1
|
Hu F, Zhang Y, Yang Y, Peng L, Cui S, Ma Q, Wang F, Wang X. A rapid and ultrasensitive RPA-assisted CRISPR-Cas12a/Cas13a nucleic acid diagnostic platform with a smartphone-based portable device. Biosens Bioelectron 2025; 280:117428. [PMID: 40179699 DOI: 10.1016/j.bios.2025.117428] [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: 04/03/2024] [Revised: 03/27/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025]
Abstract
The spread of infectious diseases can be controlled by early identification of the source of infection and timely diagnosis to stop transmission. Real-time fluorescence quantitative polymerase chain reaction (PCR) is the current gold standard for pathogen diagnosis, with high detection sensitivity and accuracy. However, due to the need for specialized equipment, laboratories, and personnel, it is difficult to achieve rapid and immediate diagnosis during large-scale infectious disease outbreaks. Herein, an optimized CRISPR-based nucleic acid detection method was developed that reduces the CRISPR detection time to 15 min while maintaining high sensitivity. By using nucleic acid extraction-free and lyophilization techniques, the 'sample-in-result-out' detection of the two target genes of SARS-CoV-2, the human internal reference gene, and the negative quality control sample can be completed in 20 min, with a sensitivity of 0.5 copies/μL. Additionally, to facilitate the application, a smartphone-based reverse transcription-recombinase polymerase amplification (RT-RPA)-assisted CRISPR-rapid, portable nucleic acid detection device was developed, integrating functions such as heating, centrifugation, mixing, optical detection and result output. Process control, output, and uploading of detection results were conducted through smartphones. The device is not dependent on a power supply and can perform on-site rapid virus detection in resource-limited settings. Real-time uploading of results helps to rapidly implement epidemic prevention and control measures, providing an innovative means of detection, control, and prevention of virus-based infectious diseases. This important work provides a new and effective tool to manage potential future outbreaks of infectious diseases.
Collapse
Affiliation(s)
- Fei Hu
- State Key Laboratory for Manufacturing Systems Engineering, School of Instrument Science and Techonology, Xi'an Jiaotong University, Xi'an, 710054, China.
| | - Yunyun Zhang
- State Key Laboratory for Manufacturing Systems Engineering, School of Instrument Science and Techonology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Yue Yang
- State Key Laboratory for Manufacturing Systems Engineering, School of Instrument Science and Techonology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Lixin Peng
- Windermere Preparatory School, Florida, 34786, United States
| | - Shuhui Cui
- State Key Laboratory for Manufacturing Systems Engineering, School of Instrument Science and Techonology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Qing Ma
- State Key Laboratory for Manufacturing Systems Engineering, School of Instrument Science and Techonology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Fangning Wang
- State Key Laboratory for Manufacturing Systems Engineering, School of Instrument Science and Techonology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Xincheng Wang
- State Key Laboratory for Manufacturing Systems Engineering, School of Instrument Science and Techonology, Xi'an Jiaotong University, Xi'an, 710054, China
| |
Collapse
|
2
|
Wu H, Diao J, Li X, Yue D, He G, Jiang X, Li P. Hydrogel-based 3D printing technology: From interfacial engineering to precision medicine. Adv Colloid Interface Sci 2025; 341:103481. [PMID: 40132296 DOI: 10.1016/j.cis.2025.103481] [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: 09/27/2024] [Revised: 01/03/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Advances in 3D printing technology and the development of hydrogel-based inks have significantly enhanced the potential of precision medicine, promoting progress in medical diagnosis and treatment. The development of 3D printing enables the fabrication of complex gradient structures that emulate natural tissue environments, while advancements in interface engineering facilitate the precise control of interface properties, thereby enhancing the performance of hydrogels in biomedical applications. This review focuses on the latest advancements in three critical 3D printing application areas: efficient real-time detection, drug delivery systems, and regenerative medicine. The application of 3D printing technology enhances nucleic acid-based molecular diagnostic platforms and wearable biosensors for real-time monitoring of physiological parameters, thereby providing robust support for early disease diagnosis. Additionally, it facilitates the development of targeted and controlled drug delivery systems, which offer promising methods for efficient drug utilization, and enables the construction of complex tissue and organ structures with bioactivity and functionality, providing new solutions for regenerative medicine. Collectively, these advancements propel the ongoing progress and development of precision medicine. Furthermore, the challenges associated with 3D printing technology in these three major applications are discussed along with an outlook on prospects.
Collapse
Affiliation(s)
- Haojie Wu
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian, Liaoning 116023, China; State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Jibo Diao
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Xinrong Li
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Dongmei Yue
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian, Liaoning 116023, China
| | - Gaohong He
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Xiaobin Jiang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China.
| | - Peipei Li
- Liaoning Ocean and Fisheries Science Research Institute, Liaoning Academy of Agricultural Sciences, Dalian, Liaoning 116023, China.
| |
Collapse
|
3
|
Ding M, Yang Y, Sun N, He Y, Dong Z, Gao Q, Tian B. Catalytic hairpin assembly-and-cyclization aptasensing for AND-logic detection of protein- and RNA-targets in ribonucleoprotein. Biosens Bioelectron 2025; 279:117388. [PMID: 40132285 DOI: 10.1016/j.bios.2025.117388] [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: 01/20/2025] [Revised: 03/04/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025]
Abstract
Both immunological and molecular diagnostics are essential in disease control. However, due to the different technical routes, existing diagnostic tools mainly focus on either immunological or molecular targets at a time, resulting in restricted information for assessment. Herein, we report a DNA operational amplifier circuit for AND-logic analysis of nucleoprotein and RNA of SARS-CoV-2 ribonucleoprotein. The operator is realized using an aptamer-hairpin probe and a padlock-hairpin probe for nucleoprotein- and ligase-catalyzed hairpin assembly-and-cyclization (CHAC). Given approximately 1000 nucleoproteins per virion in coronavirus, a rolling circle amplification (RCA)-based preamplifier is applied to adjust the input bias by converting the target sequence into an aptamer-hairpin input for CHAC. After CHAC, cyclized padlock-hairpin probes trigger another round of RCA as a post-operator amplifier, producing amplicon coils that aggregate detection probe-modified magnetic nanoparticles. These stepwise homogeneous reaction processes are conducted in a single tube for optomagnetic sensing, offering detection limits of 0.07 ng/mL and 1.5 fM for the protein and the molecular targets of SARS-CoV-2, respectively. The stability, specificity, and accuracy of the circuit are validated by testing serum samples, salmon sperm samples, biased inputs, and 33 clinical nasopharyngeal swab specimens, demonstrating the practicability of simultaneously analyzing immunological and molecular targets for accurate diagnostics.
Collapse
Affiliation(s)
- Mingming Ding
- Department of Biomedical Engineering, Xiangya School of Basic Medical Sciences, Central South University, Changsha, 410013, China
| | - Yulin Yang
- Department of Biomedical Engineering, Xiangya School of Basic Medical Sciences, Central South University, Changsha, 410013, China
| | - Nan Sun
- Department of Biomedical Engineering, Xiangya School of Basic Medical Sciences, Central South University, Changsha, 410013, China
| | - Yilong He
- Department of Biomedical Engineering, Xiangya School of Basic Medical Sciences, Central South University, Changsha, 410013, China
| | - Zhuxin Dong
- Department of Biomedical Engineering, Xiangya School of Basic Medical Sciences, Central South University, Changsha, 410013, China; Furong Laboratory, Changsha, 410008, China
| | - Qian Gao
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Bo Tian
- Department of Biomedical Engineering, Xiangya School of Basic Medical Sciences, Central South University, Changsha, 410013, China; Furong Laboratory, Changsha, 410008, China.
| |
Collapse
|
4
|
Campuzano S, Ruiz-Valdepeñas Montiel V, Torrente-Rodríguez RM, Pingarrón JM. Bioelectroanalytical Technologies for Advancing the Frontiers To Democratize Personalized Desired Health. Anal Chem 2025. [PMID: 40364744 DOI: 10.1021/acs.analchem.5c01450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
The breakthroughs experienced in the development of cutting-edge, reliable, and multipurpose (bio)electroanalytical technologies and their successful incursion into underexplored scenarios have demonstrated their unique potential to act as enablers of the ongoing transformation from reactive to predictive, preventive, personalized, and participatory healthcare, currently known as "P4" medicine. This transformation, more than a vision, is a necessity to achieve a new generation of more efficient, sustainable, and tailored to individual needs healthcare. This promising outlook is the focus of this Perspective, which in addition to highlighting some of the most shocking related research over the last 5 years, offers a prospective and insightful view of the opportunities and imminent advances that shape the future of these fascinating technologies.
Collapse
Affiliation(s)
- Susana Campuzano
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, Pza. de las Ciencias 2, Madrid 28040, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, Madrid 28029, Spain
| | - Víctor Ruiz-Valdepeñas Montiel
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, Pza. de las Ciencias 2, Madrid 28040, Spain
| | - Rebeca M Torrente-Rodríguez
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, Pza. de las Ciencias 2, Madrid 28040, Spain
| | - José M Pingarrón
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, Pza. de las Ciencias 2, Madrid 28040, Spain
| |
Collapse
|
5
|
Mirjalili S, Ikbal MA, Hou CW, Mohammadi MK, Choi Y, Kelbauskas L, VanBlargan LA, Hogue BG, Murugan V, Diamond MS, Wang C. Nanoparticle-supported, rapid, digital quantification of neutralizing antibodies against SARS-CoV-2 variants. Biosens Bioelectron 2025; 285:117549. [PMID: 40383030 DOI: 10.1016/j.bios.2025.117549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 04/23/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
The measurement of neutralizing immune responses to viral infection is essential, given the heterogeneity of human immunity and the emergence of new virus strains. However, neutralizing antibody (nAb) assays often require high-level biosafety containment, sophisticated instrumentation, and long detection times. Here, as a proof-of-principle, we designed a nanoparticle-supported, rapid, electronic detection (NasRED) assay to assess the neutralizing potency of monoclonal antibodies (mAbs) against SARS-CoV-2. The gold nanoparticles (AuNPs) coated with human angiotensin-converting enzyme 2 (ACE2) protein as nAb potency reporters were mixed with the mAbs to be tested, as well as streptavidin-conjugated multivalent spike (S) protein or their receptor binding domains (RBD). High-affinity and ACE2-competitive nAbs alter the S (or RBD)-to-ACE2 binding level and modulate AuNP cluster formation and precipitation. The amount of free-floating AuNP reporters is quantified by a semiconductor-based readout system that measures the AuNPs' optical extinction, producing nAb signals that can differentiate SARS-CoV-2 variants (Wuhan-Hu-1, Gamma, and Omicron). The modular design nature, short assay time (less than 30 min), and portable and inexpensive readout system make this NasRED-nAb assay applicable to measuring vaccine potency, immune responses to infection, and the efficacy of antibody-based therapies.
Collapse
Affiliation(s)
- Seyedsina Mirjalili
- School of Electrical, Computer, and Energy Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biodesign Center for Molecular Design and Biomimetics, Arizona State University, AZ, 85287, USA
| | - Md Ashif Ikbal
- School of Electrical, Computer, and Energy Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biodesign Center for Molecular Design and Biomimetics, Arizona State University, AZ, 85287, USA
| | - Ching-Wen Hou
- Biodesign Center for Personalized Diagnostics, Arizona State University, AZ, 85287, USA
| | - Maziyar Kalateh Mohammadi
- School of Electrical, Computer, and Energy Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biodesign Center for Molecular Design and Biomimetics, Arizona State University, AZ, 85287, USA
| | - Yeji Choi
- School of Electrical, Computer, and Energy Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biodesign Center for Molecular Design and Biomimetics, Arizona State University, AZ, 85287, USA
| | - Laimonas Kelbauskas
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, AZ, 85287, USA
| | | | - Brenda G Hogue
- Biodesign Center for Applied Structural Discovery, Arizona State University, AZ, 85287, USA; School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Vel Murugan
- Biodesign Center for Personalized Diagnostics, Arizona State University, AZ, 85287, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chao Wang
- School of Electrical, Computer, and Energy Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biodesign Center for Molecular Design and Biomimetics, Arizona State University, AZ, 85287, USA.
| |
Collapse
|
6
|
Sun Y, Liu K, Zhang H, Zhao Y, Wen J, Zhao M, Li X, Li Z. A tube-based biosensor for DNA and RNA detection. SCIENCE ADVANCES 2025; 11:eadu2271. [PMID: 40315319 PMCID: PMC12047445 DOI: 10.1126/sciadv.adu2271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/28/2025] [Indexed: 05/04/2025]
Abstract
Affordable, sensitive, and simplified DNA/RNA detection is important for disease diagnosis and enables timely medical intervention measures. Usually, high sensitivity depends on expensive instruments and sophisticated procedures, making sensitivity contradict affordability and simplicity. Here, we proposed an ultra-sensitive single-tube biosensor (USTB) where users can visually detect targets by observing the liquid motion state in a glass tube. The developed instrument-free USTB performed low-cost ($0.1), fast (1 min), and ultra-sensitive detection for both the DNA/RNA fragments (≤1 aM) and the clinical positive samples, which commercial reverse transcription polymerase chain reaction (RT-PCR) and PCR kits could not effectively recognize. Furthermore, USTB is promising to be easily applied to detect other-type biomarkers by the designed smart sensing unit.
Collapse
Affiliation(s)
| | | | - Haoli Zhang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing 100083, PR China
| | - Yuecan Zhao
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing 100083, PR China
| | - Jingshuai Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing 100083, PR China
| | - Meng Zhao
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing 100083, PR China
| | - Xiang Li
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing 100083, PR China
| | - Zhengping Li
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing 100083, PR China
| |
Collapse
|
7
|
Ashok D, Singh J, Howard HR, Cottam S, Waterhouse A, Bilek MMM. Interfacial engineering for biomolecule immobilisation in microfluidic devices. Biomaterials 2025; 316:123014. [PMID: 39708778 DOI: 10.1016/j.biomaterials.2024.123014] [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: 07/24/2024] [Revised: 11/25/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Microfluidic devices are used for various applications in biology and medicine. From on-chip modelling of human organs for drug screening and fast and straightforward point-of-care (POC) detection of diseases to sensitive biochemical analysis, these devices can be custom-engineered using low-cost techniques. The microchannel interface is essential for these applications, as it is the interface of immobilised biomolecules that promote cell capture, attachment and proliferation, sense analytes and metabolites or provide enzymatic reaction readouts. However, common microfluidic materials do not facilitate the stable immobilisation of biomolecules required for relevant applications, making interfacial engineering necessary to attach biomolecules to the microfluidic surfaces. Interfacial engineering is performed through various immobilisation mechanisms and surface treatment techniques, which suitably modify the surface properties like chemistry and energy to obtain robust biomolecule immobilisation and long-term storage stability suitable for the final application. In this review, we provide an overview of the status of interfacial engineering in microfluidic devices, covering applications, the role of biomolecules, their immobilisation pathways and the influence of microfluidic materials. We then propose treatment techniques to optimise performance for various biological and medical applications and highlight future areas of development.
Collapse
Affiliation(s)
- Deepu Ashok
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia; School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; Heart Research Institute, Newtown, NSW, 2042, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; School of Physics, Faculty of Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Jasneil Singh
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; Heart Research Institute, Newtown, NSW, 2042, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Henry Robert Howard
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Sophie Cottam
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia; School of Physics, Faculty of Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Anna Waterhouse
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Marcela M M Bilek
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia; The Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia; School of Physics, Faculty of Science, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
8
|
He Y, Jiang M, Liang Z, Luo Z, Qin J, Shen Y, Gu Y, Ma X, Wang H, Li X, Shi Y, Chen Y, Pu K, Li J. Lab-in-a-Tip: a multiplex immunoassay platform based on a self-assembled barcoded protein array. Nat Commun 2025; 16:3990. [PMID: 40295512 PMCID: PMC12037755 DOI: 10.1038/s41467-025-59390-1] [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: 09/13/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
High throughput immunoassay is increasingly crucial for both scientific and clinical applications. Here we propose a "Lab-in-a-Tip" (LIT) concept to fabricate a pipette tip containing a high-density protein array and other essential reagents. The protein array is made by self-assembling digitally encoded microparticles inside the modified tip. Mounted on a robotic workstation, it automates liquid-handling steps. Notably, compared with Luminex, the current gold standard in multiplex immunoassays, such a design enables LIT to demonstrate multiple advantages in terms of analytical sensitivity, speed, and throughput. It detects analyte concentrations as low as fg/ml, representing a sensitivity improvement of two orders of magnitude over Luminex. Incubation time is reduced to 15 minutes from Luminex's 210 minutes. Furthermore, LIT requires only 10 µl of sample, one-fifth of what Luminex needs. This makes LIT ideal for rapid diagnostics and studies with limited biological samples, greatly expanding its application scope.
Collapse
Affiliation(s)
- Yiran He
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China
| | - Min Jiang
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China
| | - Zhenlong Liang
- Department of laboratory, the first Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zhaoxu Luo
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China
| | - Jingyi Qin
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Ye Shen
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China
| | - Yayun Gu
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China
| | - Xiaodong Ma
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China
| | - Hong Wang
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China
| | - Xin Li
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China
| | - Ying Shi
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China
| | - Yanhua Chen
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China
| | - Kefeng Pu
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China.
| | - Jiong Li
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, China.
| |
Collapse
|
9
|
Lu P, Zhou Y, Niu X, Zhan C, Lang P, Zhao Y, Chen Y. Deep-Learning-Assisted Microfluidic Immunoassay via Smartphone-Based Imaging Transcoding System for On-Site and Multiplexed Biosensing. NANO LETTERS 2025; 25:6803-6812. [PMID: 40203242 DOI: 10.1021/acs.nanolett.5c01435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Point-of-care testing (POCT) with multiplexed capability, ultrahigh sensitivity, affordable smart devices, and user-friendly operation is critically needed for clinical diagnostics and food safety. This study presents a deep-learning-assisted microfluidic immunoassay platform that uses a smartphone-based imaging transcoding system, polystyrene microsphere-based encoding, and artificial-intelligence-assisted decoding. Microspheres of varying sizes act as multiprobes, with their quantities correlating to target concentrations after an immunoreaction and separation-filtration within the microfluidic chip. A smartphone with intelligent decoding software captures images of multiprobes from the chip and performs classification, counting, and concentration calculations. The "encoding-decoding" strategy and integrated microfluidic chip design allow these processes to be completed in simple steps, eliminating the need for additional immunomagnetic separation. As a proof of concept, this platform successfully detected multiple respiratory viruses and antibiotics in various real samples with high sensitivity within 30 min, demonstrating great potential as a smart, universal toolkit for next-generation POCT applications.
Collapse
Affiliation(s)
- Peng Lu
- College of Engineering, Huazhong Agricultural University, Wuhan 430070, China
| | - Yang Zhou
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Xiaohu Niu
- College of Engineering, Huazhong Agricultural University, Wuhan 430070, China
| | - Chen Zhan
- College of Engineering, Huazhong Agricultural University, Wuhan 430070, China
| | - Pengzhou Lang
- Henan Mechanical & Electrical Vocational College, Zhengzhou 451192, China
| | - Yongkun Zhao
- College of Engineering, Huazhong Agricultural University, Wuhan 430070, China
| | - Yiping Chen
- College of Engineering, Huazhong Agricultural University, Wuhan 430070, China
- Dalian Jinshiwan Laboratory, Dalian 116034, China
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
10
|
Jiao JB, Kang Q, Cui SX, Cao JL, Lin T, Ma CJ, Xiao ZH, Du T, Wang N, Du XJ, Wang S. Target-driven functionalized DNA hydrogel capillary sensor for SARS-CoV-2 dual-mode detection. Talanta 2025; 285:127342. [PMID: 39644672 DOI: 10.1016/j.talanta.2024.127342] [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: 10/22/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Coronavirus disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused secondary pandemic, which still poses a serious threat to physical health and economic development. Herein, the target-driven functionalized DNA hydrogel capillary sensor based on cascade signal amplification and carbon coated cobalt manganese modified by prussian blue and platinum nanoparticles (MnCo@C-Pt-PB NPs) has been successfully developed for dual-mode detection of SARS-CoV-2. The cascade signal amplification triggered by target RNA causes the permeability of the DNA hydrogel loaded in the capillary to be destroyed, thereby releasing the embedded MnCo@C-Pt-PB NPs as signal molecules into 3,3',5,5'-tetramethylbenzidine/hydrogen peroxide (TMB/H2O2) solution under the driving of capillarity. The colorless TMB is then catalyzed to blue oxidation products (oxTMB) due to peroxidase-like activity of MnCo@C-Pt-PB NPs, and MnCo@C-Pt-PB NPs and oxTMB with photothermal properties synergistically increase the system temperature under near-infrared irradiation, which are recorded by portable devices to achieve dual-mode detection. Signals intensity are proportional to the logarithm of T-RNA concentration in a wide detection range (100 aM-100 pM), with a detection limit of 100 aM. Moreover, the reliability of the developed method in oropharyngeal swabs samples has also been validated. The signal conversion and amplification function of functionalized DNA hydrogel enhances the convenience, sensitivity and versatility of the developed method, which is promising to be applied in environmental safety, molecular diagnostic assays and disease prevention.
Collapse
Affiliation(s)
- Jing-Bo Jiao
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Qing Kang
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Shu-Xin Cui
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Jiang-Li Cao
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Tong Lin
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Chen-Jing Ma
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ze-Hui Xiao
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ting Du
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Nan Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China.
| | - Xin-Jun Du
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China.
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
11
|
Lu TC, Xiao WB, Tian HY, Qiu QZ, Zhu YT, Chen ZH, Li X, Chen YZ, Lei Y, Liu AL. Rapid Bacterial/Viral Infections Typing Strategy Using a Portable Dual-Channel Electrochemical Biosensor Based on One-Step Assembly of Immunomagnetic Beads. Anal Chem 2025; 97:5953-5964. [PMID: 40068971 DOI: 10.1021/acs.analchem.4c03442] [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: 03/26/2025]
Abstract
Amidst multiple epidemics, a rapid, sensitive, economical, and portable infection diagnosis strategy is crucial for primary medical care, particularly through the analysis of pathogen sources to determine appropriate antibiotic use. C-reactive protein (CRP) and serum amyloid A (SAA) are host-related biomarkers, and their combined detection can effectively distinguish between bacterial and viral infections, which holds great significance for the diagnosis of unknown pathogens. In this work, a portable dual-channel electrochemical biosensor based on a one-step assembly of immunomagnetic beads was proposed for the on-site combined detection of plasma CRP and SAA, which streamlined the operation and shortened the minimum detection time to less than 3 min. The biosensor exhibited excellent linearity in the detection of 3.125-1250 ng/mL CRP and 31.25-1250 ng/mL SAA, with detection limits of 0.91 and 12 ng/mL, respectively, falling within the clinically relevant reference range. Through simulated sample tests, the biosensor effectively distinguished between bacterial infection, viral infection, and healthy plasma samples. The actual sample tests demonstrated a high correlation and comparable medical value to enzyme-linked immunosorbent assay. Overall, this proposed strategy showed potential to aid in infection diagnosis and enable rapid combined detection of multiple biomarkers.
Collapse
Affiliation(s)
- Tai-Cheng Lu
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, The School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Wen-Biao Xiao
- Department of Critical Care Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Hui-Yun Tian
- Fujian Institute of Hematology, The Affiliated Union Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Qing-Zhen Qiu
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, The School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Yu-Ting Zhu
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, The School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Zhen-Hua Chen
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, The School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Xin Li
- Fujian Institute of Hematology, The Affiliated Union Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Yuan-Zhong Chen
- Fujian Institute of Hematology, The Affiliated Union Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Yun Lei
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, The School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Ai-Lin Liu
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, The School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
12
|
Xin X, Su J, Cui H, Wang L, Song S. Recent Advances in Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-Associated Proteins System-Based Biosensors. BIOSENSORS 2025; 15:155. [PMID: 40136952 PMCID: PMC11939850 DOI: 10.3390/bios15030155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
High-sensitivity and high-specificity biodetection is critical for advancing applications in life sciences, biosafety, food safety, and environmental monitoring. CRISPR/Cas systems have emerged as transformative tools in biosensing due to their unparalleled specificity, programmability, and unique enzymatic activities. They exhibit two key cleavage behaviors: precise ON-target cleavage guided by specific protospacers, which ensures accurate target recognition, and bystander cleavage activity triggered upon target binding, which enables robust signal amplification. These properties make CRISPR/Cas systems highly versatile for designing biosensors for ultra-sensitive detection. This review comprehensively explores recent advancements in CRISPR/Cas system-based biosensors, highlighting their impact on improving biosensing performance. We discuss the integration of CRISPR/Cas systems with diverse signal readout mechanisms, including electrochemical, fluorescent, colorimetric, surface-enhanced Raman scattering (SERS), and so on. Additionally, we examine the development of integrated biosensing systems, such as microfluidic devices and portable biosensors, which leverage CRISPR/Cas technology for point-of-care testing (POCT) and high-throughput analysis. Furthermore, we identify unresolved challenges, aiming to inspire innovative solutions and accelerate the translation of these technologies into practical applications for diagnostics, food, and environment safety.
Collapse
Affiliation(s)
- Xianglin Xin
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai 200444, China; (X.X.); (H.C.); (L.W.)
| | - Jing Su
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, No. 100 Haiquan Road, Shanghai 201418, China
| | - Haoran Cui
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai 200444, China; (X.X.); (H.C.); (L.W.)
| | - Lihua Wang
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai 200444, China; (X.X.); (H.C.); (L.W.)
| | - Shiping Song
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai 200444, China; (X.X.); (H.C.); (L.W.)
| |
Collapse
|
13
|
Sakib S, Bajaj K, Sen P, Li W, Gu J, Li Y, Soleymani L. Comparative Analysis of Machine Learning Algorithms Used for Translating Aptamer-Antigen Binding Kinetic Profiles to Diagnostic Decisions. ACS Sens 2025; 10:907-920. [PMID: 39869304 DOI: 10.1021/acssensors.4c02682] [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: 01/28/2025]
Abstract
Current approaches for classifying biosensor data in diagnostics rely on fixed decision thresholds based on receiver operating characteristic (ROC) curves, which can be limited in accuracy for complex and variable signals. To address these limitations, we developed a framework that facilitates the application of machine learning (ML) to diagnostic data for the binary classification of clinical samples, when using real-time electrochemical measurements. The framework was applied to a real-time multimeric aptamer assay (RT-MAp) that captures single-frequency (12.6 Hz) impedance data during the binding of viral protein targets to trimeric aptamers. The impedance data collected from 172 COVID-19 saliva samples were processed through multiple nonlinear regression models to extract nine key features from the transient signals. These features were then used to train three supervised ML algorithms─support vector machine (SVM), artificial neural network (ANN), and random forest (RF)─using a 75:25 training-testing ratio. Traditional ROC-based classification achieved an accuracy of 83.6%, while ML-based models significantly improved performance, with SVM, ANN, and RF achieving accuracies of 86.0%, 100%, and 100%, respectively. The ANN model demonstrated superior performance in handling complex and high-variance biosensor data, providing a robust and scalable solution for improving diagnostic accuracy in point-of-care settings.
Collapse
Affiliation(s)
- Sadman Sakib
- Department of Engineering Physics, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
| | - Kulmanak Bajaj
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
| | - Payel Sen
- Department of Engineering Physics, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
| | - Wantong Li
- Department of Engineering Physics, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
| | - Jimmy Gu
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
| | - Yingfu Li
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
- Micheal G. DeGroote for Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
| | - Leyla Soleymani
- Department of Engineering Physics, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
- Micheal G. DeGroote for Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, L8S 4L8 Hamilton, Ontario, Canada
| |
Collapse
|
14
|
Doan THP, Fried JP, Tang W, Hagness DE, Wu Y, Tilley RD, Gooding JJ. Optical Nanopore Blockade Sensors for Multiplexed Detection of Proteins. NANO LETTERS 2025; 25:3233-3239. [PMID: 39949081 DOI: 10.1021/acs.nanolett.4c05934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Enduring challenges for quantitative analysis in nanopore sensing are the detection of low biomarker concentrations in reasonable time frames and the detection of multiple biomarkers in the same sample. Herein we report an optical blockade nanopore sensor strategy that can detect more than a protein at femtomolar concentrations in rapid time (approximately 12 min). This is done using a nanopore array functionalized with an aptamer that can bind to two different but related target proteins. The assay then monitors two different colors of fluorescent particles modified with antibodies specific to the protein of interest, as they block the nanopores using a wide-field microscope. By distinguishing specific and nonspecific blockade events for each nanoparticle based on whether they can be easily pulled out of the nanopores using an electric field, we can simultaneously quantify the concentrations of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) down to femtomolar concentrations.
Collapse
Affiliation(s)
- Thanh Hoang Phuong Doan
- School of Chemistry, Australian Centre for NanoMedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Jasper P Fried
- School of Chemistry, Australian Centre for NanoMedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Wenxian Tang
- School of Chemistry, Australian Centre for NanoMedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Daniel Everett Hagness
- School of Chemistry, Australian Centre for NanoMedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Yanfang Wu
- School of Chemistry, Australian Centre for NanoMedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Richard D Tilley
- School of Chemistry, Australian Centre for NanoMedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
- Electron Microscope Unit, Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - J Justin Gooding
- School of Chemistry, Australian Centre for NanoMedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
15
|
Vo DK, Trinh KTL. Polymerase Chain Reaction Chips for Biomarker Discovery and Validation in Drug Development. MICROMACHINES 2025; 16:243. [PMID: 40141854 PMCID: PMC11944077 DOI: 10.3390/mi16030243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025]
Abstract
Polymerase chain reaction (PCR) chips are advanced, microfluidic platforms that have revolutionized biomarker discovery and validation because of their high sensitivity, specificity, and throughput levels. These chips miniaturize traditional PCR processes for the speed and precision of nucleic acid biomarker detection relevant to advancing drug development. Biomarkers, which are useful in helping to explain disease mechanisms, patient stratification, and therapeutic monitoring, are hard to identify and validate due to the complexity of biological systems and the limitations of traditional techniques. The challenges to which PCR chips respond include high-throughput capabilities coupled with real-time quantitative analysis, enabling researchers to identify novel biomarkers with greater accuracy and reproducibility. More recent design improvements of PCR chips have further expanded their functionality to also include digital and multiplex PCR technologies. Digital PCR chips are ideal for quantifying rare biomarkers, which is essential in oncology and infectious disease research. In contrast, multiplex PCR chips enable simultaneous analysis of multiple targets, therefore simplifying biomarker validation. Furthermore, single-cell PCR chips have made it possible to detect biomarkers at unprecedented resolution, hence revealing heterogeneity within cell populations. PCR chips are transforming drug development, enabling target identification, patient stratification, and therapeutic efficacy assessment. They play a major role in the development of companion diagnostics and, therefore, pave the way for personalized medicine, ensuring that the right patient receives the right treatment. While this tremendously promising technology has exhibited many challenges regarding its scalability, integration with other omics technologies, and conformity with regulatory requirements, many still prevail. Future breakthroughs in chip manufacturing, the integration of artificial intelligence, and multi-omics applications will further expand PCR chip capabilities. PCR chips will not only be important for the acceleration of drug discovery and development but also in raising the bar in improving patient outcomes and, hence, global health care as these technologies continue to mature.
Collapse
Affiliation(s)
- Dang-Khoa Vo
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea;
| | - Kieu The Loan Trinh
- Bionano Applications Research Center, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
16
|
Drainas AP, McIlwain DR, Dallas A, Chu T, Delgado-González A, Baron M, Angulo-Ibáñez M, Trejo A, Bai Y, Hickey JW, Lu G, Lu S, Pineda-Ramirez J, Anglin K, Richardson ET, Prostko JC, Frias E, Servellita V, Brazer N, Chiu CY, Peluso MJ, Martin JN, Wirz OF, Pham TD, Boyd SD, Kelly JD, Sage J, Nolan GP, Rovira-Clavé X. High-throughput multiplexed serology via the mass-spectrometric analysis of isotopically barcoded beads. Nat Biomed Eng 2025:10.1038/s41551-025-01349-0. [PMID: 39939547 DOI: 10.1038/s41551-025-01349-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 01/12/2025] [Indexed: 02/14/2025]
Abstract
In serology, each sample is typically tested individually, one antigen at a time. This is costly and time consuming. Serology techniques should ideally allow recurrent measurements in parallel in small sample volumes and be inexpensive and fast. Here we show that mass cytometry can be used to scale up multiplexed serology testing by leveraging polystyrene beads uniformly loaded with combinations of stable isotopes. We generated 18,480 unique isotopically barcoded beads to simultaneously detect, in a single tube with 924 serum samples, the levels of immunoglobulins G and M against 19 proteins from SARS-CoV-2 (a total of 36,960 tests in 400 nl of sample volume and 30 μl of reaction volume). As a rapid, high-throughput and cost-effective technique, serology by mass cytometry may contribute to the effective management of public health emergencies originating from infectious diseases.
Collapse
Affiliation(s)
- Alexandros P Drainas
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
| | - David R McIlwain
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, University of Nevada Reno, Reno, NV, USA
| | - Alec Dallas
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Theresa Chu
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Antonio Delgado-González
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Maya Baron
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Angelica Trejo
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Yunhao Bai
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - John W Hickey
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Guolan Lu
- Department of Pathology, Stanford University, Stanford, CA, USA
- Otolaryngology, Stanford University, Stanford, CA, USA
| | - Scott Lu
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Jesus Pineda-Ramirez
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Khamal Anglin
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Eugene T Richardson
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - John C Prostko
- Applied Research and Technology, Abbott Laboratories Inc., Abbott Park, IL, USA
| | - Edwin Frias
- Applied Research and Technology, Abbott Laboratories Inc., Abbott Park, IL, USA
| | - Venice Servellita
- Department of Laboratory Medicine, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Noah Brazer
- Department of Laboratory Medicine, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Charles Y Chiu
- Department of Laboratory Medicine, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Michael J Peluso
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jeffrey N Martin
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Oliver F Wirz
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Tho D Pham
- Department of Pathology, Stanford University, Stanford, CA, USA
- Stanford Blood Center, Palo Alto, CA, USA
| | - Scott D Boyd
- Department of Pathology, Stanford University, Stanford, CA, USA
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Stanford, CA, USA
| | - J Daniel Kelly
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
- Institute for Global Health Sciences, University of California San Francisco, San Francisco, CA, USA
- F.I. Proctor Foundation, University of California San Francisco, San Francisco, CA, USA
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Julien Sage
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA, USA.
| | - Xavier Rovira-Clavé
- Department of Pathology, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
- Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain.
| |
Collapse
|
17
|
He XL, Song YL, Ma LY, Jiang M, Xu L, Yu X. An ultra-sensitive fluorescence biosensor with rolling circle amplification and CRISPR/Cas12a one-pot system for FEN1 detection. Talanta 2025; 283:127159. [PMID: 39515055 DOI: 10.1016/j.talanta.2024.127159] [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: 08/02/2024] [Revised: 10/28/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Flap endonuclease 1 (FEN1), a structure-specific nuclease, has been reported to be widely involved in the development of cancer, and recognized as a new biomarker for cancer. However, there remains a deficiency in the availability of simple, rapid and reliable biosensors for its detection. We have constructed a cascade signal amplification fluorescence biosensor for ultra-sensitive and rapid detection of FEN1. This biosensor relied on FEN1-induced production of the 5' flap DNA, and combined rolling circle amplification (RCA) and CRISPR/Cas12a one-pot system (RCOS). By utilizing branched dsDNA substrates to provoke FEN1 activity, the 5' flap DNA was cleaved and isolated through magnetic separation. Subsequently, these DNA fragments initiated the RCA and CRISPR/Cas12a one-pot exponential amplification reaction, activating the cis and trans-cleavage activity of Cas12a and resulting in a significant fluorescence signal for readout. By combining RCA and CRISPR/Cas12a one-pot cascade signal amplification, the detection signal was remarkable enhanced. The RCOS exhibited excellent sensitivity with a limit of detection (LOD) of 4.1 × 10-7 U/μL, which was more sensitive and expeditious than many other approaches. Furthermore, the biosensor successfully facilitated accurate determination of FEN1 in cell extracts and plasma samples, revealing the potential clinical application and providing a dependable and rapid approach for FEN1 inhibitor screening. Compared with traditional methods, this approach has several benefits including improving the selectivity and sensitivity for FEN1 assay, reducing the complex operation process, and providing a method for the FEN1 inhibition screening.
Collapse
Affiliation(s)
- Xiang-Lan He
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yong-Li Song
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Yun Ma
- Department of Pharmacy, Union Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Xu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xu Yu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
18
|
Qian J, Xia J, Chiang S, Liu JF, Li K, Li F, Wei F, Aziz M, Kim Y, Go V, Morizio J, Zhong R, He Y, Yang K, Yang OO, Wong DTW, Lee LP, Huang TJ. Rapid and comprehensive detection of viral antibodies and nucleic acids via an acoustofluidic integrated molecular diagnostics chip: AIMDx. SCIENCE ADVANCES 2025; 11:eadt5464. [PMID: 39813350 PMCID: PMC11734728 DOI: 10.1126/sciadv.adt5464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
Precise and rapid disease detection is critical for controlling infectious diseases like COVID-19. Current technologies struggle to simultaneously identify viral RNAs and host immune antibodies due to limited integration of sample preparation and detection. Here, we present acoustofluidic integrated molecular diagnostics (AIMDx) on a chip, a platform enabling high-speed, sensitive detection of viral immunoglobulins [immunoglobulin A (IgA), IgG, and IgM] and nucleic acids. AIMDx uses acoustic vortexes and Gor'kov potential wells at a 1/10,000 subwavelength scale for concurrent isolation of viruses and antibodies while excluding cells, bacteria, and large (>200 nanometers) vesicles from saliva samples. The chip facilitates on-chip viral RNA enrichment, lysis in 2 minutes, and detection via transcription loop-mediated isothermal amplification, alongside electrochemical sensing of antibodies, including mucin-masked IgA. AIMDx achieved nearly 100% recovery of viruses and antibodies, a 32-fold RNA detection improvement, and an immunity marker sensitivity of 15.6 picograms per milliliter. This breakthrough provides a transformative tool for multiplex diagnostics, enhancing early infectious disease detection.
Collapse
Affiliation(s)
- Jiao Qian
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Jianping Xia
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Samantha Chiang
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jessica F. Liu
- Department of Anesthesiology, Duke University, Durham, NC 27710, USA
| | - Ke Li
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Feng Li
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Fang Wei
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Mohammad Aziz
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yong Kim
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Vinson Go
- Department of Electrical and Computer Engineering, Duke University, Durham, NC 27710, USA
| | - James Morizio
- Department of Electrical and Computer Engineering, Duke University, Durham, NC 27710, USA
| | - Ruoyu Zhong
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Ye He
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Kaichun Yang
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Otto O. Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David T. W. Wong
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Luke P. Lee
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea
| | - Tony Jun Huang
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| |
Collapse
|
19
|
Mohammadi A, Chiang S, Li F, Wei F, Lau CS, Aziz M, Ibarrondo FJ, Fulcher JA, Yang OO, Chia D, Kim Y, Wong DTW. Direct detection of 4-dimensions of SARS-CoV-2: infection (vRNA), infectivity (antigen), binding antibody, and functional neutralizing antibody in saliva. Sci Rep 2024; 14:30792. [PMID: 39730575 DOI: 10.1038/s41598-024-81019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 11/22/2024] [Indexed: 12/29/2024] Open
Abstract
We developed a 4-parameter clinical assay using Electric Field Induced Release and Measurement (EFIRM) technology to simultaneously assess SARS-CoV-2 RNA (vRNA), nucleocapsid antigen, host binding (BAb) and neutralizing antibody (NAb) levels from a drop of saliva with performance that equals or surpasses current EUA-approved tests. The vRNA and antigen assays achieved lower limit of detection (LOD) of 100 copies/reaction and 3.5 TCID₅₀/mL, respectively. The vRNA assay differentiated between acutely infected (n = 10) and infection-naïve patients (n = 33) with an AUC of 0.9818, sensitivity of 90%, and specificity of 100%. The antigen assay similarly differentiated these patient populations with an AUC of 1.000. The BAb assay detected BAbs with an LOD of 39 pg/mL and distinguished acutely infected (n = 35), vaccinated with prior infection (n = 13), and vaccinated infection-naïve patients (n = 13) from pre-pandemic (n = 81) with AUC of 0.9481, 1.000, and 0.9962, respectively. The NAb assay detected NAbs with a LOD of 31.6 Unit/mL and differentiated between COVID-19 recovered or vaccinated patients (n = 31) and pre-pandemic controls (n = 60) with an AUC 0.923, sensitivity of 87.10%, and specificity of 86.67%. Our combo assay represents a significant technological advancement to simultaneously address SARS-CoV-2 infection and immunity, and it lays the foundation for tackling potential future pandemics.
Collapse
Grants
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
Collapse
Affiliation(s)
- Aida Mohammadi
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Samantha Chiang
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Feng Li
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Fang Wei
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | | | - Mohammad Aziz
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Francisco J Ibarrondo
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer A Fulcher
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Otto O Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - David Chia
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Yong Kim
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA.
| | - David T W Wong
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA.
| |
Collapse
|
20
|
Chen Z, Mao K, Chen Z, Feng R, Du W, Zhang H, Tu C. Isothermal nucleic acid amplification for monitoring hand-foot-and-mouth disease: current status and future implications. Mikrochim Acta 2024; 192:31. [PMID: 39720958 DOI: 10.1007/s00604-024-06899-9] [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: 09/27/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024]
Abstract
With the global prevalence of the hand-foot-and-mouth disease (HFMD) epidemic, the development of reliable point-of-care testing (POCT) is crucial for the timely identification and prevention of outbreaks. Isothermal nucleic acid amplification techniques (INAATs) have attracted much attention because of their high efficiency for rapid diagnosis. In this work, we systematically summarize the current status of INAATs for HFMD and discuss advantages and drawbacks of various INAATs for HFMD. The INAATs for HFMD detection mainly include loop-mediated isothermal amplification (LAMP), simultaneous amplification and testing (SAT), and recombinase polymerase amplification (RPA). Among them, LAMP has excelled in several diagnostic metrics and has made significant progress in the field of POCT. SAT has been effective in overcoming the problem of RNA degradation. RPA is suited for on-site testing due to its rapid amplification rate and low reaction temperature. In addition, this study explores the potential of INAATs in lateral flow strips (LFS) test and microfluidic devices for HFMD. LFS is typically used for qualitative analysis and supports multiple detection. Microfluidics can integrate necessary processes of sample pre-processing, amplification, and signal output, enabling high-throughput qualitative or quantitative detection and demonstrating the potential of monitoring HFMD. We hope the current work will provide insights into INAATs for monitoring HFMD and serve as a reference for the implementation of on-site EV detection for public health.
Collapse
Affiliation(s)
- Zhen Chen
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 561113, China
- State Key Laboratory of Environmental Geochemistry, Institute of Geochemistry, Chinese Academy of Sciences, Guiyang, 550081, China
| | - Kang Mao
- State Key Laboratory of Environmental Geochemistry, Institute of Geochemistry, Chinese Academy of Sciences, Guiyang, 550081, China.
| | - Zhuo Chen
- State Key Laboratory of Environmental Geochemistry, Institute of Geochemistry, Chinese Academy of Sciences, Guiyang, 550081, China
| | - Rida Feng
- State Key Laboratory of Environmental Geochemistry, Institute of Geochemistry, Chinese Academy of Sciences, Guiyang, 550081, China
| | - Wei Du
- Yunnan Provincial Key Laboratory of Soil Carbon Sequestration and Pollution Control, Faculty of Environmental Science & Engineering, Kunming University of Science & Technology, Kunming, 650500, China
| | - Hua Zhang
- State Key Laboratory of Environmental Geochemistry, Institute of Geochemistry, Chinese Academy of Sciences, Guiyang, 550081, China
| | - Chenglong Tu
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 561113, China.
- Toxicity Testing Center, Guizhou Medical University, Guian New Region, 561113, China.
| |
Collapse
|
21
|
Liu KZ, Tian G, Ko ACT, Geissler M, Malic L, Moon BU, Clime L, Veres T. Microfluidic methods for the diagnosis of acute respiratory tract infections. Analyst 2024; 150:9-33. [PMID: 39440426 DOI: 10.1039/d4an00957f] [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: 10/25/2024]
Abstract
Acute respiratory tract infections (ARTIs) are caused by sporadic or pandemic outbreaks of viral or bacterial pathogens, and continue to be a considerable socioeconomic burden for both developing and industrialized countries alike. Diagnostic methods and technologies serving as the cornerstone for disease management, epidemiological tracking, and public health interventions are evolving continuously to keep up with the demand for higher sensitivity, specificity and analytical throughput. Microfluidics is becoming a key technology in these developments as it allows for integrating, miniaturizing and automating bioanalytical assays at an unprecedented scale, reducing sample and reagent consumption and improving diagnostic performance in terms of sensitivity, throughput and response time. In this article, we describe relevant ARTIs-pneumonia, influenza, severe acute respiratory syndrome, and coronavirus disease 2019-along with their pathogenesis. We provide a summary of established methods for disease diagnosis, involving nucleic acid amplification techniques, antigen detection, serological testing as well as microbial culture. This is followed by a short introduction to microfluidics and how flow is governed at low volume and reduced scale using centrifugation, pneumatic pumping, electrowetting, capillary action, and propagation in porous media through wicking, for each of these principles impacts the design, functioning and performance of diagnostic tools in a particular way. We briefly cover commercial instruments that employ microfluidics for use in both laboratory and point-of-care settings. The main part of the article is dedicated to emerging methods deriving from the use of miniaturized, microfluidic systems for ARTI diagnosis. Finally, we share our thoughts on future perspectives and the challenges associated with validation, approval, and adaptation of microfluidic-based systems.
Collapse
Affiliation(s)
- Kan-Zhi Liu
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 435 Ellice Avenue, Winnipeg, MB, R3B 1Y6, Canada
| | - Ganghong Tian
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 435 Ellice Avenue, Winnipeg, MB, R3B 1Y6, Canada
| | - Alex C-T Ko
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 435 Ellice Avenue, Winnipeg, MB, R3B 1Y6, Canada
| | - Matthias Geissler
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, QC, J4B 6Y4, Canada.
| | - Lidija Malic
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, QC, J4B 6Y4, Canada.
| | - Byeong-Ui Moon
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, QC, J4B 6Y4, Canada.
| | - Liviu Clime
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, QC, J4B 6Y4, Canada.
| | - Teodor Veres
- Life Sciences Division, Medical Devices Research Centre, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, QC, J4B 6Y4, Canada.
| |
Collapse
|
22
|
Chen Y, Wang P, Zhang FN, Dai H, Jiao XY, Wang XY, Yu QW, Kang M, Su S, Wang D. Sensors for surveillance of RNA viruses: a One Health perspective. THE LANCET. MICROBE 2024:101029. [PMID: 39681124 DOI: 10.1016/j.lanmic.2024.101029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/03/2024] [Accepted: 10/17/2024] [Indexed: 12/18/2024]
Abstract
RNA viruses, especially those capable of cross-species transmission, pose a serious threat to human, animal, and environmental health, as exemplified by the 2024 outbreak of the highly pathogenic avian influenza H5N1 virus in cattle, unpasteurised milk, and workers on dairy farms in the USA. This escalating risk of a new RNA virus pandemic highlights the urgent need to implement One Health strategies. However, the centralised virus detection systems currently in use fall short of meeting the required level of virus surveillance and infection diagnosis, particularly in resource-limited regions. In this context, the latest advancements in RNA virus-sensing technologies offer promising solutions. Through interdisciplinary collaboration, these sensors can achieve sensitivity and reliability similar to that of standard laboratory equipment and offer several advantages, such as compact size, affordability, and operational simplicity. In this Review, we highlight the latest advances in sensing technologies for detecting different biomarkers of viral infections (RNA, antigens, and antibodies). We further compare the sensing principles and performances of these technologies and discuss the possibility of deployment of these sensors in the One Health approach and the challenges expected in this pursuit. In conclusion, the widespread use of RNA virus sensors is expected to enhance the effectiveness of surveillance systems for infectious diseases.
Collapse
Affiliation(s)
- Ye Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China.
| | - Peng Wang
- Department of Atmospheric and Oceanic Sciences, Fudan University, Shanghai, China
| | - Fen-Ni Zhang
- Department of Biomedical Engineering, Zhejiang University, Hangzhou, China
| | - Hui Dai
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xin-Yi Jiao
- Department of Epidemiology, School of Public Health, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Xin-Yu Wang
- Department of Epidemiology, School of Public Health, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Qi-Wen Yu
- Research Center for Frontier Fundamental Studies, Zhejiang Lab, Hangzhou, China
| | - Mei Kang
- Department of Epidemiology, School of Public Health, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China; Clinical Research Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Su
- Department of Epidemiology, School of Public Health, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| | - Di Wang
- Department of Biomedical Engineering, Zhejiang University, Hangzhou, China; Research Center for Frontier Fundamental Studies, Zhejiang Lab, Hangzhou, China.
| |
Collapse
|
23
|
Liu Z, Zhou Y, Lu J, Gong T, Ibáñez E, Cifuentes A, Lu W. Microfluidic biosensors for biomarker detection in body fluids: a key approach for early cancer diagnosis. Biomark Res 2024; 12:153. [PMID: 39639411 PMCID: PMC11622463 DOI: 10.1186/s40364-024-00697-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024] Open
Abstract
Early detection of cancer significantly improves patient outcomes, with biomarkers offering a promising avenue for earlier and more precise diagnoses. Microfluidic biosensors have emerged as a powerful tool for detecting these biomarkers in body fluids, providing enhanced sensitivity, specificity, and rapid analysis. This review focuses on recent advances in microfluidic biosensors from 2018 to 2024, detailing their operational principles, fabrication techniques, and integration with nanotechnology for cancer biomarker detection. Additionally, we have reviewed recent innovations in several aspects of microfluidic biosensors, such as novel detection technologies, nanomaterials and novel microfluidic chip structures, which significantly enhance detection capabilities. We highlight key biomarkers pertinent to early cancer detection and explore how these innovations in biosensor technology contribute to the evolving landscape of personalized medicine. We further explore how these technologies could be incorporated into clinical cancer diagnostic workflows to improve early detection and treatment outcomes. These innovations could help enable more precise and personalized cancer diagnostics. In addition, this review addresses several important issues such as enhancing the scalability and sensitivity of these biosensors in clinical settings and points out future possibilities of combining artificial intelligence diagnostics with microfluidic biosensors to optimize their practical applications. This overview aims to guide future research and clinical applications by addressing current challenges and identifying opportunities for further development in the field of biomarker research.
Collapse
Affiliation(s)
- Zhiting Liu
- School of Medicine and Health, Harbin Institute of Technology, 92 Xidazhi Street, Nangang District, Harbin, 150001, China
- National and Local Joint Engineering Laboratory for Synthesis Transformation and Separation of Extreme Environmental Nutrients, 92 Xidazhi Street, Nangang District, Harbin, 150001, China
| | - Yingyu Zhou
- School of Medicine and Health, Harbin Institute of Technology, 92 Xidazhi Street, Nangang District, Harbin, 150001, China.
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, Henan, China.
- National and Local Joint Engineering Laboratory for Synthesis Transformation and Separation of Extreme Environmental Nutrients, 92 Xidazhi Street, Nangang District, Harbin, 150001, China.
| | - Jia Lu
- School of Mechatronics Engineering, Harbin Institute of Technology, 92 Xidazhi Street, Nangang District, Harbin, 150001, China.
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, Henan, China.
| | - Ting Gong
- School of Medicine and Health, Harbin Institute of Technology, 92 Xidazhi Street, Nangang District, Harbin, 150001, China
- National and Local Joint Engineering Laboratory for Synthesis Transformation and Separation of Extreme Environmental Nutrients, 92 Xidazhi Street, Nangang District, Harbin, 150001, China
| | - Elena Ibáñez
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolás Cabrera 9, Madrid, 28049, Spain
| | - Alejandro Cifuentes
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolás Cabrera 9, Madrid, 28049, Spain
| | - Weihong Lu
- School of Medicine and Health, Harbin Institute of Technology, 92 Xidazhi Street, Nangang District, Harbin, 150001, China.
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, Henan, China.
- National and Local Joint Engineering Laboratory for Synthesis Transformation and Separation of Extreme Environmental Nutrients, 92 Xidazhi Street, Nangang District, Harbin, 150001, China.
| |
Collapse
|
24
|
Mao Z, Lei H, Chen R, Ren S, Liu B, Gao Z. CRISPR/Cas13a analysis based on NASBA amplification for norovirus detection. Talanta 2024; 280:126725. [PMID: 39167939 DOI: 10.1016/j.talanta.2024.126725] [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: 02/20/2024] [Revised: 05/22/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Human norovirus (HuNoV) is a leading cause of foodborne diseases worldwide, making rapid and accurate detection crucial for prevention and control. In recent years, the CRISPR/Cas13a system, known for its single-base resolution in RNA recognition and unique collateral cleavage activity, is particularly suitable for sensitive and rapid RNA detection. However, isothermal amplification-based CRISPR/Cas13 assays often require an external transcription step, complicating the detection process. In our study, an efficient diagnostic technique based on the NASBA/Cas13a system was established to identify conserved regions at the ORF1-ORF2 junction of norovirus. The RNA amplification techniques [Nucleic Acid Sequence-Based Amplification (NASBA)] integrates reverse transcription and transcription steps, enabling sensitive, accurate, and rapid enrichment of low-abundance RNA. Furthermore, the CRISPR/Cas13a system provides secondary precise recognition of the amplified products, generating a fluorescence signal through its activated accessory collateral cleavage activity. We optimized the reaction kinetics parameters of Cas13a and achieved a detection limit as low as 51pM. The conditions for the cascade reaction involving CRISPR analysis and RNA amplification were optimized. Finally, we validated the reliability and accuracy of the NASBA/Cas13a method by detecting norovirus in shellfish, achieving results comparable to qRT-PCR in a shorter time and detecting viral loads as low as 10 copies/μL.
Collapse
Affiliation(s)
- Zefeng Mao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Huang Lei
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Ruipeng Chen
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Shuyue Ren
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China.
| | - Baolin Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China.
| | - Zhixian Gao
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China.
| |
Collapse
|
25
|
Freko S, Nikić M, Mayer D, Weiß LJK, Simmel FC, Wolfrum B. Digital CRISPR-Powered Biosensor Concept without Target Amplification Using Single-Impact Electrochemistry. ACS Sens 2024; 9:6197-6206. [PMID: 39435883 PMCID: PMC11590096 DOI: 10.1021/acssensors.4c02060] [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: 08/07/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024]
Abstract
The rapid and reliable detection and quantification of nucleic acids is crucial for various applications, including infectious disease and cancer diagnostics. While conventional methods, such as the quantitative polymerase chain reaction are widely used, they are limited to the laboratory environment due to their complexity and the requirement for sophisticated equipment. In this study, we present a novel amplification-free digital sensing strategy by combining the collateral cleavage activity of the Cas12a enzyme with single-impact electrochemistry. In doing so, we modified silver nanoparticles using a straightforward temperature-assisted cofunctionalization process to subsequently detect the collision events of particles released by the activated Cas12a as distinct current spikes on a microelectrode array. The functionalization resulted in stable DNA-AgNP conjugates, making them suitable for numerous biosensor applications. Thus, our study demonstrates the potential of clustered regularly interspaced short palindromic repeats-based diagnostics combined with impact-based digital sensing for a rapid and amplification-free quantification of nucleic acids.
Collapse
Affiliation(s)
- Sebastian Freko
- Neuroelectronics,
Munich Institute of Biomedical Engineering, Department of Electrical
Engineering, School of Computation, Information and Technology, Technical University of Munich, 85748 Garching, Germany
| | - Marta Nikić
- Neuroelectronics,
Munich Institute of Biomedical Engineering, Department of Electrical
Engineering, School of Computation, Information and Technology, Technical University of Munich, 85748 Garching, Germany
| | - Dirk Mayer
- Institute
of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Lennart J. K. Weiß
- Department
of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Friedrich C. Simmel
- Department
of Bioscience, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Bernhard Wolfrum
- Neuroelectronics,
Munich Institute of Biomedical Engineering, Department of Electrical
Engineering, School of Computation, Information and Technology, Technical University of Munich, 85748 Garching, Germany
| |
Collapse
|
26
|
Guo W, Tao Y, Yang R, Mao K, Zhou H, Xu M, Sun T, Li X, Shi C, Ge Z, Xue R, Zhou H, Ren Y. Compact highly sensitive photothermal RT-LAMP chip for simultaneous multidisease detection. SCIENCE ADVANCES 2024; 10:eadq2899. [PMID: 39536102 PMCID: PMC11559619 DOI: 10.1126/sciadv.adq2899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Developing instant detection systems with disease diagnostic capabilities holds immense importance for remote or resource-limited areas. However, the task of creating these systems-which are simultaneously easy to operate, rapid in detection, and cost-effective-remains a challenge. In this study, we present a compact highly sensitive photothermal reverse transcriptase-loop-mediated isothermal amplification (RT-LAMP) chip (SPRC) designed for the detection of multiple diseases. The nucleic acid (NA) amplification on the chip is achieved through LAMP driven by either LED illumination or simple sunlight focusing. SPRC performs sample addition and amplification within a limited volume and autonomous enrichment of NA during the sample addition process, achieving a limit of detection (LOD) as low as 0.2 copies per microliter. Through 120 clinical samples, we achieved an accuracy of 95%, with a specificity exceeding 97.5%. Overall, SPRC has achieved promising progress in the application of point-of-care testing (POCT) by using light energy to simultaneously detect multiple diseases.
Collapse
Affiliation(s)
- Wenshang Guo
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin 150001, China
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Ye Tao
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Ruizhe Yang
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin 150001, China
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Kaihao Mao
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Hongwei Zhou
- Department of Laboratory Diagnosis, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Minghui Xu
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Tie Sun
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin 150001, China
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Xiao Li
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin 150001, China
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Changrui Shi
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin 150001, China
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Zhenyou Ge
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin 150001, China
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Rui Xue
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Haizhou Zhou
- Department of Laboratory Diagnosis, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yukun Ren
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin 150001, China
- School of Mechatronics Engineering, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
27
|
Mirjalili S, Ikbal MA, Hou CW, Mohammadi MK, Choi Y, Kelbauskas L, VanBlargan LA, Hogue BG, Murugan V, Diamond MS, Wang C. Nanoparticle-Supported, Rapid, Digital Quantification of Neutralizing Antibodies Against SARS-CoV-2 Variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.05.622148. [PMID: 39574622 PMCID: PMC11580943 DOI: 10.1101/2024.11.05.622148] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
The measurement of neutralizing immune responses to viral infection is essential, given the heterogeneity of human immunity and the emergence of new virus strains. However, neutralizing antibody (nAb) assays often require high-level biosafety containment, sophisticated instrumentation, and long detection times. Here, as a proof-of-principle, we designed a nanoparticle-supported, rapid, electronic detection (NasRED) assay to assess the neutralizing potency of monoclonal antibodies (mAbs) against SARS-CoV-2. The gold nanoparticles (AuNPs) coated with human angiotensin-converting enzyme 2 (ACE2) protein as nAb potency reporters were mixed with the mAbs to be tested, as well as streptavidin-conjugated multivalent spike (S) protein or their receptor binding domains (RBD). High-affinity and ACE2-competitive nAbs alter the S (or RBD)-to-ACE2 binding level and modulate AuNP cluster formation and precipitation. The amount of free-floating AuNP reporters is quantified by a semiconductor-based readout system that measures the AuNPs' optical extinction, producing nAb signals that can differentiate SARS-CoV-2 variants (Wuhan-Hu-1, Gamma, and Omicron). The modular design nature, short assay time (less than 30 minutes), and portable and inexpensive readout system make this NasRED-nAb assay applicable to measuring vaccine potency, immune responses to infection, and the efficacy of antibody-based therapies.
Collapse
|
28
|
Cruz Da Silva E, Gaki P, Flieg F, Messmer M, Gucciardi F, Markovska Y, Reisch A, Fafi-Kremer S, Pfeffer S, Klymchenko AS. Direct Zeptomole Detection of RNA Biomarkers by Ultrabright Fluorescent Nanoparticles on Magnetic Beads. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404167. [PMID: 39011971 DOI: 10.1002/smll.202404167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/05/2024] [Indexed: 07/17/2024]
Abstract
Nucleic acids are important biomarkers in cancer and viral diseases. However, their ultralow concentration in biological/clinical samples makes direct target detection challenging, because it leads to slow hybridization kinetics with the probe and its insufficient signal-to-noise ratio. Therefore, RNA target detection is done by molecular (target) amplification, notably by RT-PCR, which is a tedious multistep method that includes nucleic acid extraction and reverse transcription. Here, a direct method based on ultrabright dye-loaded polymeric nanoparticles in a sandwich-like hybridization assay with magnetic beads is reported. The ultrabright DNA-functionalized nanoparticle, equivalent to ≈10 000 strongly emissive rhodamine dyes, is hybridized with the magnetic bead to the RNA target, providing the signal amplification for the detection. This concept (magneto-fluorescent sandwich) enables high-throughput detection of DNA and RNA sequences of varied lengths from 48 to 1362 nt with the limit of detection down to 0.3 fm using a plate reader (15 zeptomoles), among the best reported for optical sandwich assays. Moreover, it allows semi-quantitative detection of SARS-CoV-2 viral RNA directly in clinical samples without a dedicated RNA extraction step. The developed technology, combining ultrabright nanoparticles with magnetic beads, addresses fundamental challenges in RNA detection; it is expected to accelerate molecular diagnostics of diseases.
Collapse
Affiliation(s)
- Elisabete Cruz Da Silva
- Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, UMR 7021 CNRS, Illkirch, 67401, France
- BrightSens Diagnostics SAS, 11 Rue de l'Académie, Strasbourg, 67000, France
| | - Paraskevi Gaki
- Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, UMR 7021 CNRS, Illkirch, 67401, France
- BrightSens Diagnostics SAS, 11 Rue de l'Académie, Strasbourg, 67000, France
| | - Fabien Flieg
- BrightSens Diagnostics SAS, 11 Rue de l'Académie, Strasbourg, 67000, France
| | - Melanie Messmer
- Architecture et Réactivité de l'ARN, Institut de biologie moléculaire et cellulaire du CNRS, Université de Strasbourg, UPR 9002, Strasbourg, 67084, France
| | - Floriane Gucciardi
- Architecture et Réactivité de l'ARN, Institut de biologie moléculaire et cellulaire du CNRS, Université de Strasbourg, UPR 9002, Strasbourg, 67084, France
| | | | - Andreas Reisch
- Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, UMR 7021 CNRS, Illkirch, 67401, France
| | - Samira Fafi-Kremer
- CHU de Strasbourg, Laboratoire de Virologie, Université de Strasbourg, INSERM, Strasbourg, IRM UMR-S 1109, France
| | - Sébastien Pfeffer
- Architecture et Réactivité de l'ARN, Institut de biologie moléculaire et cellulaire du CNRS, Université de Strasbourg, UPR 9002, Strasbourg, 67084, France
| | - Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, UMR 7021 CNRS, Illkirch, 67401, France
| |
Collapse
|
29
|
Jiang Z, Ye D, Xiang L, He Z, Dai X, Yang J, Xiong Q, Ma Y, Zhi D, Zou Y, Peng Q, Wang S, Li J, Zhang F, Di CA. A drug-mediated organic electrochemical transistor for robustly reusable biosensors. NATURE MATERIALS 2024; 23:1547-1555. [PMID: 39112738 DOI: 10.1038/s41563-024-01970-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/08/2024] [Indexed: 09/15/2024]
Abstract
Reusable point-of-care biosensors offer a cost-effective solution for serial biomarker monitoring, addressing the critical demand for tumour treatments and recurrence diagnosis. However, their realization has been limited by the contradictory requirements of robust reusability and high sensing capability to multiple interactions among transducer surface, sensing probes and target analytes. Here we propose a drug-mediated organic electrochemical transistor as a robust, reusable epidermal growth factor receptor sensor with striking sensitivity and selectivity. By electrostatically adsorbing protonated gefitinib onto poly(3,4-ethylenedioxythiophene):polystyrene sulfonate and leveraging its strong binding to the epidermal growth factor receptor target, the device operates with a unique refresh-in-sensing mechanism. It not only yields an ultralow limit-of-detection concentration down to 5.74 fg ml-1 for epidermal growth factor receptor but, more importantly, also produces an unprecedented regeneration cycle exceeding 200. We further validate the potential of our devices for easy-to-use biomedical applications by creating an 8 × 12 diagnostic drug-mediated organic electrochemical transistor array with excellent uniformity to clinical blood samples.
Collapse
Affiliation(s)
- Ziling Jiang
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
- Beijing National Laboratory for Molecular Sciences, CAS Kay Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Dekai Ye
- Beijing National Laboratory for Molecular Sciences, CAS Kay Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- Zhangjiang Laboratory, Shanghai, China
| | - Lanyi Xiang
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zihan He
- Beijing National Laboratory for Molecular Sciences, CAS Kay Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Xiaojuan Dai
- Beijing National Laboratory for Molecular Sciences, CAS Kay Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Junfang Yang
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qi Xiong
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Yingqiao Ma
- Beijing National Laboratory for Molecular Sciences, CAS Kay Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Danfeng Zhi
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ye Zou
- Beijing National Laboratory for Molecular Sciences, CAS Kay Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Qian Peng
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences, CAS Kay Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Jia Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China.
| | - Fengjiao Zhang
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China.
| | - Chong-An Di
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China.
- Beijing National Laboratory for Molecular Sciences, CAS Kay Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
30
|
Shen S, Wang W, Ma Y, Wang S, Zhang S, Cai X, Chen L, Zhang J, Li Y, Wu X, Wei J, Zhao Y, Huang A, Niu S, Wang D. Affinity molecular assay for detecting Candida albicans using chitin affinity and RPA-CRISPR/Cas12a. Nat Commun 2024; 15:9304. [PMID: 39468064 PMCID: PMC11519397 DOI: 10.1038/s41467-024-53693-5] [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: 02/21/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024] Open
Abstract
Invasive fungal infections (IFIs) pose a significant threat to immunocompromised individuals, leading to considerable morbidity and mortality. Prompt and accurate diagnosis is essential for effective treatment. Here we develop a rapid molecular diagnostic method that involves three steps: fungal enrichment using affinity-magnetic separation (AMS), genomic DNA extraction with silicon hydroxyl magnetic beads, and detection through a one-pot system. This method, optimized to detect 30 CFU/mL of C. albicans in blood and bronchoalveolar lavage (BAL) samples within 2.5 h, is approximately 100 times more sensitive than microscopy-based staining. Initial validation using clinical samples showed 93.93% sensitivity, 100% specificity, and high predictive values, while simulated tests demonstrated 95% sensitivity and 100% specificity. This cost-effective, highly sensitive technique offers potential for use in resource-limited clinical settings and can be easily adapted to differentiate between fungal species and detect drug resistance.
Collapse
Affiliation(s)
- Shimei Shen
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Clinical Laboratory Medicine, Chongqing Red Cross Hospital (Jiangbei District People's Hospital), Chongqing, China
| | - Wen Wang
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chengdu Medical College (Nuclear Industry 416 Hospital), Chengdu, China
| | - Yuanyan Ma
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shilei Wang
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Shaocheng Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chengdu Medical College (Nuclear Industry 416 Hospital), Chengdu, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Xuefei Cai
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Liang Chen
- Department of Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Jin Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yalan Li
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xiaoli Wu
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jie Wei
- Department of Clinical Laboratory, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Yanan Zhao
- Department of Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Ailong Huang
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Siqiang Niu
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Deqiang Wang
- Key Laboratory of Clinical Laboratory Diagnostics (Chinese Ministry of Education), Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
- Western (Chongqing) Collaborative Innovation Center for Intelligent Diagnostics and Digital Medicine, Chongqing National Biomedicine Industry Park, Chongqing, China.
| |
Collapse
|
31
|
Mao T, Nan L, Shum HC. Digital Quantification and Ultrasensitive Detection of Single Influenza Virus Using Microgel-in-Droplet Enzyme-Linked Immunosorbent Assay. Anal Chem 2024; 96:16134-16144. [PMID: 39360754 DOI: 10.1021/acs.analchem.4c02429] [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: 10/16/2024]
Abstract
Detection and quantification of viral particles (VPs) facilitate both diagnostics of pathogenic viruses and quality control testing of virus-based products. However, existing technologies fail to afford concurrent ultrasensitive detection and large-scale absolute quantification of VPs. Here, we propose a digital Microgel-in-Droplet enzyme-linked immunosorbent assay (ELISA) system that enables the processing and monitoring of millions of ELISA reactions at the single-VP level by incorporating droplet microfluidics with sandwich ELISA. Upon validating the microfluidic workflow and optimizing ELISA parameters, we demonstrate ultrasensitive VP detection at a limit of detection of 56 PFU/test. Leveraging a fluorescence-based screening platform, we further realize high-throughput digital counting of VPs with a linear detection range of 500-64 000 PFU/test. The precision is comparable to that of the gold standard, the plaque assay, across a wide range of virus concentrations. We anticipate that our system will provide a novel paradigm for the absolute enumeration of various types of viral particles.
Collapse
Affiliation(s)
- Tianjiao Mao
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Lang Nan
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong, China
| | - Ho Cheung Shum
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong, China
| |
Collapse
|
32
|
Qian X, Xu Q, Lyon CJ, Hu TY. CRISPR for companion diagnostics in low-resource settings. LAB ON A CHIP 2024; 24:4717-4740. [PMID: 39268697 PMCID: PMC11393808 DOI: 10.1039/d4lc00340c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 08/15/2024] [Indexed: 09/17/2024]
Abstract
New point-of-care tests (POCTs), which are especially useful in low-resource settings, are needed to expand screening capacity for diseases that cause significant mortality: tuberculosis, multiple cancers, and emerging infectious diseases. Recently, clustered regularly interspaced short palindromic repeats (CRISPR)-based diagnostic (CRISPR-Dx) assays have emerged as powerful and versatile alternatives to traditional nucleic acid tests, revealing a strong potential to meet this need for new POCTs. In this review, we discuss CRISPR-Dx assay techniques that have been or could be applied to develop POCTs, including techniques for sample processing, target amplification, multiplex assay design, and signal readout. This review also describes current and potential applications for POCTs in disease diagnosis and includes future opportunities and challenges for such tests. These tests need to advance beyond initial assay development efforts to broadly meet criteria for use in low-resource settings.
Collapse
Affiliation(s)
- Xu Qian
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China.
| | - Qiang Xu
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China.
| | - Christopher J Lyon
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA.
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | - Tony Y Hu
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA.
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| |
Collapse
|
33
|
Son H. Harnessing CRISPR/Cas Systems for DNA and RNA Detection: Principles, Techniques, and Challenges. BIOSENSORS 2024; 14:460. [PMID: 39451674 PMCID: PMC11506544 DOI: 10.3390/bios14100460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024]
Abstract
The emergence of CRISPR/Cas systems has revolutionized the field of molecular diagnostics with their high specificity and sensitivity. This review provides a comprehensive overview of the principles and recent advancements in harnessing CRISPR/Cas systems for detecting DNA and RNA. Beginning with an exploration of the molecular mechanisms of key Cas proteins underpinning CRISPR/Cas systems, the review navigates the detection of both pathogenic and non-pathogenic nucleic acids, emphasizing the pivotal role of CRISPR in identifying diverse genetic materials. The discussion extends to the integration of CRISPR/Cas systems with various signal-readout techniques, including fluorescence, electrochemical, and colorimetric, as well as imaging and biosensing methods, highlighting their advantages and limitations in practical applications. Furthermore, a critical analysis of challenges in the field, such as target amplification, multiplexing, and quantitative detection, underscores areas requiring further refinement. Finally, the review concludes with insights into the future directions of CRISPR-based nucleic acid detection, emphasizing the potential of these systems to continue driving innovation in diagnostics, with broad implications for research, clinical practice, and biotechnology.
Collapse
Affiliation(s)
- Heyjin Son
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| |
Collapse
|
34
|
Yuan S, Yuan H, Hay DC, Hu H, Wang C. Revolutionizing Drug Discovery: The Impact of Distinct Designs and Biosensor Integration in Microfluidics-Based Organ-on-a-Chip Technology. BIOSENSORS 2024; 14:425. [PMID: 39329800 PMCID: PMC11430660 DOI: 10.3390/bios14090425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
Traditional drug development is a long and expensive process with high rates of failure. This has prompted the pharmaceutical industry to seek more efficient drug development frameworks, driving the emergence of organ-on-a-chip (OOC) based on microfluidic technologies. Unlike traditional animal experiments, OOC systems provide a more accurate simulation of human organ microenvironments and physiological responses, therefore offering a cost-effective and efficient platform for biomedical research, particularly in the development of new medicines. Additionally, OOC systems enable quick and real-time analysis, high-throughput experimentation, and automation. These advantages have shown significant promise in enhancing the drug development process. The success of an OOC system hinges on the integration of specific designs, manufacturing techniques, and biosensors to meet the need for integrated multiparameter datasets. This review focuses on the manufacturing, design, sensing systems, and applications of OOC systems, highlighting their design and sensing capabilities, as well as the technical challenges they currently face.
Collapse
Affiliation(s)
- Sheng Yuan
- Centre of Biomedical Systems and Informatics, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), School of Medicine, International Campus, Zhejiang University, Haining 314400, China
| | - Huipu Yuan
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - David C. Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK;
| | - Huan Hu
- Zhejiang University-University of Illinois Urbana-Champaign Institute (ZJU-UIUC Institute), International Campus, Zhejiang University, Haining 314400, China
| | - Chaochen Wang
- Centre of Biomedical Systems and Informatics, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), School of Medicine, International Campus, Zhejiang University, Haining 314400, China
- Department of Gynecology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| |
Collapse
|
35
|
Lu J, Han Y, Wu Y, Wang K, Yang J, Miao P, Li G. Simplified Electrochemical Approach for End-Point Yet Quantitative Detection of Nucleic Acids in Resource-Limited Settings. ACS Sens 2024; 9:4098-4106. [PMID: 39033535 DOI: 10.1021/acssensors.4c01025] [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: 07/23/2024]
Abstract
Nucleic acid detection plays a crucial role in various aspects of health care, necessitating accessible and reliable quantification methods, especially in resource-limited settings. This work presents a simplified electrochemical approach for end-point yet quantitative nucleic acid detection. By elevating the concentration of redox species and choosing potential as the signals, we achieved enhanced signal robustness, even in the presence of interfering substances. Leveraging this robustness, we accurately measured pH-induced redox potential changes in methylene blue solution for end-point nucleic acid detection after loop-mediated isothermal amplification (LAMP). Our method demonstrated quantitative detection of the SARS-CoV-2 N gene and human ATCB gene and successful discrimination of the human BRAF V600E mutation, comparable in sensitivity to commercial kits. The developed user-friendly electrochemical method offers a simplified and reliable approach for end-point yet quantitative detection of nucleic acids, potentially expanding the benefits of nucleic acid testing in resource-limited settings.
Collapse
Affiliation(s)
- Jianyang Lu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Yiwei Han
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Yanbing Wu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Kaizhi Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Jie Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Peng Miao
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, P. R. China
| | - Genxi Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
36
|
Xu H, Hang Y, Wu Z, Lei X, Deng J, Yang J. Capillary-driven microchip integrated with nickel phosphide hybrid-modified electrode for the electrochemical detection of glucose. Anal Chim Acta 2024; 1316:342882. [PMID: 38969418 DOI: 10.1016/j.aca.2024.342882] [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: 01/13/2024] [Revised: 05/06/2024] [Accepted: 06/16/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Transition metal phosphides with properties similar to platinum metal have received increasing attention for the non-enzymatic detection of glucose. However, the requirement of highly corrosive reagent during sample pretreatment would impose a potential risk to the human body, limiting their practical applications. RESULTS In this study, we report a self-powered microfluidic device for the non-enzymatic detection of glucose using nickel phosphide (Ni2P) hybrid as the catalyst. The Ni2P hybrid is synthesized by pyrolysis of metal-organic framework (MOF)-based precursor and in-situ phosphating process, showing two linear detection ranges (1 μM-1 mM, 1 mM-6 mM) toward glucose with the detection limit of 0.32 μM. The good performance of Ni2P hybrid for glucose is attributed to the synergistic effect of Ni2P active sites and N-doped porous carbon matrix. The microchip is integrated with a NaOH-loaded paper pad and a capillary-based micropump, enabling the automatic NaOH redissolution and delivery of sample solution into the detection chamber. Under the optimized condition, the Ni2P hybrid-based microchip realized the detection of glucose in a user-friendly way. Besides, the feasibility of using this microchip for glucose detection in real serum samples has also been validated. SIGNIFICANCE This article presents a facile fabrication method utilizing a MOF template to synthesize a Ni2P hybrid catalyst. By leveraging the synergy between the Ni2P active sites and the N-doped carbon matrix, an exceptional electrochemical detection performance for glucose has been achieved. Additionally, a self-powered chip device has been developed for convenient glucose detection based on the pre-established high pH environment on the chip.
Collapse
Affiliation(s)
- Hongyang Xu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Yulu Hang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Zhangying Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Xiaoyu Lei
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Jinan Deng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing, 400044, China.
| | - Jun Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education and Bioengineering College, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
37
|
Kaur R, Gupta S, Chauhan A, Mishra V, Sharma MK, Singh J. Harnessing the power of clustered regularly interspaced short palindromic repeats (CRISPR) based microfluidics for next-generation molecular diagnostics. Mol Biol Rep 2024; 51:896. [PMID: 39115550 DOI: 10.1007/s11033-024-09840-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/31/2024] [Indexed: 02/06/2025]
Abstract
CRISPR-based (Clustered regularly interspaced short palindromic repeats-based) technologies have revolutionized molecular biology and diagnostics, offering unprecedented precision and versatility. However, challenges remain, such as high costs, demanding technical expertise, and limited quantification capabilities. To overcome these limitations, innovative microfluidic platforms are emerging as powerful tools for enhancing CRISPR diagnostics. This review explores the exciting intersection of CRISPR and microfluidics, highlighting their potential to revolutionize healthcare diagnostics. By integrating CRISPR's specificity with microfluidics' miniaturization and automation, researchers are developing more sensitive and portable diagnostic tools for a range of diseases. These microfluidic devices streamline sample processing, improve diagnostic performance, and enable point-of-care applications, allowing for rapid and accurate detection of pathogens, genetic disorders, and other health conditions. The review discusses various CRISPR/Cas systems, including Cas9, Cas12, and Cas13, and their integration with microfluidic platforms. It also examines the advantages and limitations of these systems, highlighting their potential for detecting DNA and RNA biomarkers. The review also explores the key challenges in developing and implementing CRISPR-driven microfluidic diagnostics, such as ensuring robustness, minimizing cross-contamination, and achieving robust quantification. Finally, it highlights potential future directions for this rapidly evolving field, emphasizing the transformative potential of these technologies for personalized medicine and global health.
Collapse
Affiliation(s)
- Rasanpreet Kaur
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, 281406, Mathura, Uttar Pradesh, India
| | - Saurabh Gupta
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, 281406, Mathura, Uttar Pradesh, India.
| | - Arjun Chauhan
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, 281406, Mathura, Uttar Pradesh, India
| | - Vidhi Mishra
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Chaumuhan, 281406, Mathura, Uttar Pradesh, India
| | - Manish Kumar Sharma
- Department of Biotechnology, Dr. Rammanohar Lohia Avadh University, Ayodhya, 224001, Uttar Pradesh, India
| | - Jitendra Singh
- Department of Translational Medicine, All India Institute of Medical Sciences Bhopal, Saket Nagar, Bhopal, 462020, Madhya Pradesh, India
| |
Collapse
|
38
|
Piranej S, Zhang L, Bazrafshan A, Marin M, Melikian GB, Salaita K. Rolosense: Mechanical Detection of SARS-CoV-2 Using a DNA-Based Motor. ACS CENTRAL SCIENCE 2024; 10:1332-1347. [PMID: 39071064 PMCID: PMC11273449 DOI: 10.1021/acscentsci.4c00312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 07/30/2024]
Abstract
Assays that detect viral infections play a significant role in limiting the spread of diseases such as SARS-CoV-2. Here, we present Rolosense, a virus sensing platform that leverages the motion of 5 μm DNA-based motors on RNA fuel chips to transduce the presence of viruses. Motors and chips are modified with aptamers, which are designed for multivalent binding to viral targets and lead to stalling of motion. Therefore, the motors perform a "mechanical test" of the viral target and stall in the presence of whole virions, which represents a unique mechanism of transduction distinct from conventional assays. Rolosense can detect SARS-CoV-2 spiked in artificial saliva and exhaled breath condensate with a sensitivity of 103 copies/mL and discriminates among other respiratory viruses. The assay is modular and amenable to multiplexing, as demonstrated by our one-pot detection of influenza A and SARS-CoV-2. As a proof of concept, we show that readout can be achieved using a smartphone camera with a microscopic attachment in as little as 15 min without amplification reactions. Taken together, these results show that mechanical detection using Rolosense can be broadly applied to any viral target and has the potential to enable rapid, low-cost point-of-care screening of circulating viruses.
Collapse
Affiliation(s)
- Selma Piranej
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Luona Zhang
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Alisina Bazrafshan
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Mariana Marin
- Department
of Pediatrics, Emory University School of
Medicine, Atlanta, Georgia 30322, United States
- Children’s
Healthcare of Atlanta, Atlanta, Georgia 30322, United States
| | - Gregory B. Melikian
- Department
of Pediatrics, Emory University School of
Medicine, Atlanta, Georgia 30322, United States
- Children’s
Healthcare of Atlanta, Atlanta, Georgia 30322, United States
| | - Khalid Salaita
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
39
|
Adedokun G, Alipanah M, Fan ZH. Sample preparation and detection methods in point-of-care devices towards future at-home testing. LAB ON A CHIP 2024; 24:3626-3650. [PMID: 38952234 PMCID: PMC11270053 DOI: 10.1039/d3lc00943b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Timely and accurate diagnosis is critical for effective healthcare, yet nearly half the global population lacks access to basic diagnostics. Point-of-care (POC) testing offers partial solutions by enabling low-cost, rapid diagnosis at the patient's location. At-home POC devices have the potential to advance preventive care and early disease detection. Nevertheless, effective sample preparation and detection methods are essential for accurate results. This review surveys recent advances in sample preparation and detection methods at POC. The goal is to provide an in-depth understanding of how these technologies can enhance at-home POC devices. Lateral flow assays, nucleic acid tests, and virus detection methods are at the forefront of POC diagnostic technology, offering rapid and sensitive tools for identifying and measuring pathogens, biomarkers, and viral infections. By illuminating cutting-edge research on assay development for POC diagnostics, this review aims to accelerate progress towards widely available, user-friendly, at-home health monitoring tools that empower individuals in personalized healthcare in the future.
Collapse
Affiliation(s)
- George Adedokun
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA.
| | - Morteza Alipanah
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA.
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL 32611, USA
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL 32611, USA
| |
Collapse
|
40
|
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
|
41
|
Chen J, Yang D, Zhu G, Zhang R, Wang B, Chang Z, Dai J, Wu W, Rotenberg MY, Fang Y. Automated and ultrasensitive point-of-care glycoprotein detection using boronate-affinity enhanced organic electrochemical transistor patch. Biosens Bioelectron 2024; 255:116229. [PMID: 38554574 DOI: 10.1016/j.bios.2024.116229] [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: 11/09/2023] [Revised: 02/29/2024] [Accepted: 03/14/2024] [Indexed: 04/01/2024]
Abstract
Quantifying trace glycoproteins in biofluids requires ultrasensitive components, but feedback is not available in the current portable platforms of point-of-care (POC) diagnosis technologies. A compact and ultrasensitive bioelectrochemical patch was based on boronate-affinity amplified organic electrochemical transistors (BAAOECTs) for POC use was developed to overcome this dilemma. Benefit from the cascading signal enhancement deriving from boronate-affinity targeting multiple regions of glycoprotein and OECTs' inherent signal amplification capability, the BAAOECTs achieved a detection limit of 300 aM within 25 min, displaying about 3 orders of magnitude improvement in sensitivity compared with the commercial electrochemical luminescence (ECL) kit. By using a microfluidic chip, a microcontroller module, and a wireless sensing system, the testing workflows of the above patch was automated, allowing for running the sample-to-answer pipeline even in a resource-limited environment. The reliability of such portable biosensing platform is well recognized in clinical diagnostic applications of heart failure. Overall, the remarkable enhanced sensitivity and automated workflow of BAAOECTs biosensing platform provide a prospective and generalized design policy for expanding the POC diagnosis capabilities of glycoproteins.
Collapse
Affiliation(s)
- Jing Chen
- Research Center for Translational Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Deqi Yang
- Research Center for Translational Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Guoqi Zhu
- Tongji Hospital, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Ru Zhang
- Research Center for Translational Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Bingfang Wang
- Research Center for Translational Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Zhiqiang Chang
- Research Center for Translational Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Jing Dai
- Research Center for Translational Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Wenjuan Wu
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, PR China
| | - Menahem Y Rotenberg
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yin Fang
- Research Center for Translational Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, PR China; Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital Affiliated to Tongji University, Shanghai, 200120, PR China.
| |
Collapse
|
42
|
Zhang YB, Arizti-Sanz J, Bradley A, Huang Y, Kosoko-Thoroddsen TSF, Sabeti PC, Myhrvold C. CRISPR-Based Assays for Point-of-Need Detection and Subtyping of Influenza. J Mol Diagn 2024; 26:599-612. [PMID: 38901927 DOI: 10.1016/j.jmoldx.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 02/26/2024] [Accepted: 04/02/2024] [Indexed: 06/22/2024] Open
Abstract
The high disease burden of influenza virus poses a significant threat to human health. Optimized diagnostic technologies that combine speed, sensitivity, and specificity with minimal equipment requirements are urgently needed to detect the many circulating species, subtypes, and variants of influenza at the point of need. Here, we introduce such a method using Streamlined Highlighting of Infections to Navigate Epidemics (SHINE), a clustered regularly interspaced short palindromic repeats (CRISPR)-based RNA detection platform. Four SHINE assays were designed and validated for the detection and differentiation of clinically relevant influenza species (A and B) and subtypes (H1N1 and H3N2). When tested on clinical samples, these optimized assays achieved 100% concordance with quantitative RT-PCR. Duplex Cas12a/Cas13a SHINE assays were also developed to detect two targets simultaneously. This study demonstrates the utility of this duplex assay in discriminating two alleles of an oseltamivir resistance (H275Y) mutation as well as in simultaneously detecting influenza A and human RNAse P in patient samples. These assays have the potential to expand influenza detection outside of clinical laboratories for enhanced influenza diagnosis and surveillance.
Collapse
Affiliation(s)
- Yibin B Zhang
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts; Harvard-MIT Program in Health Sciences and Technology, Cambridge, Massachusetts; Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts
| | - Jon Arizti-Sanz
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts; Harvard-MIT Program in Health Sciences and Technology, Cambridge, Massachusetts
| | - A'Doriann Bradley
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts
| | - Yujia Huang
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | | | - Pardis C Sabeti
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts; Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts; Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Cameron Myhrvold
- Department of Molecular Biology, Princeton University, Princeton, New Jersey; Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey; Omenn-Darling Bioengineering Institute, Princeton University, Princeton, New Jersey; Department of Chemistry, Princeton University, Princeton, New Jersey.
| |
Collapse
|
43
|
Xiao Y, Zhou M, Liu C, Gao S, Wan C, Li S, Dai C, Du W, Feng X, Li Y, Chen P, Liu BF. Fully integrated and automated centrifugal microfluidic chip for point-of-care multiplexed molecular diagnostics. Biosens Bioelectron 2024; 255:116240. [PMID: 38554576 DOI: 10.1016/j.bios.2024.116240] [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: 02/05/2024] [Revised: 03/12/2024] [Accepted: 03/21/2024] [Indexed: 04/01/2024]
Abstract
Public health events caused by pathogens have imposed significant economic and societal burdens. However, conventional methods still face challenges including complex operations, the need for trained operators, and sophisticated instruments. Here, we proposed a fully integrated and automated centrifugal microfluidic chip, also termed IACMC, for point-of-care multiplexed molecular diagnostics by harnessing the advantages of active and passive valves. The IACMC incorporates multiple essential components including a pneumatic balance module for sequential release of multiple reagents, a pneumatic centrifugation-assisted module for on-demand solution release, an on-chip silicon membrane module for nucleic acid extraction, a Coriolis force-mediated fluid switching module, and an amplification module. Numerical simulation and visual validation were employed to iterate and optimize the chip's structure. Upon sample loading, the chip automatically executes the entire process of bacterial sample lysis, nucleic acid capture, elution quantification, and isothermal LAMP amplification. By optimizing crucial parameters including centrifugation speed, direction of rotation, and silicone membrane thickness, the chip achieves exceptional sensitivity (twenty-five Salmonella or forty Escherichia coli) and specificity in detecting Escherichia coli and Salmonella within 40 min. The development of IACMC will drive advancements in centrifugal microfluidics for point-of-care testing and holds potential for broader applications in precision medicine including high-throughput biochemical analysis immune diagnostics, and drug susceptibility testing.
Collapse
Affiliation(s)
- Yujin Xiao
- 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; Shenzhen YHLO Biotech Co., Ltd., Shenzhen, Guangdong, 518116, China
| | - Mengfan Zhou
- 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
| | - Changgen Liu
- Shenzhen YHLO Biotech Co., Ltd., Shenzhen, Guangdong, 518116, China
| | - Siyu Gao
- 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
| | - Shunji 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
| | - Chenxi Dai
- 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
| | - Wei Du
- 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
| | - Xiaojun Feng
- 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
| | - 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.
| | - 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
|
44
|
Yin W, Li L, Yang Y, Yang Y, Liang R, Ma L, Dai J, Mao G, Ma Y. Ultra-Sensitive Detection of the SARS-CoV-2 Nucleocapsid Protein via a Clustered Regularly Interspaced Short Palindromic Repeat/Cas12a-Mediated Immunoassay. ACS Sens 2024; 9:3150-3157. [PMID: 38717584 DOI: 10.1021/acssensors.4c00432] [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: 06/29/2024]
Abstract
Tracking trace protein analytes in precision diagnostics is an ongoing challenge. Here, we developed an ultrasensitive detection method for the detection of SARS-CoV-2 nucleocapsid (N) protein by combining enzyme-linked immunosorbent assay (ELISA) with the clustered regularly interspaced short palindromic repeat/CRISPR-associated protein (CRISPR/Cas) system. First, the SARS-CoV-2 N protein bound by the capture antibody adsorbed on the well plate was sequentially coupled with the primary antibody, biotinylated secondary antibody, and streptavidin (SA), followed by biotin primer binding to SA. Subsequently, rolling circle amplification was initiated to generate ssDNA strands, which were targeted by CRISPR/Cas12a to cleave the FAM-ssDNA-BHQ1 probe in trans to generate fluorescence signals. We observed a linear relationship between fluorescence intensity and the logarithm of N protein concentration ranging from 3 fg/mL to 3 × 107 fg/mL. The limit of detection (LOD) was 1 fg/mL, with approximately nine molecules in 1 μL of the sample. This detection sensitivity was 4 orders magnitude higher than that of commercially available ELISA kits (LOD: 5.7 × 104 fg/mL). This method was highly specific and sensitive and could accurately detect SARS-CoV-2 pseudovirus and clinical samples, providing a new approach for ultrasensitive immunoassay of protein biomarkers.
Collapse
Affiliation(s)
- Wen Yin
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Leyao Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and Immunity, National Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen 518112, China
| | - Yuxin Yang
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ruijing Liang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lixin Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, China
| | - Junbiao Dai
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Guobin Mao
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yingxin Ma
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
45
|
Aminiranjbar Z, Gultakti CA, Alangari MN, Wang Y, Demir B, Koker Z, Das AK, Anantram MP, Oren EE, Hihath J. Identifying SARS-CoV-2 Variants Using Single-Molecule Conductance Measurements. ACS Sens 2024; 9:2888-2896. [PMID: 38773960 DOI: 10.1021/acssensors.3c02734] [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: 05/24/2024]
Abstract
The global COVID-19 pandemic has highlighted the need for rapid, reliable, and efficient detection of biological agents and the necessity of tracking changes in genetic material as new SARS-CoV-2 variants emerge. Here, we demonstrate that RNA-based, single-molecule conductance experiments can be used to identify specific variants of SARS-CoV-2. To this end, we (i) select target sequences of interest for specific variants, (ii) utilize single-molecule break junction measurements to obtain conductance histograms for each sequence and its potential mutations, and (iii) employ the XGBoost machine learning classifier to rapidly identify the presence of target molecules in solution with a limited number of conductance traces. This approach allows high-specificity and high-sensitivity detection of RNA target sequences less than 20 base pairs in length by utilizing a complementary DNA probe capable of binding to the specific target. We use this approach to directly detect SARS-CoV-2 variants of concerns B.1.1.7 (Alpha), B.1.351 (Beta), B.1.617.2 (Delta), and B.1.1.529 (Omicron) and further demonstrate that the specific sequence conductance is sensitive to nucleotide mismatches, thus broadening the identification capabilities of the system. Thus, our experimental methodology detects specific SARS-CoV-2 variants, as well as recognizes the emergence of new variants as they arise.
Collapse
Affiliation(s)
- Zahra Aminiranjbar
- Department of Electrical and Computer Engineering, University of California Davis, Davis, California 95616, United States
| | - Caglanaz Akin Gultakti
- Bionanodesign Laboratory, Department of Biomedical Engineering, TOBB University of Economics and Technology, Ankara 06560, Turkey
- Department of Materials Science & Nanotechnology Engineering, TOBB University of Economics and Technology, Ankara 06560, Turkey
| | - Mashari Nasser Alangari
- Department of Electrical and Computer Engineering, University of California Davis, Davis, California 95616, United States
- Department of Electrical Engineering, University of Hail, Hail 2240, Saudi Arabia
| | - Yiren Wang
- Department of Electrical Engineering, University of Washington, Seattle, Washington 98115, United States
| | - Busra Demir
- Bionanodesign Laboratory, Department of Biomedical Engineering, TOBB University of Economics and Technology, Ankara 06560, Turkey
- Department of Materials Science & Nanotechnology Engineering, TOBB University of Economics and Technology, Ankara 06560, Turkey
| | - Zeynep Koker
- Bionanodesign Laboratory, Department of Biomedical Engineering, TOBB University of Economics and Technology, Ankara 06560, Turkey
| | - Arindam K Das
- Department of Electrical Engineering, University of Washington, Seattle, Washington 98115, United States
- Department of Computer Science and Electrical Engineering, Eastern Washington University, Cheney, Washington 99004,United States
| | - M P Anantram
- Department of Electrical Engineering, University of Washington, Seattle, Washington 98115, United States
| | - Ersin Emre Oren
- Bionanodesign Laboratory, Department of Biomedical Engineering, TOBB University of Economics and Technology, Ankara 06560, Turkey
- Department of Materials Science & Nanotechnology Engineering, TOBB University of Economics and Technology, Ankara 06560, Turkey
| | - Joshua Hihath
- Department of Electrical and Computer Engineering, University of California Davis, Davis, California 95616, United States
- Center for Bioelectronics and Biosensors, School of Electrical, Computer, and Energy Engineering, Arizona State University, Phoenix, Arizona 85287, United States
| |
Collapse
|
46
|
Lee JC, Ryu SM, Lee Y, Jang H, Song J, Kang T, Lee KH, Park S. CRISPR/Cas12a antifouling nanocomposite electrochemical biosensors enable amplification-free detection of Monkeypox virus in complex biological fluids. NANOSCALE 2024; 16:11318-11326. [PMID: 38804270 DOI: 10.1039/d4nr01618a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The escalating global threat of infectious diseases, including monkeypox virus (MPXV), necessitates advancements in point-of-care diagnostics, moving beyond the constraints of conventional methods tethered to centralized laboratories. Here, we introduce multiple CRISPR RNA (crRNA)-based biosensors that can directly detect MPXV within 35 minutes without pre-amplification, leveraging the enhanced sensitivity and antifouling attributes of the BSA-based nanocomposite. Multiple crRNAs, strategically targeting diverse regions of the F3L gene of MPXV, are designed and combined to amplify Cas12a activation and its collateral cleavage of reporter probes. Notably, our electrochemical sensors exhibit the detection limit of 669 fM F3L gene without amplification, which is approximately a 15-fold improvement compared to fluorescence detection. This sensor also shows negligible changes in peak current after exposure to complex biological fluids, such as whole blood and serum, maintaining its sensitivity at 682 fM. This sensitivity is nearly identical to the conditions when only the F3L gene was present in PBS. In summary, our CRISPR-based electrochemical biosensors can be utilized as a high-performance diagnostic tool in resource-limited settings, representing a transformative leap forward in point-of-care testing. Beyond infectious diseases, the implications of this technology extend to various molecular diagnostics, establishing itself as a rapid, accurate, and versatile platform for detection of target analytes.
Collapse
Affiliation(s)
- Jeong-Chan Lee
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Seuk-Min Ryu
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - YongJin Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Hyowon Jang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Jayeon Song
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- Department of Forensic Sciences, Sungkyunkwan University (SKKU), Suwon-si, Gyeongi-do 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University (SKKU), Suwon-si, Gyeongi-do 16419, Republic of Korea
| | - Taejoon Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- School of Pharmacy, Sungkyunkwan University (SKKU), Suwon-si, Gyeongi-do 16419, Republic of Korea
| | - Kwan Hyi Lee
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Steve Park
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
47
|
Li T, Wang J, Fang J, Chen F, Wu X, Wang L, Gao M, Zhang L, Li S. A universal nucleic acid detection platform combing CRISPR/Cas12a and strand displacement amplification with multiple signal readout. Talanta 2024; 273:125922. [PMID: 38503121 DOI: 10.1016/j.talanta.2024.125922] [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: 08/07/2023] [Revised: 02/08/2024] [Accepted: 03/13/2024] [Indexed: 03/21/2024]
Abstract
Rapid and sensitive detection of nucleic acids has become crucial in various fields. However, most current nucleic acid detection methods can only be used in specific scenarios, such as RT-qPCR, which relies on fluorometer for signal readout, limiting its application at home or in the field due to its high price. In this paper, a universal nucleic acid detection platform combing CRISPR/Cas12a and strand displacement amplification (CRISPR-SDA) with multiple signal readout was established to adapt to different application scenarios. Nucleocapsid protein gene of SARS-CoV-2 (N gene) and hepatitis B virus (HBV) DNA were selected as model targets. The proposed strategy achieved the sensitivity of 53.1 fM, 0.15 pM, and 1 pM for N gene in fluorescence mode, personal glucose meter (PGM) mode and lateral flow assay (LFA) mode, respectively. It possessed the ability to differentiate single-base mismatch and the presence of salmon sperm DNA with a mass up to 105-fold of the targets did not significantly interfere with the assay signal. The general and modular design idea made CRISPR-SDA as simple as building blocks to construct nucleic acid sensing methods to meet different requirements by simply changing the SDA template and selecting suitable signal report probes, which was expected to find a breadth of applications in nucleic acids detection.
Collapse
Affiliation(s)
- Tian Li
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China.
| | - Jinjin Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China
| | - Jiaoyuan Fang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China
| | - Fei Chen
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China
| | - Xinru Wu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China
| | - Lan Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China
| | - Meng Gao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China
| | - Liping Zhang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China.
| | - Sanqiang Li
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China.
| |
Collapse
|
48
|
Hosnedlova B, Werle J, Cepova J, Narayanan VHB, Vyslouzilova L, Fernandez C, Parikesit AA, Kepinska M, Klapkova E, Kotaska K, Stepankova O, Bjorklund G, Prusa R, Kizek R. Electrochemical Sensors and Biosensors for Identification of Viruses: A Critical Review. Crit Rev Anal Chem 2024:1-30. [PMID: 38753964 DOI: 10.1080/10408347.2024.2343853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Due to their life cycle, viruses can disrupt the metabolism of their hosts, causing diseases. If we want to disrupt their life cycle, it is necessary to identify their presence. For this purpose, it is possible to use several molecular-biological and bioanalytical methods. The reference selection was performed based on electronic databases (2020-2023). This review focused on electrochemical methods with high sensitivity and selectivity (53% voltammetry/amperometry, 33% impedance, and 12% other methods) which showed their great potential for detecting various viruses. Moreover, the aforementioned electrochemical methods have considerable potential to be applicable for care-point use as they are portable due to their miniaturizability and fast speed analysis (minutes to hours), and are relatively easy to interpret. A total of 2011 articles were found, of which 86 original papers were subsequently evaluated (the majority of which are focused on human pathogens, whereas articles dealing with plant pathogens are in the minority). Thirty-two species of viruses were included in the evaluation. It was found that most of the examined research studies (77%) used nanotechnological modifications. Other ones performed immunological (52%) or genetic analyses (43%) for virus detection. 5% of the reports used peptides to increase the method's sensitivity. When evaluable, 65% of the research studies had LOD values in the order of ng or nM. The vast majority (79%) of the studies represent proof of concept and possibilities with low application potential and a high need of further research experimental work.
Collapse
Affiliation(s)
- Bozena Hosnedlova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Julia Werle
- Department of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Jana Cepova
- Department of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Vedha Hari B Narayanan
- Pharmaceutical Technology Lab, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Lenka Vyslouzilova
- Czech Institute of Informatics, Robotics and Cybernetics, Department of Biomedical Engineering & Assistive Technologies, Czech Technical University in Prague, Prague, Czech Republic
| | - Carlos Fernandez
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen, United Kingdom
| | - Arli Aditya Parikesit
- Department of Bioinformatics, School of Life Sciences, Indonesia International Institute for Life Sciences, Jakarta, Timur, Indonesia
| | - Marta Kepinska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Eva Klapkova
- Department of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Karel Kotaska
- Department of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Olga Stepankova
- Czech Institute of Informatics, Robotics and Cybernetics, Department of Biomedical Engineering & Assistive Technologies, Czech Technical University in Prague, Prague, Czech Republic
| | - Geir Bjorklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| | - Richard Prusa
- Department of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Rene Kizek
- Department of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
49
|
Zhang J, Li Z, Guo C, Guan X, Avery L, Banach D, Liu C. Intrinsic RNA Targeting Triggers Indiscriminate DNase Activity of CRISPR-Cas12a. Angew Chem Int Ed Engl 2024; 63:e202403123. [PMID: 38516796 PMCID: PMC11073899 DOI: 10.1002/anie.202403123] [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: 02/13/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 03/23/2024]
Abstract
The CRISPR-Cas12a system has emerged as a powerful tool for next-generation nucleic acid-based molecular diagnostics. However, it has long been believed to be effective only on DNA targets. Here, we investigate the intrinsic RNA-enabled trans-cleavage activity of AsCas12a and LbCas12a and discover that they can be directly activated by full-size RNA targets, although LbCas12a exhibits weaker trans-cleavage activity than AsCas12a on both single-stranded DNA and RNA substrates. Remarkably, we find that the RNA-activated Cas12a possesses higher specificity in recognizing mutated target sequences compared to DNA activation. Based on these findings, we develop the "Universal Nuclease for Identification of Virus Empowered by RNA-Sensing" (UNIVERSE) assay for nucleic acid testing. We incorporate a T7 transcription step into this assay, thereby eliminating the requirement for a protospacer adjacent motif (PAM) sequence in the target. Additionally, we successfully detect multiple PAM-less targets in HIV clinical samples that are undetectable by the conventional Cas12a assay based on double-stranded DNA activation, demonstrating unrestricted target selection with the UNIVERSE assay. We further validate the clinical utility of the UNIVERSE assay by testing both HIV RNA and HPV 16 DNA in clinical samples. We envision that the intrinsic RNA targeting capability may bring a paradigm shift in Cas12a-based nucleic acid detection and further enhance the understanding of CRISPR-Cas biochemistry.
Collapse
Affiliation(s)
- Jiongyu Zhang
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Ziyue Li
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Chong Guo
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Xin Guan
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Lori Avery
- Department of Pathology and Laboratory Medicine, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
| | - David Banach
- Department of Medicine, Division of Infectious Diseases, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
| | - Changchun Liu
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
| |
Collapse
|
50
|
Li X, Sun R, Pan J, Shi Z, An Z, Dai C, Lv J, Liu G, Liang H, Liu J, Lu Y, Zhang F, Liu Q. Rapid and on-site wireless immunoassay of respiratory virus aerosols via hydrogel-modulated resonators. Nat Commun 2024; 15:4035. [PMID: 38740742 PMCID: PMC11091083 DOI: 10.1038/s41467-024-48294-1] [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: 01/02/2024] [Accepted: 04/26/2024] [Indexed: 05/16/2024] Open
Abstract
Rapid and accurate detection of respiratory virus aerosols is highlighted for virus surveillance and infection control. Here, we report a wireless immunoassay technology for fast (within 10 min), on-site (wireless and battery-free), and sensitive (limit of detection down to fg/L) detection of virus antigens in aerosols. The wireless immunoassay leverages the immuno-responsive hydrogel-modulated radio frequency resonant sensor to capture and amplify the recognition of virus antigen, and flexible readout network to transduce the immuno bindings into electrical signals. The wireless immunoassay achieves simultaneous detection of respiratory viruses such as severe acute respiratory syndrome coronavirus 2, influenza A H1N1 virus, and respiratory syncytial virus for community infection surveillance. Direct detection of unpretreated clinical samples further demonstrates high accuracy for diagnosis of respiratory virus infection. This work provides a sensitive and accurate immunoassay technology for on-site virus detection and disease diagnosis compatible with wearable integration.
Collapse
Affiliation(s)
- Xin Li
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
- Taizhou Key Laboratory of Medical Devices and Advanced Materials, Research Institute of Zhejiang University-Taizhou, Taizhou, 318000, China
| | - Rujing Sun
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Biosafety III Laboratory, Life Science Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jingying Pan
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
- School of Medicine, Zhejiang University, Hangzhou, 310027, China
| | - Zhenghan Shi
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zijian An
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Chaobo Dai
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jingjiang Lv
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Guang Liu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Biosafety III Laboratory, Life Science Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jun Liu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
- Taizhou Key Laboratory of Medical Devices and Advanced Materials, Research Institute of Zhejiang University-Taizhou, Taizhou, 318000, China
| | - Yanli Lu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
- Intelligent Perception Research Institute, Zhejiang Lab, Hangzhou, 311100, China
| | - Fenni Zhang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qingjun Liu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China.
- Taizhou Key Laboratory of Medical Devices and Advanced Materials, Research Institute of Zhejiang University-Taizhou, Taizhou, 318000, China.
| |
Collapse
|