (J.Q.); (Y.Z.)
| | - Yushan Zhang
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada; (A.E.); (J.Q.); (Y.Z.)
| | - Chang-qing Xu
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada; (A.E.); (J.Q.); (Y.Z.)
- Department of Engineering Physics, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada;
| |
Collapse
|
38
|
Rais Y, Fu Z, Drabovich AP. Mass spectrometry-based proteomics in basic and translational research of SARS-CoV-2 coronavirus and its emerging mutants. Clin Proteomics 2021; 18:19. [PMID: 34384361 PMCID: PMC8358260 DOI: 10.1186/s12014-021-09325-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 08/07/2021] [Indexed: 01/08/2023] Open
Abstract
Molecular diagnostics of the coronavirus disease of 2019 (COVID-19) now mainly relies on the measurements of viral RNA by RT-PCR, or detection of anti-viral antibodies by immunoassays. In this review, we discussed the perspectives of mass spectrometry-based proteomics as an analytical technique to identify and quantify proteins of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and to enable basic research and clinical studies on COVID-19. While RT-PCR and RNA sequencing are indisputably powerful techniques for the detection of SARS-CoV-2 and identification of the emerging mutations, proteomics may provide confirmatory diagnostic information and complimentary biological knowledge on protein abundance, post-translational modifications, protein-protein interactions, and the functional impact of the emerging mutations. Pending advances in sensitivity and throughput of mass spectrometry and liquid chromatography, shotgun and targeted proteomic assays may find their niche for the differential quantification of viral proteins in clinical and environmental samples. Targeted proteomic assays in combination with immunoaffinity enrichments also provide orthogonal tools to evaluate cross-reactivity of serology tests and facilitate development of tests with the nearly perfect diagnostic specificity, this enabling reliable testing of broader populations for the acquired immunity. The coronavirus pandemic of 2019-2021 is another reminder that the future global pandemics may be inevitable, but their impact could be mitigated with the novel tools and assays, such as mass spectrometry-based proteomics, to enable continuous monitoring of emerging viruses, and to facilitate rapid response to novel infectious diseases.
Collapse
Affiliation(s)
- Yasmine Rais
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Zhiqiang Fu
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Andrei P Drabovich
- Division of Analytical and Environmental Toxicology, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
39
|
Yan S, Ahmad KZ, Warden AR, Ke Y, Maboyi N, Zhi X, Ding X. One-pot pre-coated interface proximity extension assay for ultrasensitive co-detection of anti-SARS-CoV-2 antibodies and viral RNA. Biosens Bioelectron 2021; 193:113535. [PMID: 34399194 PMCID: PMC8336976 DOI: 10.1016/j.bios.2021.113535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/12/2021] [Accepted: 07/24/2021] [Indexed: 12/11/2022]
Abstract
In the field of in vitro diagnostics, detection of nucleic acids and proteins from biological samples is typically performed with independent platforms; however, co-detection remains a major technical challenge. Specifically, during the coronavirus disease 2019 (COVID-19) pandemic, the ability to simultaneously detect viral RNA and human antibodies would prove highly useful for efficient diagnosis and disease course management. Herein, we present a multiplex one-pot pre-coated interface proximity extension (OPIPE) assay that facilitates the simultaneous recognition of antibodies using a pre-coated antigen interface and a pair of anti-antibodies labeled with oligonucleotides. Following anti-antibody-bound nucleic acid chain extension to form templates in proximity, antibody signals can be amplified, together with that of targeted RNA, via a reverse transcription-polymerase chain reaction. Using four-color fluorescent TaqMan probes, we demonstrate the co-detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific antibodies and viral nucleic acids in a single bio-complex sample, including nucleocapsid protein-specific IgG and IgM, and the RNA fragments of RdRp and E genes. The serum detection limit for this platform is 100 fg/mL (0.67 fM) for the anti-SARS-CoV-2 antibody and 10 copies/μL for viral RNA. The OPIPE assay offers a practical and affordable solution for ultrasensitive co-detection of nucleic acids and antibodies from the same trace biological sample without the additional requirement of complicated equipment.
Collapse
Affiliation(s)
- Sijia Yan
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Khan Zara Ahmad
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Antony R Warden
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yuqing Ke
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Nokuzola Maboyi
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiao Zhi
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xianting Ding
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
40
|
A plug-and-play platform of ratiometric bioluminescent sensors for homogeneous immunoassays. Nat Commun 2021; 12:4586. [PMID: 34321486 PMCID: PMC8319308 DOI: 10.1038/s41467-021-24874-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 07/08/2021] [Indexed: 01/07/2023] Open
Abstract
Heterogeneous immunoassays such as ELISA have become indispensable in modern bioanalysis, yet translation into point-of-care assays is hindered by their dependence on external calibration and multiple washing and incubation steps. Here, we introduce RAPPID (Ratiometric Plug-and-Play Immunodiagnostics), a mix-and-measure homogeneous immunoassay platform that combines highly specific antibody-based detection with a ratiometric bioluminescent readout. The concept entails analyte-induced complementation of split NanoLuc luciferase fragments, photoconjugated to an antibody sandwich pair via protein G adapters. Introduction of a calibrator luciferase provides a robust ratiometric signal that allows direct in-sample calibration and quantitative measurements in complex media such as blood plasma. We developed RAPPID sensors that allow low-picomolar detection of several protein biomarkers, anti-drug antibodies, therapeutic antibodies, and both SARS-CoV-2 spike protein and anti-SARS-CoV-2 antibodies. With its easy-to-implement standardized workflow, RAPPID provides an attractive, fast, and low-cost alternative to traditional immunoassays, in an academic setting, in clinical laboratories, and for point-of-care applications. Many current immunoassays require multiple washing, incubation and optimization steps. Here the authors present Ratiometric Plug-and-Play Immunodiagnostics (RAPPID), a generic assay platform that uses ratiometric bioluminescent detection to allow sandwich immunoassays to be performed directly in solution.
Collapse
|
41
|
Li L, Zhu C, Zhao X, Qu F. [Applications of separation technology in novel coronavirus research, epidemic prevention and detection]. Se Pu 2021; 39:679-685. [PMID: 34227364 PMCID: PMC9421565 DOI: 10.3724/sp.j.1123.2021.03022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
新型冠状病毒肺炎(COVID-19)疫情的爆发给世界公共卫生安全带来前所未有的挑战。随着新型冠状病毒(SARS-CoV-2)相关研究的不断深入,众多分析检测技术相继被应用,推动了病毒检测、疫苗和创新疗法的研发,从而使疫情早日得到控制。分离技术作为生命科学、医学、药学领域的关键技术,操作简单,分离效率高,选择性强,在新型冠状病毒的分离、检测、诊疗及防疫中起到不可替代的作用。该文以SARS-CoV-2或COVID-19为关键词在ISI Web of Science中进行主题检索,归纳了2020年度新型冠状病毒相关的研究论文,简要介绍主要的研究方向,并对国际顶级学术期刊Nature, Science, Cell的论文发表情况进行了统计。通过检索影响因子较高的期刊,综述了新型冠状病毒研究中主要应用的分离技术,并从亲和色谱和尺寸排阻色谱、液相色谱、磁珠分离、离心、微纳分离以及电泳6个方面进行说明。综述统计了亲和色谱和尺寸排阻色谱纯化的病毒相关蛋白,并介绍了其在新型冠状病毒传播、感染机制以及药物筛选中的应用;介绍了液相色谱对病毒候选药物评估以及复杂基质中单一成分的鉴定;介绍了磁珠分离在细胞分离、核酸提取和免疫学检测中的应用;介绍了离心对病毒颗粒、细胞以及血清的分离;介绍了微纳分离结合其他技术以实现病毒蛋白的高灵敏检测;简要介绍了电泳在聚合酶链式反应(PCR)产物分析中的应用。该文综述了2020年度新型冠状病毒研究和防疫检测中分离技术的应用情况,分析了分离技术在新型冠状病毒检测中发挥的作用,旨在为从事分离研究的科研工作者提供一些参考。
Collapse
Affiliation(s)
- Linsen Li
- School of Life Science, Beijing Institute of Technology, "Molecular Medicine and Biological Diagnosis" Key Laboratory of Ministry of Industry and Information Technology, Beijing 100081, China
| | - Chao Zhu
- School of Life Science, Beijing Institute of Technology, "Molecular Medicine and Biological Diagnosis" Key Laboratory of Ministry of Industry and Information Technology, Beijing 100081, China
| | | | - Feng Qu
- School of Life Science, Beijing Institute of Technology, "Molecular Medicine and Biological Diagnosis" Key Laboratory of Ministry of Industry and Information Technology, Beijing 100081, China
| |
Collapse
|
42
|
Wang C, Wang C, Qiu J, Gao J, Liu H, Zhang Y, Han L. Ultrasensitive, high-throughput, and rapid simultaneous detection of SARS-CoV-2 antigens and IgG/IgM antibodies within 10 min through an immunoassay biochip. Mikrochim Acta 2021; 188:262. [PMID: 34282508 PMCID: PMC8289455 DOI: 10.1007/s00604-021-04896-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/12/2021] [Indexed: 12/24/2022]
Abstract
COVID-19 is now a severe threat to global health. Facing this pandemic, we developed a space-encoding microfluidic biochip for high-throughput, rapid, sensitive, simultaneous quantitative detection of SARS-CoV-2 antigen proteins and IgG/IgM antibodies in serum. The proposed immunoassay biochip integrates the advantages of graphene oxide quantum dots (GOQDs) and microfluidic chip and is capable of conducting multiple SARS-CoV-2 antigens or IgG/IgM antibodies of 60 serum samples simultaneously with only 2 μL sample volume of each patient. Fluorescence intensity of antigens and IgG antibody detection at emission wavelength of ~680 nm was used to quantify the target concentration at excitation wavelength of 632 nm, and emission wavelength of ~519 nm was used during the detection of IgM antibodies at excitation wavelength of 488 nm. The method developed has a large linear quantification detection regime of 5 orders of magnitude, an ultralow detection limit of ~0.3 pg/mL under optimized conditions, and less than 10-min qualitative detection time. The proposed biosensing platform will not only greatly facilitate the rapid diagnosis of COVID-19 patients, but also provide a valuable screening approach for infected patients, medical therapy, and vaccine recipients.
Collapse
Affiliation(s)
- Chunhua Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266000, China
| | - Chao Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266000, China
| | - Jiaoyan Qiu
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266000, China
| | - Jianwei Gao
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266000, China
| | - Hong Liu
- Institute of Crystal Materials, Shandong University, Jinan, 250100, Shandong, China
| | - Yu Zhang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266000, China.
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266000, China.
| |
Collapse
|
43
|
Xu W, Liu J, Song D, Li C, Zhu A, Long F. Rapid, label-free, and sensitive point-of-care testing of anti-SARS-CoV-2 IgM/IgG using all-fiber Fresnel reflection microfluidic biosensor. Mikrochim Acta 2021; 188:261. [PMID: 34278534 PMCID: PMC8286882 DOI: 10.1007/s00604-021-04911-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/21/2021] [Indexed: 01/28/2023]
Abstract
The ongoing global pandemic of SARS-CoV-2 has promoted to develop novel serological testing technologies since they can be effectively complementary to RT-PCR. Here, a new all-fiber Fresnel reflection microfluidic biosensor (FRMB) was constructed through combining all-fiber optical system, microfluidic chip, and multimode fiber bio-probe. The transmission of the incident light and the collection and transmission of Fresnel reflection light are achieved using a single-multi-mode fiber optic coupler (SMFC) without any other optical separation elements. This compact design greatly simplifies the whole system structure and improves light transmission efficiency, which makes it suitable for the label-free, sensitive, and easy-to-use point-of-care testing (POCT) of targets in nanoliter samples. Based on Fresnel reflection mechanism and immunoassay principle, both the SARS-CoV-2 IgM and IgG antibodies against the SARS-CoV-2 spike protein could be sensitively quantified in 7 min using the secondary antibodies-modified multimode fiber bio-probe. The FRMB performs in one-step, is accurate, label-free, and sensitive in situ/on-site detection of SARS-CoV-2 IgM or IgG in serum with simple dilution only. The limits of detection of SARS-CoV-2 IgM and SARS-CoV-2 IgG were 0.82 ng/mL and 0.45 ng/mL, respectively. Based on our proposed theory, the affinity constants of SARS-CoV-2 IgM or IgG antibody and their respective secondary antibodies were also determined. The FRMB can be readily extended as a universal platform for the label-free, rapid, and sensitive in situ/on-site measurement of other biomarkers and the investigation of biomolecular interaction.
Collapse
Affiliation(s)
- Wenjuan Xu
- School of Environment and Natural Resources, Renmin University of China, Beijing, 100872, China
| | - Jiayao Liu
- School of Environment and Natural Resources, Renmin University of China, Beijing, 100872, China
| | - Dan Song
- School of Environment and Natural Resources, Renmin University of China, Beijing, 100872, China
| | - Chunsheng Li
- Biology Institute of Hebei Academy of Sciences, Shijiazhuang, China
| | - Anna Zhu
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China.
| | - Feng Long
- School of Environment and Natural Resources, Renmin University of China, Beijing, 100872, China.
- Department of Chemistry, Renmin University of China, Beijing, 100872, China.
| |
Collapse
|
44
|
Safiabadi Tali SH, LeBlanc JJ, Sadiq Z, Oyewunmi OD, Camargo C, Nikpour B, Armanfard N, Sagan SM, Jahanshahi-Anbuhi S. Tools and Techniques for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)/COVID-19 Detection. Clin Microbiol Rev 2021; 34:e00228-20. [PMID: 33980687 PMCID: PMC8142517 DOI: 10.1128/cmr.00228-20] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory disease coronavirus 2 (SARS-CoV-2), has led to millions of confirmed cases and deaths worldwide. Efficient diagnostic tools are in high demand, as rapid and large-scale testing plays a pivotal role in patient management and decelerating disease spread. This paper reviews current technologies used to detect SARS-CoV-2 in clinical laboratories as well as advances made for molecular, antigen-based, and immunological point-of-care testing, including recent developments in sensor and biosensor devices. The importance of the timing and type of specimen collection is discussed, along with factors such as disease prevalence, setting, and methods. Details of the mechanisms of action of the various methodologies are presented, along with their application span and known performance characteristics. Diagnostic imaging techniques and biomarkers are also covered, with an emphasis on their use for assessing COVID-19 or monitoring disease severity or complications. While the SARS-CoV-2 literature is rapidly evolving, this review highlights topics of interest that have occurred during the pandemic and the lessons learned throughout. Exploring a broad armamentarium of techniques for detecting SARS-CoV-2 will ensure continued diagnostic support for clinicians, public health, and infection prevention and control for this pandemic and provide advice for future pandemic preparedness.
Collapse
Affiliation(s)
- Seyed Hamid Safiabadi Tali
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
- Department of Mechanical, Industrial, and Aerospace Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| | - Jason J LeBlanc
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medicine (Infectious Diseases), Dalhousie University, Halifax, Nova Scotia, Canada
- Division of Microbiology, Department of Pathology and Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
| | - Zubi Sadiq
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| | - Oyejide Damilola Oyewunmi
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| | - Carolina Camargo
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | - Bahareh Nikpour
- Department of Electrical and Computer Engineering, McGill University, Montréal, Québec, Canada
| | - Narges Armanfard
- Department of Electrical and Computer Engineering, McGill University, Montréal, Québec, Canada
- Mila-Quebec AI Institute, Montréal, Québec, Canada
| | - Selena M Sagan
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Sana Jahanshahi-Anbuhi
- Department of Chemical and Materials Engineering, Gina Cody School of Engineering, Concordia University, Montréal, Québec, Canada
| |
Collapse
|
45
|
Zhang Z, Wang X, Wei X, Zheng SW, Lenhart BJ, Xu P, Li J, Pan J, Albrecht H, Liu C. Multiplex quantitative detection of SARS-CoV-2 specific IgG and IgM antibodies based on DNA-assisted nanopore sensing. Biosens Bioelectron 2021; 181:113134. [PMID: 33761415 PMCID: PMC7927651 DOI: 10.1016/j.bios.2021.113134] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/24/2021] [Accepted: 02/27/2021] [Indexed: 02/06/2023]
Abstract
The coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread into a global pandemic. Early and accurate diagnosis and quarantine remain the most effective mitigation strategy. Although reverse transcriptase polymerase chain reaction (RT-qPCR) is the gold standard for COVID-19 diagnosis, recent studies suggest that nucleic acids were undetectable in a significant number of cases with clinical features of COVID-19. Serologic assays that detect human antibodies to SARS-CoV-2 serve as a complementary method to diagnose these cases, as well as to identify asymptomatic cases and qualified convalescent serum donors. However, commercially available enzyme-linked immunosorbent assays (ELISA) are laborious and non-quantitative, while point-of-care assays suffer from low detection accuracy. To provide a serologic assay with high performance and portability for potential point-of-care applications, we developed DNA-assisted nanopore sensing for quantification of SARS-CoV-2 related antibodies in human serum. Different DNA structures were used as detection reporters for multiplex quantification of immunoglobulin M (IgM) and immunoglobulin G (IgG) antibodies against the nucleocapsid protein of SARS-CoV-2 in serum specimens from patients with conformed or suspected infection. Comparing to a clinically used point-of-care assay and an ELISA assay, our technology can reliably quantify SARS-CoV-2 antibodies with higher accuracy, large dynamic range, and potential for assay automation.
Collapse
Affiliation(s)
- Zehui Zhang
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA
| | - Xiaoqin Wang
- Department of Chemical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA
| | - Xiaojun Wei
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA; Department of Chemical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA
| | - Sophia W Zheng
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA
| | - Brian J Lenhart
- Department of Chemical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA
| | - Peisheng Xu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Jie Li
- Department of Chemistry and Biochemistry, College of Arts and Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Jing Pan
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Helmut Albrecht
- Department of Internal Medicine, School of Medicine, University of South Carolina, Columbia, SC 29209, USA; Department of Internal Medicine, Palmetto Health USC Medical Group, Columbia, SC 29203, USA
| | - Chang Liu
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA; Department of Chemical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
46
|
Song Q, Sun X, Dai Z, Gao Y, Gong X, Zhou B, Wu J, Wen W. Point-of-care testing detection methods for COVID-19. LAB ON A CHIP 2021; 21:1634-1660. [PMID: 33705507 DOI: 10.1039/d0lc01156h] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
COVID-19 is an acute respiratory disease caused by SARS-CoV-2, which has high transmissibility. People infected with SARS-CoV-2 can develop symptoms including cough, fever, pneumonia and other complications, which in severe cases could lead to death. In addition, a proportion of people infected with SARS-CoV-2 may be asymptomatic. At present, the primary diagnostic method for COVID-19 is reverse transcription-polymerase chain reaction (RT-PCR), which tests patient samples including nasopharyngeal swabs, sputum and other lower respiratory tract secretions. Other detection methods, e.g., isothermal nucleic acid amplification, CRISPR, immunochromatography, enzyme-linked immunosorbent assay (ELISA) and electrochemical sensors are also in use. As the current testing methods are mostly performed at central hospitals and third-party testing centres, the testing systems used mostly employ large, high-throughput, automated equipment. Given the current situation of the epidemic, point-of-care testing (POCT) is advantageous in terms of its ease of use, greater approachability on the user's end, more timely detection, and comparable accuracy and sensitivity, which could reduce the testing load on central hospitals. POCT is thus conducive to daily epidemic control and achieving early detection and treatment. This paper summarises the latest research advances in POCT-based SARS-CoV-2 detection methods, compares three categories of commercially available products, i.e., nucleic acid tests, immunoassays and novel sensors, and proposes the expectations for the development of POCT-based SARS-CoV-2 detection including greater accessibility, higher sensitivity and lower costs.
Collapse
Affiliation(s)
- Qi Song
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China. and Guangzhou HKUST Fok Ying Tung Research Institute, Guangzhou, Guangdong, China
| | - Xindi Sun
- Materials Genome Institute, Shanghai University, Shanghai, China.
| | - Ziyi Dai
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Macau, China.
| | - Yibo Gao
- Shenzhen Shineway Technology Corporation, Shenzhen, Guangdong, China
| | - Xiuqing Gong
- Materials Genome Institute, Shanghai University, Shanghai, China.
| | - Bingpu Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Macau, China.
| | - Jinbo Wu
- Materials Genome Institute, Shanghai University, Shanghai, China.
| | - Weijia Wen
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
47
|
Yang X, Gong C, Wang Y, Luo Y, Rao YJ, Peng GD, Gong Y. A sequentially bioconjugated optofluidic laser for wash-out-free and rapid biomolecular detection. LAB ON A CHIP 2021; 21:1686-1693. [PMID: 33949394 DOI: 10.1039/d0lc01332c] [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
Microstructures can improve both sensitivity and assay time in heterogeneous assays (such as ELISA) for biochemical analysis; however, it remains a challenge to perform the essential wash process in those microstructure-based heterogeneous assays. Here, we propose a sequential bioconjugation protocol to solve this problem and demonstrate a new type of fiber optofluidic laser for biosensing. Except for acting as an optical microresonator and a microstructured substrate, the miniaturized hollow optical fiber (HOF) is used as a microfluidic channel for storing and transferring reagents thanks to its capability in length extension. Through the capillary action, different reagents were sequentially withdrawn into the fiber for specific binding and washing purposes. By using the sequentially bioconjugated FOFL, avidin molecules are detected based on competitive binding with a limit of detection of 9.5 pM, ranging from 10 pM to 100 nM. It is demonstrated that a short incubation time of 10 min is good enough to allow the biomolecules to conjugate on the inner surface of the HOF. Owing to its miniaturized size, only 589 nL of liquid is required for incubation, which reduces the sample consumption and cost for each test. This work provides a tool to exploit the potential of microstructured optical fibers in high-performance biosensing.
Collapse
Affiliation(s)
- Xi Yang
- Key Laboratory of Optical Fiber Sensing and Communications (Ministry of Education of China), School of Information and Communication Engineering, University of Electronic Science and Technology of China, No. 2006, Xiyuan Ave., Chengdu, 611731 China.
| | - Chaoyang Gong
- School of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Ave, Singapore, 639798 Singapore
| | - Yanqiong Wang
- Key Laboratory of Optical Fiber Sensing and Communications (Ministry of Education of China), School of Information and Communication Engineering, University of Electronic Science and Technology of China, No. 2006, Xiyuan Ave., Chengdu, 611731 China.
| | - Yanhua Luo
- School of Electrical Engineering and Telecommunications, University of New South Wales, Sydney, NSW 2052, Australia
| | - Yun-Jiang Rao
- Key Laboratory of Optical Fiber Sensing and Communications (Ministry of Education of China), School of Information and Communication Engineering, University of Electronic Science and Technology of China, No. 2006, Xiyuan Ave., Chengdu, 611731 China. and Research Center for Optical Fiber Sensing, Zhejiang Laboratory, Hangzhou, Zhejiang 310000, China
| | - Gang-Ding Peng
- School of Electrical Engineering and Telecommunications, University of New South Wales, Sydney, NSW 2052, Australia
| | - Yuan Gong
- Key Laboratory of Optical Fiber Sensing and Communications (Ministry of Education of China), School of Information and Communication Engineering, University of Electronic Science and Technology of China, No. 2006, Xiyuan Ave., Chengdu, 611731 China.
| |
Collapse
|
48
|
Ghodake GS, Shinde SK, Kadam AA, Saratale RG, Saratale GD, Syed A, Elgorban AM, Marraiki N, Kim DY. Biological characteristics and biomarkers of novel SARS-CoV-2 facilitated rapid development and implementation of diagnostic tools and surveillance measures. Biosens Bioelectron 2021; 177:112969. [PMID: 33434780 PMCID: PMC7836906 DOI: 10.1016/j.bios.2021.112969] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/30/2020] [Accepted: 01/02/2021] [Indexed: 01/08/2023]
Abstract
Existing coronavirus named as a severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has speeded its spread across the globe immediately after emergence in China, Wuhan region, at the end of the year 2019. Different techniques, including genome sequencing, structural feature classification by electron microscopy, and chest imaging using computed tomography, are primarily used to diagnose and screen SARS-CoV-2 suspected individuals. Determination of the viral structure, surface proteins, and genome sequence has provided a design blueprint for the diagnostic investigations of novel SARS-CoV-2 virus and rapidly emerging diagnostic technologies, vaccine trials, and cell-entry-inhibiting drugs. Here, we describe recent understandings on the spike glycoprotein (S protein), receptor-binding domain (RBD), and angiotensin-converting enzyme 2 (ACE2) and their receptor complex. This report also aims to review recently established diagnostic technologies and developments in surveillance measures for SARS-CoV-2 as well as the characteristics and performance of emerging techniques. Smartphone apps for contact tracing can help nations to conduct surveillance measures before a vaccine and effective medicines become available. We also describe promising point-of-care (POC) diagnostic technologies that are under consideration by researchers for advancement beyond the proof-of-concept stage. Developing novel diagnostic techniques needs to be facilitated to establish automatic systems, without any personal involvement or arrangement to curb an existing SARS-CoV-2 epidemic crisis, and could also be appropriate for avoiding the emergence of a future epidemic crisis.
Collapse
Affiliation(s)
- Gajanan Sampatrao Ghodake
- Department of Biological and Environmental Science, Dongguk University-Seoul, Medical Center Ilsan, Goyang-si, 10326, Gyeonggi-do, South Korea
| | - Surendra Krushna Shinde
- Department of Biological and Environmental Science, Dongguk University-Seoul, Medical Center Ilsan, Goyang-si, 10326, Gyeonggi-do, South Korea
| | - Avinash Ashok Kadam
- Research Institute of Biotechnology and Medical Converged Science, Dongguk University-Seoul, Ilsandong-gu, Goyang-si, 10326, Gyeonggi-do, South Korea
| | - Rijuta Ganesh Saratale
- Research Institute of Biotechnology and Medical Converged Science, Dongguk University-Seoul, Ilsandong-gu, Goyang-si, 10326, Gyeonggi-do, South Korea
| | - Ganesh Dattatraya Saratale
- Department of Food Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, 10326, Gyeonggi-do, South Korea
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455 Riyadh, 11451, Saudi Arabia
| | - Abdallah M Elgorban
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455 Riyadh, 11451, Saudi Arabia
| | - Najat Marraiki
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455 Riyadh, 11451, Saudi Arabia
| | - Dae-Young Kim
- Department of Biological and Environmental Science, Dongguk University-Seoul, Medical Center Ilsan, Goyang-si, 10326, Gyeonggi-do, South Korea.
| |
Collapse
|
49
|
Chen L, Zhang G, Liu L, Li Z. Emerging biosensing technologies for improved diagnostics of COVID-19 and future pandemics. Talanta 2021; 225:121986. [PMID: 33592734 PMCID: PMC7733602 DOI: 10.1016/j.talanta.2020.121986] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/25/2020] [Accepted: 12/05/2020] [Indexed: 02/07/2023]
Abstract
Diagnostic tools play significant roles in the fight against COVID-19 and other pandemics. Existing tests, such as RT-qPCR, have limitations including long assay time, low throughput, inadequate sensitivity, and suboptimal portability. Emerging biosensing technologies hold the promise to develop tests that are rapid, highly sensitive, and suitable for point-of-care testing, which could significantly facilitate the testing of COVID-19. Despite that, practical applications of such biosensors in pandemics have yet to be achieved. In this review, we consolidate the newly developed diagnostic tools for COVID-19 using emerging biosensing technologies and discuss their application promise. In particular, we present nucleic acid tests and antibody tests of COVID-19 based on both conventional and emerging biosensing methods. We then provide perspectives on the existing challenges and potential solutions.
Collapse
Affiliation(s)
- Linzhe Chen
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Guoliang Zhang
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | | | - Zida Li
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
50
|
Aziz A, Asif M, Ashraf G, Farooq U, Yang Q, Wang S. Trends in biosensing platforms for SARS-CoV-2 detection: A critical appraisal against standard detection tools. Curr Opin Colloid Interface Sci 2021; 52:101418. [PMID: 33495685 PMCID: PMC7817481 DOI: 10.1016/j.cocis.2021.101418] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In this ongoing theme of coronavirus disease 2019 (COVID-19) pandemic, highly sensitive analytical testing platforms are extremely necessary to detect SARS-CoV-2 RNA and antiviral antibodies. To limit the viral spread, prompt and precise diagnosis is crucial to facilitate treatment and ensure effective isolation. Accurate detection of antibodies (IgG and IgM) is imperative to understand the prevalence of SARS-CoV-2 in public and to inspect the proportion of immune individuals. In this review, we demonstrate and evaluate some tests that have been used commonly to detect SARS-CoV-2. These include nucleic acid and serological tests for the detection of SARS-CoV-2 RNA and specific antibodies in infected people. Moreover, the vitality of biosensing technologies emphasizing on optical and electrochemical biosensors toward the detection of SARS-CoV-2 has also been discussed here. The early diagnosis of COVID-19 based on detection of reactive oxygen species overproduction because of virus-induced dysfunctioning of lung cells has also been highlighted.
Collapse
Affiliation(s)
- Ayesha Aziz
- Advanced Biomaterials and Tissue Engineering Centre, School of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Muhammad Asif
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Ghazala Ashraf
- Advanced Biomaterials and Tissue Engineering Centre, School of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Umer Farooq
- Advanced Biomaterials and Tissue Engineering Centre, School of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Qiaoli Yang
- Advanced Biomaterials and Tissue Engineering Centre, School of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Shenqi Wang
- Advanced Biomaterials and Tissue Engineering Centre, School of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| |
Collapse
|