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
|
Foroughi M, Torabinejad M, Angelov N, Ojcius DM, Parang K, Ravnan M, Lam J. Bridging oral and systemic health: exploring pathogenesis, biomarkers, and diagnostic innovations in periodontal disease. Infection 2025:10.1007/s15010-025-02568-y. [PMID: 40418274 DOI: 10.1007/s15010-025-02568-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Accepted: 05/16/2025] [Indexed: 05/27/2025]
Abstract
PURPOSE This narrative review explores the multifaceted links between periodontal diseases (gingivitis and periodontitis) and systemic health conditions, including cardiovascular disease, diabetes, adverse pregnancy outcomes, Alzheimer's disease, cancers, rheumatoid arthritis, and respiratory infections. It aims to synthesize evidence on how local oral infections exert systemic effects and evaluate the potential of diagnostic technologies to monitor these interactions. METHODS This narrative review synthesizes current scientific literature on periodontal disease pathogenesis, focusing on key pathogens (e.g., Porphyromonas gingivalis, Fusobacterium nucleatum) and their roles in driving local and systemic inflammation via virulence factors and microbial dysbiosis. It examines biomarker-based diagnostic approaches (e.g., IL-1β, TNF-α, microbial DNA) in saliva, blood, and gingival crevicular fluid (GCF) and evaluates current and emerging diagnostic tools (e.g., ELISA, PCR, lateral flow assays, biosensors, microfluidics). RESULTS The review highlights that periodontal pathogens contribute to systemic disease through complex mechanisms including persistent inflammation (driven by cytokines like IL-1β, TNF-α), endotoxemia (via LPS, noting pathogen-specific structural variations impacting immune response), molecular mimicry, and immune modulation. Current diagnostic methods provide valuable information but often face limitations in speed, portability, and multiplexing capability needed for comprehensive point-of-care assessment. Emerging technologies, particularly multiplex platforms integrating biosensors or microfluidics, demonstrate significant potential for rapid, user-friendly analysis of multiple biomarkers, facilitating earlier detection and personalized risk stratification, especially in high-risk populations. CONCLUSION Periodontal diseases significantly impact systemic health via intricate microbial and inflammatory pathways. The complexity of these interactions necessitates moving beyond conventional diagnostics towards integrated, advanced technologies. Implementing rapid, multiplex biomarker detection platforms within a multidisciplinary healthcare framework holds the potential to revolutionize early detection of linked conditions, improve personalized management strategies, and ultimately reduce the systemic burden of periodontal disease.
Collapse
Affiliation(s)
- Max Foroughi
- Department of Preventive and Restorative Dentistry, Arthur A. Dugoni School of Dentistry, University of the Pacific, 155 Fifth Street, San Francisco, CA, 94103, USA.
| | - Mahmoud Torabinejad
- Department of Endodontics, School of Dentistry, Loma Linda University School of Dentistry, Loma Linda, CA, USA
| | - Nikola Angelov
- Department of Periodontics and Dental Hygiene, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA
| | - David M Ojcius
- Department of Biomedical Sciences, Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, CA, USA
| | - Keykavous Parang
- Department of Biomedical and Pharmaceutical Sciences, Center for Targeted Drug Delivery, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA, USA
| | - Marcus Ravnan
- Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, CA, USA
| | - Jerika Lam
- Department of Pharmacy Practice, School of Pharmacy, Chapman University, Irvine, CA, USA
| |
Collapse
|
3
|
Kim M, Yun SH, Kim SH, Kim JD. A Compact Real-Time PCR System for Point-of-Care Detection Using a PCB-Based Disposable Chip and Open-Platform CMOS Camera. SENSORS (BASEL, SWITZERLAND) 2025; 25:3159. [PMID: 40431952 PMCID: PMC12115960 DOI: 10.3390/s25103159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2025] [Revised: 05/15/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025]
Abstract
We present a compact and cost-effective real-time PCR system designed for point-of-care testing (POCT), utilizing a PCB-based disposable chip and an open-platform CMOS camera. The system integrates precise thermal cycling with software-synchronized fluorescence detection and provides real-time analysis through a dedicated user interface. To minimize cost and complexity, a polycarbonate reaction chamber was integrated with a PCB-based heater and thermistor. A slanted LED illumination setup and an open-platform USB camera were employed for fluorescence imaging. Signal alignment was enhanced using device-specific region-of-interest (ROI) tracking based on copper pad corner detection. Thermal cycling performance achieved a heating rate of 8.0 °C/s and a cooling rate of -9.3 °C/s, with steady-state accuracy within ±0.1 °C. Fluorescence images exhibited high dynamic range without saturation, and the 3σ-based ROI correction method improved signal reliability. System performance was validated using Chlamydia trachomatis DNA standard (103 copies), yielding consistent amplification curves with a Ct standard deviation below 0.3 cycles. These results demonstrate that the proposed system enables rapid, accurate, and reproducible nucleic acid detection, making it a strong candidate for field-deployable molecular diagnostics.
Collapse
Affiliation(s)
- MinGin Kim
- Thermo Fisher Scientific, South San Francisco, CA 94080, USA;
| | - Sung-Hun Yun
- School of Software, Hallym University, Chuncheon-si 24252, Republic of Korea;
| | - Sun-Hee Kim
- Department of Fashion Industry, Incheon National University, Incheon 22012, Republic of Korea
| | - Jong-Dae Kim
- School of Software, Hallym University, Chuncheon-si 24252, Republic of Korea;
| |
Collapse
|
4
|
Iarossi M, Verma NC, Bhattacharya I, Meller A. The Emergence of Nanofluidics for Single-Biomolecule Manipulation and Sensing. Anal Chem 2025; 97:8641-8653. [PMID: 40244645 PMCID: PMC12044595 DOI: 10.1021/acs.analchem.4c06684] [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: 12/10/2024] [Revised: 03/16/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Driven by recent advancements in nanofabrication techniques, single-molecule sensing and manipulations in nanofluidic devices are rapidly evolving. These sophisticated biosensors have already had significant impacts on basic research as well as on applications in molecular diagnostics. The nanoscale dimensions of these devices introduce new physical phenomena by confining the biomolecules in at least one dimension, creating effects such as biopolymer linearization, stretching, and separation by mass that are utilized to enhance the biomolecule sensing resolutions. At the same time, the suppressed diffusional motion allows for better single-molecule SNR (signal-to-noise ratio) sensing over time. In particular, nanofluidic devices based on nanochannels have been established as promising technologies for the linearization of ultralong genomic DNA molecules and for optical genome mapping, opening a window to directly observe and infer genome organization. More recently, nanochannels have shown promising capabilities for single-molecule protein sizing, separation, and identification. Consequently, this technology is attracting remarkable interest for applications in single-molecule proteomics. In this review, we discuss the recent advancements of nanochannel-based technologies, focusing on their applications for single-molecule sensing and the characterization of a wide range of biomolecules.
Collapse
Affiliation(s)
- Marzia Iarossi
- Faculty of Biomedical Engineering, Technion -IIT, Haifa 3200003, Israel
| | | | - Ivy Bhattacharya
- Faculty of Biomedical Engineering, Technion -IIT, Haifa 3200003, Israel
| | - Amit Meller
- Faculty of Biomedical Engineering, Technion -IIT, Haifa 3200003, Israel
| |
Collapse
|
5
|
Fang F, Guo H, Guo Z, Lin L, Lai L, Shi Y, You W, Chen S, Liu C, Zhao M, Guo S, Ou Q, Fu Y. A simple and colorimetric method utilizing cell-free toehold switch sensors for the detection of Chlamydia trachomatis, Ureaplasma urealyticum and Neisseria gonorrhoeae. Anal Chim Acta 2025; 1339:343622. [PMID: 39832877 DOI: 10.1016/j.aca.2025.343622] [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: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Sexually transmitted infections (STIs) rank among the most prevalent acute infectious conditions and remain a major global public health concern. Notable STI pathogens include Chlamydia trachomatis (CT), Ureaplasma urealyticum (UU), and Neisseria gonorrhoeae (NG). Early detection and diagnosis are crucial for controlling the spread of STIs. RESULTS In this study, we utilized toehold switches integrated with a cell-free system to develop a simple, colorimetric, sensitive, specific and rapid method for the parallel detection of CT, UU, and NG. Target DNA and sensor DNA were transcribed into target trigger RNA and toehold switch sensor RNA respectively, within a cell-free transcription system. The binding of target RNA to the toehold switch RNA activated the switch, subsequently initiating the translation of the downstream lacZ gene. The expressed LacZ protein hydrolyzed the substrate chlorophenol red-β-d-galactopyranoside (CPRG), resulting in a color change from yellow to purple, which provided a visible colorimetric output. The three screened sensors exhibited excellent orthogonality without any observed cross-reactivity. By enhancing sensitivity with recombinase polymerase amplification (RPA), we reliably detected NG in clinical samples using this method, with no interference from other pathogens. Moreover, we selected high-performance toehold switch sensor for paper-based detection, further enhancing portability. SIGNIFICANCE In summary, this technique enables the simple snd sensitive parallel detection of CT, UU, and NG, generating visible colorimetric results without the need for specialized personnel or sophisticated equipment. Given these advantages, this method holds significant potential as a simple and portable diagnostic tool in resource-limited settings or point-of-care testing (POCT) scenarios.
Collapse
Affiliation(s)
- Fengling Fang
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Clinical Laboratory Diagnostics, The First Clinical College, Fujian Medical University, Fuzhou, 350004, China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, 350004, China
| | - Hongyan Guo
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Clinical Laboratory Diagnostics, The First Clinical College, Fujian Medical University, Fuzhou, 350004, China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, 350004, China; The School of Public Health, Fujian Medical University, 350122, Fuzhou, China
| | - Zhaopei Guo
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Clinical Laboratory Diagnostics, The First Clinical College, Fujian Medical University, Fuzhou, 350004, China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, 350004, China
| | - Lin Lin
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Clinical Laboratory Diagnostics, The First Clinical College, Fujian Medical University, Fuzhou, 350004, China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, 350004, China
| | - Lu Lai
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Clinical Laboratory Diagnostics, The First Clinical College, Fujian Medical University, Fuzhou, 350004, China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, 350004, China
| | - Yue Shi
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Clinical Laboratory Diagnostics, The First Clinical College, Fujian Medical University, Fuzhou, 350004, China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, 350004, China
| | - Weiquan You
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Clinical Laboratory Diagnostics, The First Clinical College, Fujian Medical University, Fuzhou, 350004, China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, 350004, China
| | - Shanjian Chen
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Clinical Laboratory Diagnostics, The First Clinical College, Fujian Medical University, Fuzhou, 350004, China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, 350004, China
| | - Can Liu
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Clinical Laboratory Diagnostics, The First Clinical College, Fujian Medical University, Fuzhou, 350004, China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, 350004, China; Fujian Clinical Research Center for Clinical Immunology Laboratory Test, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350207, China
| | - Mingming Zhao
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, China
| | - Shaobin Guo
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, China
| | - Qishui Ou
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Clinical Laboratory Diagnostics, The First Clinical College, Fujian Medical University, Fuzhou, 350004, China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, 350004, China; Fujian Clinical Research Center for Clinical Immunology Laboratory Test, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350207, China.
| | - Ya Fu
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Clinical Laboratory Diagnostics, The First Clinical College, Fujian Medical University, Fuzhou, 350004, China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Gene Diagnosis Research Center, Fujian Medical University, Fuzhou, 350004, China; Fujian Clinical Research Center for Clinical Immunology Laboratory Test, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China; Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350207, China.
| |
Collapse
|
6
|
Berasarte I, Albizu G, Santos WF, de Lima LF, Ostra M, Vidal M, de Araujo WR. Chemometrics and digital image colorimetry approaches applied to paper-based analytical devices: A review. Anal Chim Acta 2025; 1339:343577. [PMID: 39832870 DOI: 10.1016/j.aca.2024.343577] [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: 05/16/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
Colorimetric paper-based analytical devices (CPADs) are cost-efficient and high-throughput technologies that use readily available materials for point-of-need (PON) applications by leveraging color changes in response to target analytes. However, the complexity of samples can limit the precision and accuracy of CPAD applications. Therefore, CPADs have been combined with chemometric approaches to enhance analytical performance and provide simple solutions to complex systems. The integration of formal optimization techniques, such as Design of Experiments (DoE), classification tools, quantification methods, and other advanced algorithms enables optimal experimental analytical conditions and extracting meaningful information from the complex colorimetric data generated by CPADs. These approaches facilitate robust calibration and prediction models, enabling reliable quantifications or sample differentiation. In addition, chemometrics combined with CPADs contributes to Green Analytical Chemistry once they have the potential to minimize the number of experiments, provide optimal designs and consumption of reagents, and decrease waste generation. The synergy between digital colorimetry using CPADs and smart devices with chemometric techniques holds great promise for portable analysis in resource-limited settings, with applications ranging from environmental monitoring to point-of-care diagnostics. Herein, we review recent advances in the development of CPADs, ranging from manufacturing methods to extraction of color patterns and data treatment using several chemometric tools, performance assessment, and potential transfer to onsite applications for relevant analytical problems.
Collapse
Affiliation(s)
- Irati Berasarte
- Department of Applied Chemistry, University of the Basque Country (UPV/EHU), 20018, Donostia/San Sebastian, Spain
| | - Gorka Albizu
- Department of Applied Chemistry, University of the Basque Country (UPV/EHU), 20018, Donostia/San Sebastian, Spain
| | - Wendel Ferreira Santos
- Laboratório de Sensores Químicos Portáteis, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, 13083-861, Brazil
| | - Lucas Felipe de Lima
- Laboratório de Sensores Químicos Portáteis, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, 13083-861, Brazil
| | - Miren Ostra
- Department of Applied Chemistry, University of the Basque Country (UPV/EHU), 20018, Donostia/San Sebastian, Spain.
| | - Maider Vidal
- Department of Applied Chemistry, University of the Basque Country (UPV/EHU), 20018, Donostia/San Sebastian, Spain.
| | - William Reis de Araujo
- Laboratório de Sensores Químicos Portáteis, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, 13083-861, Brazil.
| |
Collapse
|
7
|
Teymur A, Hussain I, Tang C, Saxena R, Erickson D, Wu T. Portable Fluorescence Microarray Reader-Enabled Biomarker Panel Detection System for Point-of-Care Diagnosis of Lupus Nephritis. MICROMACHINES 2025; 16:156. [PMID: 40047601 PMCID: PMC11857597 DOI: 10.3390/mi16020156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 03/09/2025]
Abstract
Point-of-care (POC) testing has revolutionized diagnostics by providing rapid, accessible solutions outside traditional laboratory settings. However, many POC systems lack the sensitivity or multiplexing capability required for complex diseases. This study introduces an LED-based fluorescence reader designed for POC applications, enabling multiplex detection of lupus nephritis (LN) biomarkers using a biomarker microarray (BMA) slide. The reader integrates an LED excitation source, neutral density (ND) filters for precise intensity control, and onboard image processing with Gaussian smoothing and centroid thresholding to enhance signal detection and localization. Five LN biomarkers (VSIG4, OPN, VCAM1, ALCAM, and TNFRSF1B) were assessed, and performance was validated against a Genepix laser-based scanner. The LED reader demonstrated strong correlation coefficients (r = 0.96-0.98) with the Genepix system for both standard curves and patient samples, achieving robust signal-to-noise ratios and reproducibility across all biomarkers. The multiplex format reduced sample volume and allowed simultaneous analysis of multiple biomarkers. These results highlight the reader's potential to bridge the gap between laboratory-grade precision and POC accessibility. By combining portability, cost-effectiveness, and high analytical performance, this fluorescence reader provides a practical solution for POC diagnostics, particularly in resource-limited settings, improving the feasibility of routine monitoring and early intervention for diseases requiring comprehensive biomarker analysis.
Collapse
Affiliation(s)
- Aygun Teymur
- Department of Biomedical Engineering, University of Houston, Houston, TX 77024, USA; (A.T.); (C.T.)
| | - Iftak Hussain
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14850, USA;
| | - Chenling Tang
- Department of Biomedical Engineering, University of Houston, Houston, TX 77024, USA; (A.T.); (C.T.)
| | - Ramesh Saxena
- Department of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - David Erickson
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14850, USA;
| | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX 77024, USA; (A.T.); (C.T.)
| |
Collapse
|
8
|
Liu Z, Huang X, Liu Z, Zheng S, Yao C, Zhang T, Huang S, Zhang J, Wang J, Farah S, Xie X, Chen HJ. Plug-In Design of the Microneedle Electrode Array for Multi-Parameter Biochemical Sensing in Gouty Arthritis. ACS Sens 2025; 10:159-174. [PMID: 39783825 DOI: 10.1021/acssensors.4c01595] [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: 01/12/2025]
Abstract
Gouty arthritis is one of the most common forms of inflammatory arthritis and has brought a significant burden on patients and society. Current strategies for managing gout primarily focus on long-term urate-lowering therapy. With the rapid advancement of point-of-care testing (POCT) technology, continuous monitoring of gout-related biomarkers like uric acid (UA) or inflammatory cytokines can provide rapid and personalized diagnosis for gout management. In this study, a plug-in design of a microneedle electrode array (PIMNA) was developed and integrated into a multi-parameter sensing portable system in combination with embedded circuits and a mobile application. The system enabled real-time, in situ, and dynamic monitoring of biomarkers, including UA, reactive oxygen species (ROS), and pH at gouty joints. The multi-parameter monitoring system demonstrated a wide linear response range, excellent selectivity, stability, reproducibility, and reliable signal transmission performance. In vivo experiments demonstrated the real-time monitoring capability of PIMNA for UA, ROS, and pH, showing the potential to facilitate urate-lowering management and inflammation assessment. Prospectively, the system enables quantitative analysis of the complexity and diversity of gout, presenting promising applications in clinical practice. This work provides a unique strategy with potential for broader applications in gout management and arthritic disease treatment.
Collapse
Affiliation(s)
- Zhibo Liu
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology; Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xinshuo Huang
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology; Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zhengjie Liu
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology; Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-Sen University, Guangzhou 510006, China
| | - Shantao Zheng
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology; Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-Sen University, Guangzhou 510006, China
| | - Chuanjie Yao
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology; Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-Sen University, Guangzhou 510006, China
| | - Tao Zhang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen 518107, China
| | - Shuang Huang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen 518107, China
| | - Junrui Zhang
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jizhuang Wang
- College of Chemistry and Materials Science, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou 510632, China
| | - Shady Farah
- The Laboratory for Advanced Functional/Medicinal Polymers & Smart Drug Delivery, Technologies, The Wolfson Faculty of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology; Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-Sen University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen 518107, China
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, School of Electronics and Information Technology; Guangdong Province Key Laboratory of Display Material and Technology, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
9
|
Leuthner M, Helou M, Reisbeck M, Hayden O. Advancing magnetic flow cytometry to quantitative epitope analysis in high hematocrit conditions for point-of-care testing. Biosens Bioelectron 2025; 268:116867. [PMID: 39476540 DOI: 10.1016/j.bios.2024.116867] [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: 09/14/2024] [Accepted: 10/20/2024] [Indexed: 11/25/2024]
Abstract
Quantitative cell function measurements are essential for many clinical decisions but are primarily tied to centralized laboratories. Limited access to these laboratories in low-resource settings or for immobile patients highlights the urgent need for Point-of-Care testing (POCT) infrastructure. Magnetic flow cytometers (MFC) offer a solution, albeit phenotyping is limited, and sample processing steps like cell lysis or washing increase MFC's workflow complexity. Here, we investigate conditions for novel phenotyping and direct cell concentration quantification in a streamlined workflow suitable for POCT in high hematocrit environments. We characterize magnetic nanoparticles (MNP) by their size, magnetic moment, and opportunities for high signal-to-noise ratios. With adapted theoretical models, we provide the framework for quantifying bound MNPs per cell. This reveals labeling quality and gives insight into system requirements for reliable cell detection and rational cell phenotyping. We investigate temporal labeling dynamics, which show suboptimal MNP binding kinetics in whole blood (WB), leading to long incubation periods and only 50% recovery concentrations. With our streamlined workflow favoring small (<50 nm) MNPs, we quantify CD14+ monocytes in WB and achieve coefficients of variation of <11%. By simultaneously assessing quantitative epitope expression, we extend MFC's capabilities to clinical subtyping for POCT.
Collapse
Affiliation(s)
- Moritz Leuthner
- Heinz-Nixdorf-Chair of Biomedical Electronics, School of Computation, Information and Technology & Munich Institute of Biomedical Engineering, Technical University of Munich, TranslaTUM, Einsteinstraße 25, 81675 Munich, Germany.
| | - Michael Helou
- EarlyBio GmbH, Bottroper Weg 2, 13507 Berlin, Germany
| | - Mathias Reisbeck
- Heinz-Nixdorf-Chair of Biomedical Electronics, School of Computation, Information and Technology & Munich Institute of Biomedical Engineering, Technical University of Munich, TranslaTUM, Einsteinstraße 25, 81675 Munich, Germany
| | - Oliver Hayden
- Heinz-Nixdorf-Chair of Biomedical Electronics, School of Computation, Information and Technology & Munich Institute of Biomedical Engineering, Technical University of Munich, TranslaTUM, Einsteinstraße 25, 81675 Munich, Germany.
| |
Collapse
|
10
|
Favaretto R, Ardoino N, Pucker G, Bellotto N, Mancinelli M, Piccoli G, Bernard M, Vanzetti L, Potrich C, Lunelli L, Pederzolli C, Guardiani C, Pasquardini L. A ring resonators optical sensor for multiple biomarkers detection. Talanta 2025; 282:127035. [PMID: 39418982 DOI: 10.1016/j.talanta.2024.127035] [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: 04/22/2024] [Revised: 09/25/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
In the recent years, the number of Point-Of-Care-Tests (POCTs) available for clinical diagnostic has steadily increased. POCTs provide a near-patient testing with the potential to generate a result quickly so that appropriate treatment can be implemented, leading to improved clinical outcomes compared to traditional laboratory testing. Technological advances, such as miniaturization of sensors and improved instrumentation, have revolutionized POCTs, enabling the development of smaller and more accurate devices. In this context, it has also gained increasing importance the screening of various analytes simultaneously to increase specificity and improve the characterization of the disease. This study is aimed at developing and characterizing a photonic integrated circuit for multiple markers detection, which represents the functional core towards a full developed POCT device for clinical pathology applications. The photonic sensor, based on microring resonators (MRRs), is functionalized by immobilizing specific antibodies on a copolymer layer deposited on the MRR's surfaces. Surface chemical techniques were employed to analyse the surface chemical characteristics while fluorescence microscopy was involved to analyse the resulting bioreceptor surface density. The photonic sensor is characterized for the parallel detection of two biomarkers, the C-Reactive Protein (CRP) and the Creatine-Kinase-MB (CK-MB). The analyte-antibody binding curves were obtained both in buffer and in filtered un-diluted artificial saliva showing promising results both in terms of sensitivity, with limit of detection (LOD) of 103 pM for CRP and 140 pM for CK-MB, and in terms of specificity. These encouraging results let the assembly of a highly sensitive POC device for molecular diagnostics.
Collapse
Affiliation(s)
- Rachele Favaretto
- FTH srl, Via Sommarive 18, 38123, Trento, Italy; Department of Physics, University of Trento, Via Sommarive 14, 38123, Trento, Italy
| | | | - Georg Pucker
- Fondazione Bruno Kessler, Sensors and Devices Center, Via Sommarive 18, 38123, Trento, Italy
| | | | - Mattia Mancinelli
- Department of Physics, University of Trento, Via Sommarive 14, 38123, Trento, Italy
| | - Gioele Piccoli
- Fondazione Bruno Kessler, Sensors and Devices Center, Via Sommarive 18, 38123, Trento, Italy
| | - Martino Bernard
- Fondazione Bruno Kessler, Sensors and Devices Center, Via Sommarive 18, 38123, Trento, Italy
| | - Lia Vanzetti
- Fondazione Bruno Kessler, Sensors and Devices Center, Via Sommarive 18, 38123, Trento, Italy
| | - Cristina Potrich
- Fondazione Bruno Kessler, Sensors and Devices Center, Via Sommarive 18, 38123, Trento, Italy; CNR Institute of Biophysics, Via alla Cascata 56, Povo, 38123, Trento, Italy
| | - Lorenzo Lunelli
- Fondazione Bruno Kessler, Sensors and Devices Center, Via Sommarive 18, 38123, Trento, Italy; CNR Institute of Biophysics, Via alla Cascata 56, Povo, 38123, Trento, Italy
| | - Cecilia Pederzolli
- Fondazione Bruno Kessler, Sensors and Devices Center, Via Sommarive 18, 38123, Trento, Italy
| | | | - Laura Pasquardini
- Indivenire srl, Via Sommarive 18, 38123, Trento, Italy; Department of Engineering, University of Campania "Luigi Vanvitelli", Via Roma 29, 81031, Aversa, Italy.
| |
Collapse
|
11
|
Strohmaier-Nguyen D, Horn C, Baeumner AJ. Innovations in one-step point-of-care testing within microfluidics and lateral flow assays for shaping the future of healthcare. Biosens Bioelectron 2025; 267:116795. [PMID: 39332251 DOI: 10.1016/j.bios.2024.116795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 09/29/2024]
Abstract
Point-of-care testing (POCT) technology, using lateral flow assays and microfluidic systems, facilitates cost-effective diagnosis, timely treatment, ongoing monitoring, and prevention of life-threatening outcomes. Aside from significant advancements demonstrated in academic research, implementation in real-world applications remains frustratingly limited. The divergence between academic developments and practical utility is often due to factors such as operational complexity, low sensitivity and the need for trained personnel. Taking this into consideration, our objective is to present a critical and objective overview of the latest advancements in fully integrated one-step POCT assays for home-testing which would be commercially viable. In particular, aspects of signal amplification, assay design modification, and sample preparation are critically evaluated and their features and medical applications along with future perspective and challenges with respect to minimal user intervention are summarized. Associated with and very important for the one-step POCT realization are also readout devices and fabrication processes. Critical analysis of available and useful technologies are presented in the SI section.
Collapse
Affiliation(s)
- Dan Strohmaier-Nguyen
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93053, Regensburg, Germany
| | - Carina Horn
- Roche Diagnostics GmbH, 68305, Mannheim, Germany
| | - Antje J Baeumner
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93053, Regensburg, Germany.
| |
Collapse
|
12
|
Hamidizadeh M, Martins RF, Bier FF. Point-of-Care Diagnostics Using Self-heating Elements from Smart Food Packaging: Moving Towards Instrument-Free Nucleic Acid-Based Detection. Mol Diagn Ther 2025; 29:67-80. [PMID: 39550729 PMCID: PMC11742007 DOI: 10.1007/s40291-024-00753-7] [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] [Accepted: 10/22/2024] [Indexed: 11/18/2024]
Abstract
Compromising between accuracy and rapidity is an important issue in analytics and diagnostics, often preventing timely and appropriate reactions to disease. This issue is particularly critical for infectious diseases, where reliable and rapid diagnosis is crucial for effective treatment and easier containment, thereby reducing economic and societal impacts. Diagnostic technologies are vital in disease modeling, tracking, treatment decision making, and epidemic containment. At the point-of-care level in modern healthcare, accurate diagnostics, especially those involving genetic-level analysis and nucleic acid amplification techniques, are still needed. However, implementing these techniques in remote or non-laboratory settings poses challenges because of the need for trained personnel and specialized equipment, as all nucleic acid-based diagnostic techniques, such as polymerase chain reaction and isothermal nucleic acid amplification, require temperature cycling or elevated and stabilized temperatures. However, in smart food packaging, there are approved and commercially available methods that use temperature regulation to enable autonomous heat generation without external sources, such as chemical heaters with phase change materials. These approaches could be applied in diagnostics, facilitating point-of-care, electricity-free molecular diagnostics, especially with nucleic acid-based detection methods such as isothermal nucleic acid amplification. In this review, we explore the potential interplay between self-heating elements, isothermal nucleic acid amplification techniques, and phase change materials. This paves the way for the development of truly portable, electricity-free, point-of-care diagnostic tools, particularly advantageous for on-site detection in resource-limited remote settings and for home use.
Collapse
Affiliation(s)
- Mojdeh Hamidizadeh
- Institute of Biochemistry and Biology, Chair of Molecular Bioanalytics and Bioelectronics, University of Potsdam, Potsdam, Germany.
| | - Renata F Martins
- Institute of Molecular Diagnostics and Bioanalytics (IMDB) gGmbH, Potsdam, Germany
| | - Frank F Bier
- Institute of Biochemistry and Biology, Chair of Molecular Bioanalytics and Bioelectronics, University of Potsdam, Potsdam, Germany
- Institute of Molecular Diagnostics and Bioanalytics (IMDB) gGmbH, Potsdam, Germany
| |
Collapse
|
13
|
Wang Y, Wei W, Han Z, Guan X, Yang Y, Li T, Chang Y, Duan X. Acoustic Streaming Tunnel Enables Particle Velocity Stretching in Multiplex Flow Cytometry. Anal Chem 2024; 96:16397-16405. [PMID: 39359114 DOI: 10.1021/acs.analchem.4c03947] [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: 10/04/2024]
Abstract
Multiplexed flow cytometry, known for its powerful high-throughput identification capability, is widely applied across various biomedical and clinical fields. However, classical flow cytometry relies on multichannel lasers and detectors, which are significant in cost and size, limiting their application in miniaturized assays. Herein, we developed an acoustic streaming-based flow cytometry technique that focuses on multisized microbeads flowing sheathlessly. This method enables the discrimination of particle types and the quantification of target protein concentrations using only a single detector. Microbeads of different sizes exhibit distinct behaviors in the continuous acoustic streaming tunnel, leading to an increased velocity difference during their transition under the laser spot. Consequently, a size detection method based on "velocity stretching" has been established. A multiplex assay of three proteins: cardiac troponin I, creatine kinase-MB and myoglobin, in acute myocardial infarction is performed to validate the feasibility and evaluate the performance of the system. This new multiplexed flow cytometry strategy is expected to enable low-cost and onsite detection of multiple biomarkers.
Collapse
Affiliation(s)
- Yaping Wang
- State Key Laboratory of Precision Measuring Technology & Instruments, Tianjin University, Tianjin 300072, China
| | - Wei Wei
- State Key Laboratory of Precision Measuring Technology & Instruments, Tianjin University, Tianjin 300072, China
| | - Ziyu Han
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, Hong Kong SAR 999077, China
| | - Xieruiqi Guan
- State Key Laboratory of Precision Measuring Technology & Instruments, Tianjin University, Tianjin 300072, China
| | - Yang Yang
- State Key Laboratory of Precision Measuring Technology & Instruments, Tianjin University, Tianjin 300072, China
| | - Tiechuan Li
- State Key Laboratory of Precision Measuring Technology & Instruments, Tianjin University, Tianjin 300072, China
| | - Ye Chang
- State Key Laboratory of Precision Measuring Technology & Instruments, Tianjin University, Tianjin 300072, China
| | - Xuexin Duan
- State Key Laboratory of Precision Measuring Technology & Instruments, Tianjin University, Tianjin 300072, China
| |
Collapse
|
14
|
Anzar N, Suleman S, Singh Y, Kumari S, Parvez S, Pilloton R, Narang J. The Evolution of Illicit-Drug Detection: From Conventional Approaches to Cutting-Edge Immunosensors-A Comprehensive Review. BIOSENSORS 2024; 14:477. [PMID: 39451690 PMCID: PMC11506482 DOI: 10.3390/bios14100477] [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: 08/06/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024]
Abstract
The increasing use of illicit drugs has become a major global concern. Illicit drugs interact with the brain and the body altering an individual's mood and behavior. As the substance-of-abuse (SOA) crisis continues to spread across the world, in order to reduce trafficking and unlawful activity, it is important to use point-of-care devices like biosensors. Currently, there are certain conventional detection methods, which include gas chromatography (GC), mass spectrometry (MS), surface ionization, surface-enhanced Raman spectroscopy (SERS), surface plasmon resonance (SPR), electrochemiluminescence (ECL), high-performance liquid chromatography (HPLC), etc., for the detection of abused drugs. These methods have the advantage of high accuracy and sensitivity but are generally laborious, expensive, and require trained operators, along with high sample requirements, and they are not suitable for on-site drug detection scenarios. As a result, there is an urgent need for point-of-care technologies for a variety of drugs that can replace conventional techniques, such as a biosensor, specifically an immunosensor. An immunosensor is an analytical device that integrates an antibody-based recognition element with a transducer to detect specific molecules (antigens). In an immunosensor, the highly selective antigen-antibody interaction is used to identify and quantify the target analyte. The binding event between the antibody and antigen is converted by the transducer into a measurable signal, such as electrical, optical, or electrochemical, which corresponds to the presence and concentration of the analyte in the sample. This paper provides a comprehensive overview of various illicit drugs, the conventional methods employed for their detection, and the advantages of immunosensors over conventional techniques. It highlights the critical need for on-site detection and explores emerging point-of-care testing methods. The paper also outlines future research goals in this field, emphasizing the potential of advanced technologies to enhance the accuracy, efficiency, and convenience of drug detection.
Collapse
Affiliation(s)
- Nigar Anzar
- Department of Biotechnology, School of Chemical and Life Science, Jamia Hamdard University, New Delhi 110062, India; (N.A.); (S.S.); (Y.S.); (S.K.)
| | - Shariq Suleman
- Department of Biotechnology, School of Chemical and Life Science, Jamia Hamdard University, New Delhi 110062, India; (N.A.); (S.S.); (Y.S.); (S.K.)
| | - Yashda Singh
- Department of Biotechnology, School of Chemical and Life Science, Jamia Hamdard University, New Delhi 110062, India; (N.A.); (S.S.); (Y.S.); (S.K.)
| | - Supriya Kumari
- Department of Biotechnology, School of Chemical and Life Science, Jamia Hamdard University, New Delhi 110062, India; (N.A.); (S.S.); (Y.S.); (S.K.)
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Science, Jamia Hamdard University, New Delhi 110062, India;
| | - Roberto Pilloton
- National Research Council, Department of Chemical Sciences and Materials Technology, Institute of Crystallography, 00015 Rome, Italy
| | - Jagriti Narang
- Department of Biotechnology, School of Chemical and Life Science, Jamia Hamdard University, New Delhi 110062, India; (N.A.); (S.S.); (Y.S.); (S.K.)
| |
Collapse
|
15
|
Li Y, Cheng H. Microfluidic point-of-care testing device for multiplexed detection of liver function blood markers. BIOMEDICAL OPTICS EXPRESS 2024; 15:5803-5817. [PMID: 39421784 PMCID: PMC11482181 DOI: 10.1364/boe.533855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/15/2024] [Accepted: 08/15/2024] [Indexed: 10/19/2024]
Abstract
In this work, we developed a novel microfluidic paper-based analytical device to quantify the blood markers of liver function from human fingertips and whole blood samples. The device can quickly acquire information for screening liver injury and supporting clinical decision-making by simultaneously performing quantitative tests for alanine aminotransferase, aspartate aminotransferase, and albumin. We evaluated the detection accuracy and the storage stability of the device using fingertip samples. The yielded results of our device correlated well with those from Mindray BS350s, even under the conditions of 35 °C and 90%RH. Thus, it offers an effective platform for clinical assessment of liver injury particularly in resource-limited areas.
Collapse
Affiliation(s)
- Yingchun Li
- School of Optics and Photonics, Beijing Institute of Technology, Beijing 100000, China
- Dept. of IVD, Jiangsu Konsung Bio-medical Science and Technology Co., Ltd, Jiangsu 212300, China
| | - Haobo Cheng
- School of Optics and Photonics, Beijing Institute of Technology, Beijing 100000, China
| |
Collapse
|
16
|
Wachholz Junior D, Kubota LT. CRISPR-based electrochemical biosensors: an alternative for point-of-care diagnostics? Talanta 2024; 278:126467. [PMID: 38968657 DOI: 10.1016/j.talanta.2024.126467] [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: 04/11/2024] [Revised: 06/17/2024] [Accepted: 06/22/2024] [Indexed: 07/07/2024]
Abstract
The combination of CRISPR technology and electrochemical sensors has sparked a paradigm shift in the landscape of point-of-care (POC) diagnostics. This review explores the dynamic convergence between CRISPR and electrochemical sensing, elucidating their roles in rapid and precise biosensing platforms. CRISPR, renowned for its remarkable precision in genome editing and programmability capability, has found a novel application in conjunction with electrochemical sensors, promising highly sensitive and specific detection of nucleic acids and biomarkers associated with diverse diseases. This article navigates through fundamental principles, research developments, and applications of CRISPR-based electrochemical sensors, highlighting their potential to revolutionize healthcare accessibility and patient outcomes. In addition, some key points and challenges regarding applying CRISPR-powered electrochemical sensors in real POC settings are presented. By discussing recent advancements and challenges in this interdisciplinary field, this review evaluates the potential of these innovative sensors as an alternative for decentralized, rapid, and accurate POC testing, offering some insights into their applications across clinical scenarios and their impact on the future of diagnostics.
Collapse
Affiliation(s)
- Dagwin Wachholz Junior
- Department of Analytical Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), 13083-970, Brazil; National Institute of Science and Technology in Bioanalytic (INCTBio), Brazil
| | - Lauro Tatsuo Kubota
- Department of Analytical Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), 13083-970, Brazil; National Institute of Science and Technology in Bioanalytic (INCTBio), Brazil.
| |
Collapse
|
17
|
Urban N, Hörner M, Weber W, Dincer C. OptoAssay-Light-controlled dynamic bioassay using optogenetic switches. SCIENCE ADVANCES 2024; 10:eadp0911. [PMID: 39321291 PMCID: PMC11423887 DOI: 10.1126/sciadv.adp0911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/21/2024] [Indexed: 09/27/2024]
Abstract
Circumventing the limitations of current bioassays, we introduce a light-controlled assay, OptoAssay, toward wash- and pump-free point-of-care diagnostics. Extending the capabilities of standard bioassays with light-dependent and reversible interaction of optogenetic switches, OptoAssays enable a bidirectional movement of assay components, only by changing the wavelength of light. Demonstrating exceptional versatility, the OptoAssay showcases its efficacy on various substrates, delivering a dynamic bioassay format. The applicability of the OptoAssay is successfully demonstrated by the calibration of a competitive model assay, resulting in a superior limit of detection of 8 pg ml-1, which is beyond those of conventional ELISA tests. In the future, combined with smartphones, OptoAssays could obviate the need for external flow control systems such as pumps or valves and signal readout devices, enabling on-site analysis in resource-limited settings.
Collapse
Affiliation(s)
- Nadine Urban
- University of Freiburg, FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, 79110 Freiburg, Germany
- University of Freiburg, Department of Microsystems Engineering (IMTEK), 79110 Freiburg, Germany
- University of Freiburg, Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, 79104 Freiburg, Germany
| | - Maximillian Hörner
- University of Freiburg, Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, 79104 Freiburg, Germany
| | - Wilfried Weber
- University of Freiburg, Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, 79104 Freiburg, Germany
- INM–Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany
- Saarland University, Department of Materials Science and Engineering, Campus D2 2, 66123 Saarbrücken, Germany
| | - Can Dincer
- University of Freiburg, FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, 79110 Freiburg, Germany
- University of Freiburg, Department of Microsystems Engineering (IMTEK), 79110 Freiburg, Germany
| |
Collapse
|
18
|
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
|
19
|
Mirgh D, Sonar S, Ghosh S, Adhikari MD, Subramaniyan V, Gorai S, Anand K. Landscape of exosomes to modified exosomes: a state of the art in cancer therapy. RSC Adv 2024; 14:30807-30829. [PMID: 39328877 PMCID: PMC11426072 DOI: 10.1039/d4ra04512b] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024] Open
Abstract
Exosomes are a subpopulation of extracellular vesicles (EVs) that naturally originate from endosomes. They play a significant role in cellular communication. Tumor-secreted exosomes play a crucial role in cancer development and significantly contribute to tumorigenesis, angiogenesis, and metastasis by intracellular communication. Tumor-derived exosomes (TEXs) are a promising biomarker source of cancer detection in the early stages. On the other hand, they offer revolutionary cutting-edge approaches to cancer therapeutics. Exosomes offer a cell-free approach to cancer therapeutics, which overcomes immune cell and stem cell therapeutics-based limitations (complication, toxicity, and cost of treatment). There are multiple sources of therapeutic exosomes present (stem cells, immune cells, plant cells, and synthetic and modified exosomes). This article explores the dynamic source of exosomes (plants, mesenchymal stem cells, and immune cells) and their modification (chimeric, hybrid exosomes, exosome-based CRISPR, and drug delivery) based on cancer therapeutic development. This review also highlights exosomes based clinical trials and the challenges and future orientation of exosome research. We hope that this article will inspire researchers to further explore exosome-based cancer therapeutic platforms for precision oncology.
Collapse
Affiliation(s)
- Divya Mirgh
- Vaccine and Immunotherapy Centre, Massachusetts General Hospital Boston USA
| | - Swarup Sonar
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical and Technical Sciences Chennai Tamil Nadu 602105 India
| | - Srestha Ghosh
- Department of Microbiology, Lady Brabourne College Kolkata West Bengal 700017 India
| | - Manab Deb Adhikari
- Department of Biotechnology, University of North Bengal Darjeeling West Bengal India
| | - Vetriselvan Subramaniyan
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University Bandar Sunway Subang Jaya Selangor 47500 Malaysia
| | - Sukhamoy Gorai
- Department of Neurological Sciences, Rush University Medical Center Chicago IL USA
| | - Krishnan Anand
- Precision Medicine and Integrated Nano-Diagnostics (P-MIND) Research Group, Faculty of Health Sciences, University of the Free State Bloemfontein 9300 South Africa
| |
Collapse
|
20
|
Kshirsagar A, Politza AJ, Guan W. Deep Learning Enabled Universal Multiplexed Fluorescence Detection for Point-of-Care Applications. ACS Sens 2024; 9:4017-4027. [PMID: 39010300 PMCID: PMC11421847 DOI: 10.1021/acssensors.4c00860] [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] [Indexed: 07/17/2024]
Abstract
There is a significant demand for multiplexed fluorescence sensing and detection across a range of applications. Yet, the development of portable and compact multiplexable systems remains a substantial challenge. This difficulty largely stems from the inherent need for spectrum separation, which typically requires sophisticated and expensive optical components. Here, we demonstrate a compact, lens-free, and cost-effective fluorescence sensing setup that incorporates machine learning for scalable multiplexed fluorescence detection. This method utilizes low-cost optical components and a pretrained machine learning (ML) model to enable multiplexed fluorescence sensing without optical adjustments. Its multiplexing capability can be easily scaled up through updates to the machine learning model without altering the hardware. We demonstrate its real-world application in a probe-based multiplexed Loop-Mediated Isothermal Amplification (LAMP) assay designed to simultaneously detect three common respiratory viruses within a single reaction. The effectiveness of this approach highlights the system's potential for point-of-care applications that require cost-effective and scalable solutions. The machine learning-enabled multiplexed fluorescence sensing demonstrated in this work would pave the way for widespread adoption in diverse settings, from clinical laboratories to field diagnostics.
Collapse
Affiliation(s)
- Aneesh Kshirsagar
- Department of Electrical Engineering, The Pennsylvania State University, University Park 16802, USA
| | - Anthony J. Politza
- Department of Biomedical Engineering, The Pennsylvania State University, University Park 16802, USA
| | - Weihua Guan
- Department of Electrical Engineering, The Pennsylvania State University, University Park 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park 16802, USA
| |
Collapse
|
21
|
Gagni P, Lodigiani G, Frigerio R, Cretich M, Gori A, Bergamaschi G. Supramolecular Hydrogels for 3D Biosensors and Bioassays. Chemistry 2024; 30:e202400974. [PMID: 38871646 DOI: 10.1002/chem.202400974] [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: 03/08/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/15/2024]
Abstract
Supramolecular hydrogels play a pivotal role in many fields of biomedical research, including emerging applications in designing advanced tools for point-of-care testing, clinical diagnostics, and lab-on-chip analysis. This review outlines the growing relevance of supramolecular hydrogels in biosensing and bioassay devices, highlighting recent advancements that deliver increased sensitivity, real-time monitoring, and multiplexing capabilities through the distinctive properties of these nanomaterials. Furthermore, the exploration extends to additional applications, such as using hydrogels as three-dimensional matrices for cell-based assays.
Collapse
Affiliation(s)
- Paola Gagni
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy (SCITEC-CNR), 20131, Milan, Italy
| | - Giulia Lodigiani
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy (SCITEC-CNR), 20131, Milan, Italy
| | - Roberto Frigerio
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy (SCITEC-CNR), 20131, Milan, Italy
| | - Marina Cretich
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy (SCITEC-CNR), 20131, Milan, Italy
| | - Alessandro Gori
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy (SCITEC-CNR), 20131, Milan, Italy
| | - Greta Bergamaschi
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy (SCITEC-CNR), 20131, Milan, Italy
| |
Collapse
|
22
|
Xia CJ, Li BH, Guo YN, Zhou XH, Zhang RL, Niu YN. Establishment and performance analysis of a new multiplex detection method for influenza an and B virus antigen. World J Clin Cases 2024; 12:5338-5345. [PMID: 39156089 PMCID: PMC11238682 DOI: 10.12998/wjcc.v12.i23.5338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/04/2024] [Accepted: 06/20/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Influenza A and B virus detection is pivotal in epidemiological surveillance and disease management. Rapid and accurate diagnostic techniques are crucial for timely clinical intervention and outbreak prevention. Quantum dot-encoded microspheres have been widely used in immunodetection. The integration of quantum dot-encoded microspheres with flow cytometry is a well-established technique that enables rapid analysis. Thus, establishing a multiplex detection method for influenza A and B virus antigens based on flow cytometry quantum dot microspheres will help in disease diagnosis. AIM To establish a codetection method of influenza A and B virus antigens based on flow cytometry quantum dot-encoded microsphere technology, which forms the foundation for the assays of multiple respiratory virus biomarkers. METHODS Different quantum dot-encoded microspheres were used to couple the monoclonal antibodies against influenza A and B. The known influenza A and B antigens were detected both separately and simultaneously on a flow cytometer, and the detection conditions were optimized to establish the influenza A and B antigen codetection method, which was utilized for their detection in clinical samples. The results were compared with the fluorescence quantitative polymerase chain reaction (PCR) method to validate the clinical performance of this method. RESULTS The limits of detection of this method were 26.1 and 10.7 pg/mL for influenza A and B antigens, respectively, which both ranged from 15.6 to 250000 pg/mL. In the clinical sample evaluation, the proposed method well correlated with the fluorescent quantitative PCR method, with positive, negative, and overall compliance rates of 57.4%, 100%, and 71.6%, respectively. CONCLUSION A multiplex assay for quantitative detection of influenza A and B virus antigens has been established, which is characterized by high sensitivity, good specificity, and a wide detection range and is promising for clinical applications.
Collapse
Affiliation(s)
- Cheng-Jing Xia
- Department of Clinical Laboratory, West Wing, Shenzhen Hospital (Guangming) of University of Chinese Academy of Sciences, Shenzhen 518106, Guangdong Province, China
| | - Bao-Hua Li
- Department of Clinical Laboratory, West Wing, Shenzhen Hospital (Guangming) of University of Chinese Academy of Sciences, Shenzhen 518106, Guangdong Province, China
| | - Yan-Ni Guo
- Department of Clinical Laboratory, West Wing, Shenzhen Hospital (Guangming) of University of Chinese Academy of Sciences, Shenzhen 518106, Guangdong Province, China
| | - Xiao-He Zhou
- Department of Clinical Laboratory, West Wing, Shenzhen Hospital (Guangming) of University of Chinese Academy of Sciences, Shenzhen 518106, Guangdong Province, China
| | - Run-Ling Zhang
- Department of Clinical Laboratory, West Wing, Shenzhen Hospital (Guangming) of University of Chinese Academy of Sciences, Shenzhen 518106, Guangdong Province, China
| | - Ying-No Niu
- Laboratory, Nanjing Vazyme Biotech Co. Ltd, Nanjing 210033, Jiangsu Province, China
| |
Collapse
|
23
|
Gu Z, Chang H, Yang G, Xu B, Miao B, Li J. An integrated electronic tag-based vertical flow assay (e-VFA) with micro-sieve and AlGaN/GaN HEMT sensors for multi-target detection in actual saliva. Analyst 2024; 149:4267-4275. [PMID: 38904993 DOI: 10.1039/d4an00510d] [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: 06/22/2024]
Abstract
Vertical flow assay (VFA) is an effective point-of-care (POC) diagnostic tool for widespread application. Nevertheless, the lack of multi-target detection and multi-signal readout capability still remains a challenge. Herein, a brand new VFA scheme for multi-target saliva detection based on electronic tags was proposed, where AlGaN/GaN HEMT sensors modified with different bio-receptors as electronic tags endowed the VFA with multi-target detection capability. In addition, the use of electronic tags instead of optical tags allowed the VFA to simultaneously carry out direct multi-target readouts, which ensure effective POC diagnostics for saliva analysis. Moreover, by integrating a hydrophilically optimized micro-sieve, impurities like sticky filaments, epidermal cells and other large-scale charged particles in saliva were effectively screened, which enabled the direct detection of saliva using AlGaN/GaN HEMT sensors. Glucose, urea, and cortisol were selected to verify the feasibility of the multi-target e-VFA scheme, and the results showed that the limit of detection (LOD) was as low as 100 aM. The linear response was demonstrated in the dynamic range of 100 aM to 100 μM, and the specificity, long-term stability and validity of the actual saliva test were also verified. These results demonstrated that the as-proposed e-VFA has potential for application in saliva detection for simultaneous multi-target detection, and it is expected to achieve the real-time detection of more biological targets in saliva.
Collapse
Affiliation(s)
- Zhiqi Gu
- Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215125, People's Republic of China.
| | - Hui Chang
- Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215125, People's Republic of China.
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Guo Yang
- Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215125, People's Republic of China.
- School of Electrical and Mechanical Engineering, Changchun University of Science and Technology, Changchun 130022, China
| | - Boxuan Xu
- Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215125, People's Republic of China.
- The College of Materials Science and Engineering, Shanghai University, Shanghai, 200072, People's Republic of China
| | - Bin Miao
- Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215125, People's Republic of China.
| | - Jiadong Li
- Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215125, People's Republic of China.
| |
Collapse
|
24
|
Zhang Y, Guo Y, Liu G, Zhou S, Su R, Ma Q, Ge Y, Lu YQ, Cui L, Wang G. Portable all-in-one microfluidic system for CRISPR-Cas13a-based fully integrated multiplexed nucleic acid detection. LAB ON A CHIP 2024; 24:3367-3376. [PMID: 38845509 DOI: 10.1039/d4lc00326h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Point-of-care testing of "sample in, answer out" is urgently needed for communicable diseases. Recently, rapid nucleic acid tests for infectious diseases have been developed for use in resource-limited areas, but they require types of equipment in central laboratories and are poorly integrated. In this work, a portable centrifugal microfluidic testing system is developed, integrated with magnetic bead-based nucleic acid extraction, recombinase-assisted amplification and CRISPR-Cas13a detection. The system, with the advantage of its power-supplied active rotating chip and highly programable flow control through integrated addressable active thermally-triggered wax valves, has a rapid turnaround time within 45 min, requiring only one user step. All reagents are preloaded into the chip and can be automatically released. By exploiting a multichannel chip, it is capable of simultaneously detecting 10 infectious viruses with limits of detection of 1 copy per reaction and 5 copies per reaction in plasmid samples and mock plasma samples, respectively. The system was used to analyse clinical plasma samples with good consistency compared to laboratory-based molecular testing. Moreover, the generalizability of our device is reported by successfully testing nasopharyngeal swabs and whole blood samples. The portable device does not require the operation of professional technicians, making it an excellent assay for on-site testing.
Collapse
Affiliation(s)
- Ya Zhang
- College of Engineering and Applied Sciences, Nanjing University, Jiangsu 210093, China.
- Key Laboratory of Intelligent Optical Sensing and Integration of the Ministry of Education, Nanjing University, Jiangsu 210093, China
| | - Yue Guo
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Medical Key Laboratory of Pathogenic Microbiology in Emerging Major Infectious Diseases, Jiangsu 210009, China
- Jiangsu Province Engineering Research Center of Health Emergency, Jiangsu Provincial Center for Disease Control and Prevention, Jiangsu 210009, China.
| | - Guozhen Liu
- College of Engineering and Applied Sciences, Nanjing University, Jiangsu 210093, China.
- Key Laboratory of Intelligent Optical Sensing and Integration of the Ministry of Education, Nanjing University, Jiangsu 210093, China
| | - Shiqi Zhou
- College of Engineering and Applied Sciences, Nanjing University, Jiangsu 210093, China.
- Key Laboratory of Intelligent Optical Sensing and Integration of the Ministry of Education, Nanjing University, Jiangsu 210093, China
| | - Rouyu Su
- College of Engineering and Applied Sciences, Nanjing University, Jiangsu 210093, China.
- Key Laboratory of Intelligent Optical Sensing and Integration of the Ministry of Education, Nanjing University, Jiangsu 210093, China
| | - Qian Ma
- College of Engineering and Applied Sciences, Nanjing University, Jiangsu 210093, China.
- Key Laboratory of Intelligent Optical Sensing and Integration of the Ministry of Education, Nanjing University, Jiangsu 210093, China
| | - Yiyue Ge
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Medical Key Laboratory of Pathogenic Microbiology in Emerging Major Infectious Diseases, Jiangsu 210009, China
- Jiangsu Province Engineering Research Center of Health Emergency, Jiangsu Provincial Center for Disease Control and Prevention, Jiangsu 210009, China.
| | - Yan-Qing Lu
- College of Engineering and Applied Sciences, Nanjing University, Jiangsu 210093, China.
| | - Lunbiao Cui
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Medical Key Laboratory of Pathogenic Microbiology in Emerging Major Infectious Diseases, Jiangsu 210009, China
- Jiangsu Province Engineering Research Center of Health Emergency, Jiangsu Provincial Center for Disease Control and Prevention, Jiangsu 210009, China.
| | - Guanghui Wang
- College of Engineering and Applied Sciences, Nanjing University, Jiangsu 210093, China.
- Key Laboratory of Intelligent Optical Sensing and Integration of the Ministry of Education, Nanjing University, Jiangsu 210093, China
| |
Collapse
|
25
|
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
|
26
|
Wei C, Lei X, Yu S. Multiplexed Detection Strategies for Biosensors Based on the CRISPR-Cas System. ACS Synth Biol 2024; 13:1633-1646. [PMID: 38860462 DOI: 10.1021/acssynbio.4c00161] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
A growing number of applications require simultaneous detection of multiplexed nucleic acid targets in a single reaction, which enables higher information density in combination with reduced assay time and cost. Clustered regularly interspaced short palindromic repeats (CRISPR) and the CRISPR-Cas system have broad applications for the detection of nucleic acids due to their strong specificity, high sensitivity, and excellent programmability. However, realizing multiplexed detection is still challenging for the CRISPR-Cas system due to the nonspecific collateral cleavage activity, limited signal reporting strategies, and possible cross-reactions. In this review, we summarize the principles, strategies, and features of multiplexed detection based on the CRISPR-Cas system and further discuss the challenges and perspective.
Collapse
Affiliation(s)
- Cong Wei
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Xueying Lei
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Songcheng Yu
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
27
|
Zhao X, Jiang Y, Zhou Y, Yan J. Force-dependent rapid immunoassay of high specificity and sensitivity. MECHANOBIOLOGY IN MEDICINE 2024; 2:100061. [PMID: 40395855 PMCID: PMC12082306 DOI: 10.1016/j.mbm.2024.100061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 05/22/2025]
Abstract
The significance of early detection and isolation of infected individuals, along with the quantitative assessment of antibodies against the virus, has gained widespread recognition during the ongoing covid-19 pandemic. This necessitates the development of cost-effective, user-friendly, decentralized testing methods characterized by both high sensitivity and specificity. In this article, we present a comprehensive review of an innovative, low-cost rapid decentralized immunoassay technology, applicable across various diagnostic and quantitative testing scenarios. Distinguishing itself from conventional immunoassay technologies, this method is featured with mechanically enhanced specificity without compromising sensitivity. We delve into the basic principle of the technology and a comparative analysis of this technology in relation to other immunodiagnostic methods, highlighting its potential applications in a wide spectrum of diagnostic tests.
Collapse
Affiliation(s)
- Xiaodan Zhao
- Department of Physics, National University of Singapore, 117542, Singapore
- Centre for Bioimaging Sciences, National University of Singapore, 117557, Singapore
| | - Yanqige Jiang
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Yu Zhou
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Jie Yan
- Department of Physics, National University of Singapore, 117542, Singapore
- Centre for Bioimaging Sciences, National University of Singapore, 117557, Singapore
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China
| |
Collapse
|
28
|
Stibbards-Lyle M, Malinovska J, Badawy S, Schedin P, Rinker KD. Status of breast cancer detection in young women and potential of liquid biopsy. Front Oncol 2024; 14:1398196. [PMID: 38835377 PMCID: PMC11148378 DOI: 10.3389/fonc.2024.1398196] [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: 03/11/2024] [Accepted: 05/01/2024] [Indexed: 06/06/2024] Open
Abstract
Young onset breast cancer (YOBC) is an increasing demographic with unique biology, limited screening, and poor outcomes. Further, women with postpartum breast cancers (PPBCs), cancers occurring up to 10 years after childbirth, have worse outcomes than other young breast cancer patients matched for tumor stage and subtype. Early-stage detection of YOBC is critical for improving outcomes. However, most young women (under 45) do not meet current age guidelines for routine mammographic screening and are thus an underserved population. Other challenges to early detection in this population include reduced performance of standard of care mammography and reduced awareness. Women often face significant barriers in accessing health care during the postpartum period and disadvantaged communities face compounding barriers due to systemic health care inequities. Blood tests and liquid biopsies targeting early detection may provide an attractive option to help address these challenges. Test development in this area includes understanding of the unique biology involved in YOBC and in particular PPBCs that tend to be more aggressive and deadly. In this review, we will present the status of breast cancer screening and detection in young women, provide a summary of some unique biological features of YOBC, and discuss the potential for blood tests and liquid biopsy platforms to address current shortcomings in timely, equitable detection.
Collapse
Affiliation(s)
- Maya Stibbards-Lyle
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Cellular and Molecular Bioengineering Research Lab, University of Calgary, Calgary, AB, Canada
| | - Julia Malinovska
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Cellular and Molecular Bioengineering Research Lab, University of Calgary, Calgary, AB, Canada
| | - Seleem Badawy
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Cellular and Molecular Bioengineering Research Lab, University of Calgary, Calgary, AB, Canada
| | - Pepper Schedin
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Kristina D Rinker
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Cellular and Molecular Bioengineering Research Lab, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
29
|
Yu J, Liu Q, Qi L, Fang Q, Shang X, Zhang X, Du Y. Fluorophore and nanozyme-functionalized DNA walking: A dual-mode DNA logic biocomputing platform for microRNA sensing in clinical samples. Biosens Bioelectron 2024; 252:116137. [PMID: 38401282 DOI: 10.1016/j.bios.2024.116137] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/12/2024] [Accepted: 02/16/2024] [Indexed: 02/26/2024]
Abstract
Inspired by the programmability and modifiability of nucleic acids, point-of-care (POC) diagnostics for nucleic acid target detection is evolving to become more diversified and intelligent. In this study, we introduce a fluorescent and photothermal dual-mode logic biosensing platform that integrates catalytic hairpin assembly (CHA), toehold-mediated stand displacement reaction (SDR) and a DNA walking machine. Dual identification and signal reporting modules are incorporated into DNA circuits, orchestrated by an AND Boolean logic gate operator and magnetic beads (MBs). In the presence of bispecific microRNAs (miRNAs), the AND logic gate activates, driving the DNA walking machine, and facilitating the collection of hairpin DNA stands modified with FAM fluorescent group and CeO2@Au nanoparticles. The CeO2@Au nanoparticles, served as a nanozyme, can oxidize TMB into oxidation TMB (TMBox), enabling a near-infrared (NIR) laser-driven photothermal effect following the magnetic separation of MBs. This versatile platform was employed to differentiate between plasma samples from breast cancer patients, lung cancer patients, and healthy donors. The thermometer-readout transducers, derived from the CeO2@Au@DNA complexes, provided reliable results, further corroborated by fluorescence assays, enhancing the confidence in the diagnostics compared to singular detection method. The dual-mode logic biosensor can be easily customized to various nucleic acid biomarkers and other POC signal readout modalities by adjusting recognition sequences and modification strategies, heralding a promising future in the development of intelligent, flexible diagnostics for POC testing.
Collapse
Affiliation(s)
- Jingyuan Yu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Quanyi Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Lijuan Qi
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Qi Fang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Xudong Shang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Xiaojun Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.
| | - Yan Du
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China.
| |
Collapse
|
30
|
Lou Y, Shi X, Zhou S, Tian J, Cao R. Smartphone-based paper microfluidic detection implementing a versatile quick response code conversion strategy. SENSORS AND ACTUATORS B: CHEMICAL 2024; 406:135393. [DOI: 10.1016/j.snb.2024.135393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
31
|
Kaur G, Tintelott M, Suranglikar M, Masurier A, Vu XT, Gines G, Rondelez Y, Ingebrandt S, Coffinier Y, Pachauri V, Vlandas A. Time-encoded electrical detection of trace RNA biomarker by integrating programmable molecular amplifier on chip. Biosens Bioelectron 2024; 257:116311. [PMID: 38677018 DOI: 10.1016/j.bios.2024.116311] [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: 12/31/2023] [Revised: 04/02/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
One of the serious challenges facing modern point-of-care (PoC) molecular diagnostic platforms relate to reliable detection of low concentration biomarkers such as nucleic acids or proteins in biological samples. Non-specific analyte-receptor interactions due to competitive binding in the presence of abundant molecules, inefficient mass transport and very low number of analyte molecules in sample volume, in general pose critical hurdles for successful implementation of such PoC platforms for clinical use. Focusing on these specific challenges, this work reports a unique PoC biosensor that combines the advantages of nanoscale biologically-sensitive field-effect transistor arrays (BioFET-arrays) realized in a wafer-scale top-down nanofabrication as high sensitivity electrical transducers with that of sophisticated molecular programs (MPs) customized for selective recognition of analyte miRNAs and amplification resulting in an overall augmentation of signal transduction strategy. The MPs realize a programmable universal molecular amplifier (PUMA) in fluidic matrix on chip and provide a biomarker-triggered exponential release of small nucleic acid sequences easily detected by receptor-modified BioFETs. A common miRNA biomarker LET7a was selected for successful demonstration of this novel biosensor, achieving limit of detection (LoD) down to 10 fM and wide dynamic ranges (10 pM-10 nM) in complex physiological solutions. As the determination of biomarker concentration is implemented by following the electrical signal related to analyte-triggered PUMA in time-domain instead of measuring the threshold shifts of BioFETs, and circumvents direct hybridization of biomarkers at transducer surface, this new strategy also allows for multiple usage (>3 times) of the biosensor platform suggesting exceptional cost-effectiveness for practical use.
Collapse
Affiliation(s)
- Gurpreet Kaur
- Institut D'Électronique, de Microélectronique et de Nanotechnologie (IEMN) - UMR CNRS 8520, Univ. Lille Avenue Poincaré, BP 60069, Villeneuve D'Ascq, Cedex, 59652, France
| | - Marcel Tintelott
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, Sommerfeldstrasse 24, 52074, Aachen, Germany
| | - Mohit Suranglikar
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, Sommerfeldstrasse 24, 52074, Aachen, Germany
| | - Antoine Masurier
- Laboratoire Gulliver, Ecole Supérieure de Physique et de Chimie Industrielles, PSL Research University, and CNRS, Paris, France
| | - Xuan-Thang Vu
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, Sommerfeldstrasse 24, 52074, Aachen, Germany
| | - Guillaume Gines
- Laboratoire Gulliver, Ecole Supérieure de Physique et de Chimie Industrielles, PSL Research University, and CNRS, Paris, France
| | - Yannick Rondelez
- Laboratoire Gulliver, Ecole Supérieure de Physique et de Chimie Industrielles, PSL Research University, and CNRS, Paris, France
| | - Sven Ingebrandt
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, Sommerfeldstrasse 24, 52074, Aachen, Germany
| | - Yannick Coffinier
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, Sommerfeldstrasse 24, 52074, Aachen, Germany
| | - Vivek Pachauri
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, Sommerfeldstrasse 24, 52074, Aachen, Germany.
| | - Alexis Vlandas
- Institut D'Électronique, de Microélectronique et de Nanotechnologie (IEMN) - UMR CNRS 8520, Univ. Lille Avenue Poincaré, BP 60069, Villeneuve D'Ascq, Cedex, 59652, France
| |
Collapse
|
32
|
Diforti JF, Cunningham T, Piccinini E, Marmisollé WA, Piccinini JM, Azzaroni O. Noninvasive and Multiplex Self-Test of Kidney Disease Biomarkers with Graphene-Based Lab-on-a-Chip (G-LOC): Toward Digital Diagnostics in the Hands of Patients. Anal Chem 2024; 96:5832-5842. [PMID: 38573917 DOI: 10.1021/acs.analchem.3c05148] [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: 04/06/2024]
Abstract
Chronic kidney disease is one of the major health issues worldwide. However, diagnosis is now highly centralized in large laboratories, resulting in low access to patient monitoring and poor personalized treatments. This work reports the development of a graphene-based lab-on-a-chip (G-LOC) for the digital testing of renal function biomarkers in serum and saliva samples. G-LOC integrates multiple bioelectronic sensors with a microfluidic system that enables multiplex self-testing of urea, potassium, sodium, and chloride. The linearity, limit of detection (LOD), accuracy, and coefficient of variability (CV) were studied. Accuracy values higher than 95.5% and CV lower than 9% were obtained for all of the biomarkers. The analytical performance was compared against three reference lab benchtop analyzers by measuring healthy- and renal-failure-level samples of serum. From receiver operating characteristic (ROC) plots, sensitivities (%) of 99.7, 97.6, 99.1, and 89.0 were obtained for urea, potassium, sodium, and chloride, respectively. Then, the test was evaluated in noninvasive saliva samples and compared against reference methods. Correlation and Bland-Altman plots showed good correlation and agreement of the G-LOC with the reference methods. It is noteworthy that the precision of G-LOC was similar to better than benchtop lab analyzers, with the advantage of being highly portable. Finally, a user testing study was conducted. The analytical performance obtained with untrained volunteers was similar to that obtained with trained chemists. Additionally, based on a user experience survey, G-LOC was found to have very simple usability and would be suitable for at-home diagnostics.
Collapse
Affiliation(s)
- Joaquin F Diforti
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA)-Departamento de Química-Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), CONICET. 64 and 113, 1900 Buenos Aires, Argentina
- Department of Bioelectronic, GISENS BIOTECH, 2150 Shattuck Ave PH, Berkeley, California 94704, United States
| | - Thomas Cunningham
- Department of Bioelectronic, GISENS BIOTECH, 2150 Shattuck Ave PH, Berkeley, California 94704, United States
| | - Esteban Piccinini
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA)-Departamento de Química-Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), CONICET. 64 and 113, 1900 Buenos Aires, Argentina
| | - Waldemar A Marmisollé
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA)-Departamento de Química-Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), CONICET. 64 and 113, 1900 Buenos Aires, Argentina
| | - Jose M Piccinini
- Department of Bioelectronic, GISENS BIOTECH, 2150 Shattuck Ave PH, Berkeley, California 94704, United States
| | - Omar Azzaroni
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA)-Departamento de Química-Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), CONICET. 64 and 113, 1900 Buenos Aires, Argentina
| |
Collapse
|
33
|
Drevinek P, Flisiak R, Nemes R, Nogales Crespo KA, Tomasiewicz K. The Role and Value of Professional Rapid Testing of Acute Respiratory Infections (ARIs) in Europe: A Special Focus on the Czech Republic, Poland, and Romania. Diagnostics (Basel) 2024; 14:631. [PMID: 38535051 PMCID: PMC10969707 DOI: 10.3390/diagnostics14060631] [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: 02/02/2024] [Revised: 03/04/2024] [Accepted: 03/13/2024] [Indexed: 01/03/2025] Open
Abstract
This review aims to explore the role of professional diagnostic rapid testing of acute respiratory infections (ARIs), especially COVID-19 and influenza, ensuring proper disease management and treatment in Europe, and particularly in Czech Republic, Poland, and Romania. The paper was constructed based on a review of scientific evidence and national and international policies and recommendations, as well as a process of validation by four experts. The development of new testing technologies, treatment options, and increased awareness of the negative multidimensional impact of ARI profiles transformed differential diagnosis into a tangible and desirable reality. This review covers the following topics: (1) the multidimensional impact of ARIs, (2) ARI rapid diagnostic testing platforms and their value, (3) the policy landscape, (4) challenges and barriers to implementation, and (5) a set of recommendations illustrating a path forward. The findings indicate that rapid diagnostic testing, including at the point of care (POC), can have a positive impact on case management, antimicrobial and antibiotic stewardship, epidemiological surveillance, and decision making. Integrating this strategy will require the commitment of governments and the international and academic communities, especially as we identified room for improvement in the access and expansion of POC rapid testing in the focus countries and the inclusion of rapid testing in relevant policies.
Collapse
Affiliation(s)
- Pavel Drevinek
- Department of Medical Microbiology, Second Faculty of Medicine, Charles University and Motol University Hospital, 150 06 Prague, Czech Republic;
- Czech Society for Medical Microbiology, Czech Medical Association of J.E. Purkyne, 142 20 Prague, Czech Republic
| | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Bialystok, 15-540 Białystok, Poland;
- Polish Association of Epidemiologists and Infectiologists, 15-540 Białystok, Poland;
| | - Roxana Nemes
- Department of Preclinical Disciplines, Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania;
- Romanian Society of Pneumonology, 050159 Bucharest, Romania
| | | | - Krzysztof Tomasiewicz
- Polish Association of Epidemiologists and Infectiologists, 15-540 Białystok, Poland;
- Department and Clinic of Infectious Diseases and Hepatology SPSK-1, Medical University of Lublin, 20-081 Lublin, Poland
| |
Collapse
|
34
|
Li X, Dang Z, Tang W, Zhang H, Shao J, Jiang R, Zhang X, Huang F. Detection of Parasites in the Field: The Ever-Innovating CRISPR/Cas12a. BIOSENSORS 2024; 14:145. [PMID: 38534252 DOI: 10.3390/bios14030145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024]
Abstract
The rapid and accurate identification of parasites is crucial for prompt therapeutic intervention in parasitosis and effective epidemiological surveillance. For accurate and effective clinical diagnosis, it is imperative to develop a nucleic-acid-based diagnostic tool that combines the sensitivity and specificity of nucleic acid amplification tests (NAATs) with the speed, cost-effectiveness, and convenience of isothermal amplification methods. A new nucleic acid detection method, utilizing the clustered regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) nuclease, holds promise in point-of-care testing (POCT). CRISPR/Cas12a is presently employed for the detection of Plasmodium falciparum, Toxoplasma gondii, Schistosoma haematobium, and other parasites in blood, urine, or feces. Compared to traditional assays, the CRISPR assay has demonstrated notable advantages, including comparable sensitivity and specificity, simple observation of reaction results, easy and stable transportation conditions, and low equipment dependence. However, a common issue arises as both amplification and cis-cleavage compete in one-pot assays, leading to an extended reaction time. The use of suboptimal crRNA, light-activated crRNA, and spatial separation can potentially weaken or entirely eliminate the competition between amplification and cis-cleavage. This could lead to enhanced sensitivity and reduced reaction times in one-pot assays. Nevertheless, higher costs and complex pre-test genome extraction have hindered the popularization of CRISPR/Cas12a in POCT.
Collapse
Affiliation(s)
- Xin Li
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Zhisheng Dang
- National Institute of Parasitic Diseases, Chinese Center for Diseases Control and Prevention (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, National Health Commission of the People's Republic of China (NHC), World Health Organization (WHO) Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai 200025, China
| | - Wenqiang Tang
- State Key Laboratory of Hulless Barley and Yak Germplasm Resources and Genetic Improvement, Lhasa 850002, China
- Tibet Academy of Agriculture and Animal Husbandry Sciences, Lhasa 850002, China
| | - Haoji Zhang
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Jianwei Shao
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Rui Jiang
- College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xu Zhang
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Fuqiang Huang
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| |
Collapse
|
35
|
Link JS, O'Donnell-Sloan J, Curdts S, Geiss BJ, Dandy DS, Henry CS. Multiplexed Capillary-Flow Driven Immunoassay for Respiratory Illnesses. Anal Chem 2024; 96:4111-4119. [PMID: 38417100 DOI: 10.1021/acs.analchem.3c04977] [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: 03/01/2024]
Abstract
Multiplexed analysis in medical diagnostics is widely accepted as a more thorough and complete method compared to single-analyte detection. While analytical methods like polymerase chain reaction and enzyme-linked immunosorbent assay (ELISA) exist for multiplexed detection of biomarkers, they remain time-consuming and expensive. Lateral flow assays (LFAs) are an attractive option for point-of-care testing, and examples of multiplexed LFAs exist. However, these devices are limited by spatial resolution of test lines, large sample volume requirements, cross-reactivity, and poor sensitivity. Recent work has developed capillary-flow microfluidic ELISA platforms as a more sensitive alternative to LFAs; however, multiplexed detection on these types of devices has yet to be demonstrated. In the aftermath of the initial SARS-CoV-2 pandemic, the need for rapid, sensitive point-of-care devices has become ever clearer. Moving forward, devices that can distinguish between diseases with similar presenting symptoms would be the ideal home diagnostic. Here, the first example of a multiplexed capillary-flow immunoassay device for the simultaneous detection of multiple biomarkers is reported. From a single sample addition step, the reagents and washing steps required for two simultaneous ELISAs are delivered to spatially separated test strips. Visual results can be obtained in <15 min, and images captured with a smartphone can be analyzed for quantitative data. This device was used to distinguish between and quantify H1N1 hemagglutinin (HA) and SARS-CoV-2 nucleocapsid protein (N-protein). Using this device, analytical detection limits of 840 and 133 pg/mL were obtained for hemagglutinin and nucleocapsid protein, respectively. The presence of one target in the device did not increase the signal on the other test line, indicating no cross-reactivity between the assays. Additionally, simultaneous detection of both N-protein and HA was performed as well as simultaneous detection of N-protein and human C-reactive protein (CRP). Elevated levels of CRP in a patient infected with SARS-CoV-2 have been shown to correlate with more severe outcomes and a greater risk of death as well. To further expand on the simultaneous detection of two biomarkers, CRP and N-protein were detected simultaneously, and the presence of SARS-CoV-2 N-protein did not interfere with the detection of CRP when both targets were present in the sample.
Collapse
Affiliation(s)
- Jeremy S Link
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523, United States
| | - John O'Donnell-Sloan
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado 80523-1019, United States
| | - Sierra Curdts
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Brian J Geiss
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States
| | - David S Dandy
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado 80523-1019, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523, United States
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado 80523-1019, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States
- Metalluragy and Materials Research Institute, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
36
|
Minea C, Gruson D. Assessment of novel POCT to evaluate liver function. Pract Lab Med 2024; 39:e00367. [PMID: 38328513 PMCID: PMC10847987 DOI: 10.1016/j.plabm.2024.e00367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/09/2024] Open
Abstract
Objectives Point of care testing (POCT) offers the possibility of near bedside patient testing with a reduction of the turn-around time of analysis. The aim of our study was to determine the analytical performances and usability of a recently developed POCT device for the measurement of tests related to liver function. We evaluated the performance of a liver tests panel performed on the LINX EVO® POCT device. Design and methods The imprecision was determined with the Bio-Rad Liquichek Unassayed Chemistry Control. Method comparison was performed with a Cobas® 8000 analyzer. Samples from twenty healthy volunteers were used to verify the reference intervals. Furthermore, practicality was assessed by the healthcare staff handling the POCT device through a dedicated questionnaire. Results The imprecision observed was matching the criteria for the in-lab assay with only one exception, globulin, with an observed imprecision of 6.3 % and a criteria of 5.7 %. With the exception of total and direct bilirubin, the POCT method showed good agreement with the in-lab methods. The verification of reference intervals showed that more than 90 % of the healthy volunteer values were included into the reference interval claimed by the manufacturer except for glucose and globulin. The POCT practicality questionnaire was satisfying overall for users. Conclusions Our study showed very good analytical performances overall for the liver test panel performed on the LINX EVO® POCT instrument.
Collapse
Affiliation(s)
- Carmen Minea
- Department of Clinical Biochemistry, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Damien Gruson
- Department of Clinical Biochemistry, Cliniques Universitaires St-Luc, Université Catholique de Louvain, Brussels, Belgium
- Pôle de recherche en Endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Cliniques Universitaires St-Luc, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
37
|
Guo Z, Cao Y, Fan L, Liu W, Wei L, Ma Y, Ren J, Zhang Q, Cao C. A temperature-independent model of dual calibration standards for onsite and point-of-care quantification analyses via electrophoresis titration chip. Anal Chim Acta 2024; 1289:342207. [PMID: 38245206 DOI: 10.1016/j.aca.2024.342207] [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: 09/22/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024]
Abstract
Electrophoresis titration chip (ETC) is a versatile tool for onsite and point-of-care quantification analyses because it affords naked-eye detection and a straightforward quantification format. However, it is vulnerable to changes in environmental temperature, which regulates the electrophoretic migration by affecting the ion mobility and the target recognition by influencing the enzyme activity. Therefore, the quantification accuracy of the ETC tests was severely compromised. Rather than using the dry bath or heating/cooling units, we proposed a facile model of dual calibration standards (DCS) to mathematically eliminate the effects of temperature on quantification accuracy. To verify our model, we deployed the ETC device at different temperatures ranging from 5 to 40 °C. We further utilized the DCS-ETC to determine the protein content and uric acid concentration in real samples outside the laboratory. All the experimental results showed that our model significantly stabilized the quantification recovery from 35.31-153.44 % to 99.38-103.44 % for protein titration; the recovery of uric acid titration is also stable at 96.25-106.42 %, suggesting the enhanced robustness of the ETC tests. Therefore, DCS-ETC is a field-deployable test that can offer reliable quantification performance without extra equipment for temperature control. We envision that it is promising to be used for onsite applications, including food safety control and disease diagnostics.
Collapse
Affiliation(s)
- Zehua Guo
- School of Sensing Science and Engineering, School of Electronic Information & Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yiren Cao
- School of Sensing Science and Engineering, School of Electronic Information & Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China; School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Liuyin Fan
- Student Innovation Center, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Weiwen Liu
- School of Sensing Science and Engineering, School of Electronic Information & Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Li Wei
- Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai, 200235, China
| | - Yixin Ma
- School of Sensing Science and Engineering, School of Electronic Information & Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jicun Ren
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qiang Zhang
- School of Sensing Science and Engineering, School of Electronic Information & Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Chengxi Cao
- School of Sensing Science and Engineering, School of Electronic Information & Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China; School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
38
|
Chauke SH, Nzuza S, Ombinda-Lemboumba S, Abrahamse H, Dube FS, Mthunzi-Kufa P. Advances in the detection and diagnosis of tuberculosis using optical-based devices. Photodiagnosis Photodyn Ther 2024; 45:103906. [PMID: 38042235 DOI: 10.1016/j.pdpdt.2023.103906] [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: 10/12/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 12/04/2023]
Abstract
Tuberculosis (TB), a bacterial infection caused by Mycobacterium tuberculosis, is highly contagious and can lead to severe health complications if left untreated. This review article discusses the importance of early detection and treatment and its global incidence and epidemiology, emphasizing its impact on vulnerable populations and its role as a major cause of death worldwide. Furthermore, it highlights the challenges faced with diagnosing TB. To overcome these challenges, point-of-care devices have emerged as promising tools for rapid and accurate TB detection. These include devices such as nucleic acid amplification tests (NAATs), lateral flow assays (LFAs), and microfluidic-based assays, which offer advantages such as rapid results, portability, and the ability to detect drug-resistant strains. Optical-based devices, such as photonic micro-ring sensors, silicon platform-based sensors, plasmonic-based platforms, microfluidics, and smartphone imaging, are some of the highlighted optical-based devices with the potential to detect TB. These devices can detect TB in sputum samples with high sensitivity and specificity. Optical-based diagnostic devices have the potential to offer the advantages of detecting low concentrations of target molecules and being adaptable to detect multiple targets simultaneously. Using these devices in a clinical setting makes them suitable for their application in improving access to diagnostic testing that enables earlier detection and treatment of TB. Furthermore, these devices would improve TB's global health issue, which requires comprehensive research, prevention, and treatment efforts.
Collapse
Affiliation(s)
- Sipho H Chauke
- Biophotonics, Photonic Centre, Manufacturing Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria 0001, South Africa; Molecular and Cell Biology Department, University of Cape Town, Cape Town 7701, South Africa.
| | - Sinegugu Nzuza
- Biophotonics, Photonic Centre, Manufacturing Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria 0001, South Africa; Laser Research Centre Department, University of Johannesburg, Johannesburg 2028, South Africa
| | - Saturnin Ombinda-Lemboumba
- Biophotonics, Photonic Centre, Manufacturing Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria 0001, South Africa
| | - Heidi Abrahamse
- Laser Research Centre Department, University of Johannesburg, Johannesburg 2028, South Africa
| | - Felix S Dube
- Molecular and Cell Biology Department, University of Cape Town, Cape Town 7701, South Africa
| | - Patience Mthunzi-Kufa
- Biophotonics, Photonic Centre, Manufacturing Cluster, Council for Scientific and Industrial Research (CSIR), Pretoria 0001, South Africa; Molecular and Cell Biology Department, University of Cape Town, Cape Town 7701, South Africa; School of Interdisciplinary Research and Graduate Studies (UNESCO), University of South Africa, GroenKloof Campus, Pretoria, South Africa
| |
Collapse
|
39
|
Imran JH, Shourav MK, Kim JK. Integrated Point-of-Care Immune Cell Analyzer with Rapid Blood Sample Reaction and Wide Field-of-View Detection. Anal Chem 2024; 96:1640-1650. [PMID: 38247122 DOI: 10.1021/acs.analchem.3c04503] [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: 01/23/2024]
Abstract
The development of affordable, reliable, and rapid diagnostic devices is crucial for monitoring immunological responses using a drop of blood. However, conventional automated diagnostic devices typically involve expensive and impractical robotic fluid-handling approaches. Herein, we developed an integrated cell analyzer comprising a cylindrical sample cartridge connected to a direct current motor and a compact fluorescence imaging module. Sample mixing and loading are performed automatically by a programmable sequence of single motor rotation controlled by an Android application. Two distinct stained immune cell samples can be identified by using two types of fluorescence imaging modes. The effectiveness of mixing performance in antigen-antibody (Ag-Ab) reactions was assessed through a compound objective lens that collects weak fluorescence emitted by the cell membrane. Active mixing with bidirectional rotation of the cartridge in a confined space shortened the Ag-Ab reaction time by a factor of 3.3 and achieved cell counting with higher accuracy while reducing reagent consumption by 4 times compared to the conventional incubation method. High-intensity fluorescence images of cells labeled with a nucleic acid stain were acquired through a single-lens-based fluorescence imaging module with a large field of view (FOV) in an unconventional detection chamber with a curved substrate. Compared with a flat chamber, the curved detection chamber reduces the effects of field curvature and provides aberration-free wide-FOV images, even with a simple lens. Our integrated cell analyzer thus offers a practical and cost-effective solution for monitoring patient immune responses in point-of-care settings.
Collapse
Affiliation(s)
- Jakir Hossain Imran
- Department of Mechanical Engineering, Graduate School, Kookmin University, Seoul 02707, Republic of Korea
| | - Mohiuddin Khan Shourav
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University, Baltimore, Maryland 21231, United States
| | - Jung Kyung Kim
- School of Mechanical Engineering, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
40
|
Klebes A, Ates HC, Verboket RD, Urban GA, von Stetten F, Dincer C, Früh SM. Emerging multianalyte biosensors for the simultaneous detection of protein and nucleic acid biomarkers. Biosens Bioelectron 2024; 244:115800. [PMID: 37925943 DOI: 10.1016/j.bios.2023.115800] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/17/2023] [Accepted: 10/27/2023] [Indexed: 11/07/2023]
Abstract
Traditionally, biosensors are designed to detect one specific analyte. Nevertheless, disease progression is regulated in a highly interactive way by different classes of biomolecules like proteins and nucleic acids. Therefore, a more comprehensive analysis of biomarkers from a single sample is of utmost importance to further improve both, the accuracy of diagnosis as well as the therapeutic success. This review summarizes fundamentals like biorecognition and sensing strategies for the simultaneous detection of proteins and nucleic acids and discusses challenges related to multianalyte biosensor development. We present an overview of the current state of biosensors for the combined detection of protein and nucleic acid biomarkers associated with widespread diseases, among them cancer and infectious diseases. Furthermore, we outline the multianalyte analysis in the rapidly evolving field of single-cell multiomics, to stress its significance for the future discovery and validation of biomarkers. Finally, we provide a critical perspective on the performance and translation potential of multianalyte biosensors for medical diagnostics.
Collapse
Affiliation(s)
- Anna Klebes
- Hahn-Schickard, 79110, Freiburg, Germany; University of Freiburg, IMTEK - Department of Microsystems Engineering, Laboratory for MEMS Applications, 79110, Freiburg, Germany
| | - H Ceren Ates
- University of Freiburg, IMTEK - Department of Microsystems Engineering, Disposable Microsystems Group, 79110, Freiburg, Germany; University of Freiburg, FIT Freiburg Centre for Interactive Materials and Bioinspired Technology, 79110, Freiburg, Germany
| | - René D Verboket
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Johann Wolfgang Goethe University, 60590, Frankfurt am Main, Germany
| | - Gerald A Urban
- University of Freiburg, IMTEK - Department of Microsystems Engineering, Laboratory for Sensors, 79110, Freiburg, Germany; University of Freiburg, Freiburg Materials Research Centre - FMF, 79104, Freiburg, Germany
| | - Felix von Stetten
- Hahn-Schickard, 79110, Freiburg, Germany; University of Freiburg, IMTEK - Department of Microsystems Engineering, Laboratory for MEMS Applications, 79110, Freiburg, Germany
| | - Can Dincer
- University of Freiburg, IMTEK - Department of Microsystems Engineering, Disposable Microsystems Group, 79110, Freiburg, Germany; University of Freiburg, FIT Freiburg Centre for Interactive Materials and Bioinspired Technology, 79110, Freiburg, Germany
| | - Susanna M Früh
- Hahn-Schickard, 79110, Freiburg, Germany; University of Freiburg, IMTEK - Department of Microsystems Engineering, Laboratory for MEMS Applications, 79110, Freiburg, Germany
| |
Collapse
|
41
|
Ahmadsaidulu S, Banik O, Kumar P, Kumar S, Banoth E. Microfluidic Point-of-Care Diagnostics for Multi-Disease Detection Using Optical Techniques: A Review. IEEE Trans Nanobioscience 2024; 23:140-147. [PMID: 37399163 DOI: 10.1109/tnb.2023.3291544] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
The lifestyle of modern society is a major contributing factor for the majority of patients suffering from more than one disease. To Screen and diagnose each of those diseases, there is a great need for portable, and economical diagnostic tools, which are highly stipulated to yield rapid and accurate results using a small volume of the samples such as blood, saliva, sweat, etc. Point-of-care Testing (POCT) is one of the approaches to harvest prompt diagnosis of numerous diseases. The Majority of Point-of-Care Devices (POCD) are developed to diagnose one disease within the specimen. On the other hand, multi-disease detection capabilities in the same point-of-care devices are considered to be an efficient candidate to execute the state-of-the-art platform for multi-disease detection. Most of the literature reviews in this field focus on Point-of-Care (POC) devices, their underlying principles of operation, and their potential applications. It is evident from a perusal of the scholarly works that no review articles have been written on multi-disease detection POC devices. A review study analyzing the current level and functionality of multi-disease detection POC devices would be of great use to future researchers and device manufacturers. This review paper is addressing the above gap by focusing on various optical techniques like fluorescence, Absorbance, and Surface Plasmon Resonance (SPR) for multi-disease detection by harnessing the microfluidic-based POC device.
Collapse
|
42
|
Mazzaracchio V, Rios Maciel M, Porto Santos T, Toda-Peters K, Shen AQ. Duplex Electrochemical Microfluidic Sensor for COVID-19 Antibody Detection: Natural versus Vaccine-Induced Humoral Response. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207731. [PMID: 36916701 DOI: 10.1002/smll.202207731] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/03/2023] [Indexed: 06/18/2023]
Abstract
The rapid transmission and resilience of coronavirus disease 2019 (COVID-19) have led to urgent demands in monitoring humoral response for effective vaccine development, thus a multiplex co-detection platform to discriminate infection-induced from vaccine-induced antibodies is needed. Here a duplex electrochemical immunosensor for co-detection of anti-nucleocapsid IgG (N-IgG) and anti-spike IgG (S-IgG) is developed by using a two-working electrode system, via an indirect immunoassay, with antibody quantification obtained by differential pulse voltammetry. The screen-printed electrodes (SPEs) are modified by carbon black and electrodeposited gold nanoflowers for maximized surface areas, enabling the construction of an immunological chain for S-IgG and N-IgG electrochemical detection with enhanced performance. Using an optimized immunoassay protocol, a wide linear range between 30-750 and 20-1000 ng mL-1 , and a limit of detection of 28 and 15 ng mL-1 are achieved to detect N-IgG and S-IgG simultaneously in serum samples. This duplex immunosensor is then integrated in a microfluidic device to obtain significantly reduced detection time (≤ 7 min) while maintaining its analytical performance. The duplex microfluidic immunosensor can be easily expanded into multiplex format to achieve high throughput screening for the sero-surveillance of COVID-19 and other infectious diseases.
Collapse
Affiliation(s)
- Vincenzo Mazzaracchio
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, 904-0495, Japan
- Department of Chemical Science and Technologies, University of Rome "Tor Vergata,", Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Mauricio Rios Maciel
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, 904-0495, Japan
| | - Tatiana Porto Santos
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, 904-0495, Japan
| | - Kazumi Toda-Peters
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, 904-0495, Japan
| | - Amy Q Shen
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, 904-0495, Japan
| |
Collapse
|
43
|
Brannetti S, Gentile S, Chamorro-Garcia A, Barbero L, Del Grosso E, Ricci F. Decorated DNA-Based Scaffolds as Lateral Flow Biosensors. Angew Chem Int Ed Engl 2023; 62:e202313243. [PMID: 37804080 DOI: 10.1002/anie.202313243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/08/2023]
Abstract
Here we develop Lateral Flow Assays (LFAs) that employ as functional elements DNA-based structures decorated with reporter tags and recognition elements. We have rationally re-engineered tile-based DNA tubular structures that can act as scaffolds and can be decorated with recognition elements of different nature (i.e. antigens, aptamers or proteins) and with orthogonal fluorescent dyes. As a proof-of-principle we have developed sandwich and competitive multiplex lateral flow platforms for the detection of several targets, ranging from small molecules (digoxigenin, Dig and dinitrophenol, DNP), to antibodies (Anti-Dig, Anti-DNP and Anti-MUC1/EGFR bispecific antibodies) and proteins (thrombin). Coupling the advantages of functional DNA-based scaffolds together with the simplicity of LFAs, our approach offers the opportunity to detect a wide range of targets with nanomolar sensitivity and high specificity.
Collapse
Affiliation(s)
- Simone Brannetti
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Serena Gentile
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Alejandro Chamorro-Garcia
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Luca Barbero
- RBM-Merck an affiliate of Merck KGaA, Via Ribes 1, 10010, Turin, Italy
| | - Erica Del Grosso
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Francesco Ricci
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| |
Collapse
|
44
|
Bloemen B, Gand M, Vanneste K, Marchal K, Roosens NHC, De Keersmaecker SCJ. Development of a portable on-site applicable metagenomic data generation workflow for enhanced pathogen and antimicrobial resistance surveillance. Sci Rep 2023; 13:19656. [PMID: 37952062 PMCID: PMC10640560 DOI: 10.1038/s41598-023-46771-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/04/2023] [Indexed: 11/14/2023] Open
Abstract
Rapid, accurate and comprehensive diagnostics are essential for outbreak prevention and pathogen surveillance. Real-time, on-site metagenomics on miniaturized devices, such as Oxford Nanopore Technologies MinION sequencing, could provide a promising approach. However, current sample preparation protocols often require substantial equipment and dedicated laboratories, limiting their use. In this study, we developed a rapid on-site applicable DNA extraction and library preparation approach for nanopore sequencing, using portable devices. The optimized method consists of a portable mechanical lysis approach followed by magnetic bead-based DNA purification and automated sequencing library preparation, and resulted in a throughput comparable to a current optimal, laboratory-based protocol using enzymatic digestion to lyse cells. By using spike-in reference communities, we compared the on-site method with other workflows, and demonstrated reliable taxonomic profiling, despite method-specific biases. We also demonstrated the added value of long-read sequencing by recovering reads containing full-length antimicrobial resistance genes, and attributing them to a host species based on the additional genomic information they contain. Our method may provide a rapid, widely-applicable approach for microbial detection and surveillance in a variety of on-site settings.
Collapse
Affiliation(s)
- Bram Bloemen
- Transversal Activities in Applied Genomics, Sciensano, Rue Juliette Wytsman 14, 1050, Brussels, Belgium
- Department of Information Technology, IDLab, Ghent University, IMEC, 9052, Ghent, Belgium
| | - Mathieu Gand
- Transversal Activities in Applied Genomics, Sciensano, Rue Juliette Wytsman 14, 1050, Brussels, Belgium
| | - Kevin Vanneste
- Transversal Activities in Applied Genomics, Sciensano, Rue Juliette Wytsman 14, 1050, Brussels, Belgium
| | - Kathleen Marchal
- Department of Information Technology, IDLab, Ghent University, IMEC, 9052, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052, Ghent, Belgium
| | - Nancy H C Roosens
- Transversal Activities in Applied Genomics, Sciensano, Rue Juliette Wytsman 14, 1050, Brussels, Belgium
| | - Sigrid C J De Keersmaecker
- Transversal Activities in Applied Genomics, Sciensano, Rue Juliette Wytsman 14, 1050, Brussels, Belgium.
| |
Collapse
|
45
|
Guo Z, Smutok O, Ayva CE, Walden P, Parker J, Whitfield J, Vickers CE, Ungerer JPJ, Katz E, Alexandrov K. Development of epistatic YES and AND protein logic gates and their assembly into signalling cascades. NATURE NANOTECHNOLOGY 2023; 18:1327-1334. [PMID: 37500780 DOI: 10.1038/s41565-023-01450-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 06/09/2023] [Indexed: 07/29/2023]
Abstract
The construction and assembly of artificial allosteric protein switches into information and energy processing networks connected to both biological and non-biological systems is a central goal of synthetic biology and bionanotechnology. However, designing protein switches with the desired input, output and performance parameters is challenging. Here we use a range of reporter proteins to demonstrate that their chimeras with duplicated receptor domains produce YES gate protein switches with large (up to 9,000-fold) dynamic ranges and fast (minutes) response rates. In such switches, the epistatic interactions between largely independent synthetic allosteric sites result in an OFF state with minimal background noise. We used YES gate protein switches based on β-lactamase to develop quantitative biosensors of therapeutic drugs and protein biomarkers. Furthermore, we demonstrated the reconfiguration of YES gate switches into AND gate switches controlled by two different inputs, and their assembly into signalling networks regulated at multiple nodes.
Collapse
Affiliation(s)
- Zhong Guo
- ARC Centre of Excellence in Synthetic Biology, Brisbane, Queensland, Australia
- Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biology and Environmental Science, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Oleh Smutok
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY, USA
| | - Cagla Ergun Ayva
- Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biology and Environmental Science, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Patricia Walden
- Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biology and Environmental Science, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Jake Parker
- Yakka Bio, Canberra, New South Wales, Australia
| | - Jason Whitfield
- UNSW Founders, University of New South Wales, Sydney, New South Wales, Australia
| | - Claudia E Vickers
- ARC Centre of Excellence in Synthetic Biology, Brisbane, Queensland, Australia
- Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biology and Environmental Science, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Jacobus P J Ungerer
- Department of Chemical Pathology, Pathology Queensland, Brisbane, Queensland, Australia
- Faculty of Health and Behavioural Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Evgeny Katz
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY, USA
| | - Kirill Alexandrov
- ARC Centre of Excellence in Synthetic Biology, Brisbane, Queensland, Australia.
- Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, Queensland, Australia.
- School of Biology and Environmental Science, Queensland University of Technology, Brisbane, Queensland, Australia.
- CSIRO-QUT Synthetic Biology Alliance, Brisbane, Queensland, Australia.
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, Queensland, Australia.
| |
Collapse
|
46
|
Jia N, Torres de Oliveira L, Bégin-Drolet A, Greener J. A spectIR-fluidic reactor for monitoring fast chemical reaction kinetics with on-chip attenuated total reflection Fourier transform infrared spectroscopy. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:5129-5138. [PMID: 37609867 DOI: 10.1039/d3ay00842h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Microfluidics has emerged as a powerful technology with diverse applications in microbiology, medicine, chemistry, and physics. While its potential for controlling and studying chemical reactions is well recognized, the extraction and analysis of useful chemical information generated within microfluidic devices remain challenging. This is mainly due to the limited tools available for in situ measurements of chemical reactions. In this study, we present a proof-of-concept spectIR-fluidic reactor design that combines microfluidics with Fourier transform infrared (FTIR) spectroscopy for in situ kinetic studies of fast reactions. By integrating a multi-ridge silicon attenuated total reflection (ATR) wafer into the microfluidic device, we enable multi-point measurements for precise reaction time monitoring. As such, this work establishes a validated foundation for studying fast chemical reactions using on-chip ATR-FTIR spectroscopy in a microfluidic reactor environment, which enables simultaneous monitoring of reagents, intermediates, and products using a phosphate proton transfer reaction. The spectIR-fluidic reactor platform offers customizable designs, allowing for the investigation of reactions with various time scales, and has the potential to significantly advance studies exploring reaction mechanisms and optimization.
Collapse
Affiliation(s)
- Nan Jia
- Département de Chimie, Faculté des Sciences et de Génie, Université Laval, Québec, G1V 0A6, Canada.
| | - Leon Torres de Oliveira
- Département de Chimie, Faculté des Sciences et de Génie, Université Laval, Québec, G1V 0A6, Canada.
| | - André Bégin-Drolet
- Département de Génie Mécanique, Faculté des Sciences et de Génie, Université Laval, Québec, G1V 0A6, Canada
| | - Jesse Greener
- Département de Chimie, Faculté des Sciences et de Génie, Université Laval, Québec, G1V 0A6, Canada.
- CHU de Québec, Centre de Recherche du CHU de Québec, Université Laval, Québec, G1L 3L5, Canada
| |
Collapse
|
47
|
Zheng A, Li C, Xu S, Guo Z, Li C, Zhang C, Yao J, Zhang Z, Li J, Du L, Zhao S, Wang C, Zhang W, Zhou L. Efficient Simultaneous Detection of Metabolites Based on Electroenzymatic Assembly Strategy. BME FRONTIERS 2023; 4:0027. [PMID: 37849675 PMCID: PMC10530654 DOI: 10.34133/bmef.0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/18/2023] [Indexed: 10/19/2023] Open
Abstract
Objective and Impact Statement: We describe an electroenzymatic mediator (EM) sensor based on an electroenzymatic assembly peak separation strategy, which can efficiently realize the simultaneous detection of 3 typical cardiovascular disease (CVD) metabolites in 5 μl of plasma under one test. This work has substantial implications toward improving the efficiency of chronic CVD assessment. Introduction: Monitoring CVD of metabolites is strongly associated with disease risk. Independent and time-consuming detection in hospitals is unfavorable for chronic CVD management. Methods: The EM was flexibly designed by the cross-linking of electron mediators and enzymes, and 3 EM layers with different characteristics were assembled on one electrode. Electrons were transferred under tunable potential; 3 metabolites were quantitatively detected by 3 peak currents that correlated with metabolite concentrations. Results: In this study, the EM sensor showed high sensitivity for the simultaneous detection of 3 metabolites with a lower limit of 0.01 mM. The linear correlation between the sensor and clinical was greater than 0.980 for 242 patients, and the consistency of risk assessment was 94.6%. Conclusion: Metabolites could be expanded by the EM, and the sensor could be a promising candidate as a home healthcare tool for CVD risk assessment.
Collapse
Affiliation(s)
- Anran Zheng
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Chao Li
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- Ji Hua Laboratory, Foshan 528000, China
| | - Shengkai Xu
- Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou 215153, Jiangsu Province, China
| | - Zhen Guo
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Ji Hua Laboratory, Foshan 528000, China
| | - Chuanyu Li
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou CASENS Co. Ltd., Suzhou 215163, China
| | - Changsong Zhang
- Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou 215153, Jiangsu Province, China
| | - Jia Yao
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Zhiqi Zhang
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Jinze Li
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou CASENS Co. Ltd., Suzhou 215163, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
| | - Shasha Zhao
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
| | - Wei Zhang
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Ji Hua Laboratory, Foshan 528000, China
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
| | - Lianqun Zhou
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou CASENS Co. Ltd., Suzhou 215163, China
- Ji Hua Laboratory, Foshan 528000, China
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
| |
Collapse
|
48
|
Dey MK, Iftesum M, Devireddy R, Gartia MR. New technologies and reagents in lateral flow assay (LFA) designs for enhancing accuracy and sensitivity. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:4351-4376. [PMID: 37615701 DOI: 10.1039/d3ay00844d] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Lateral flow assays (LFAs) are a popular method for quick and affordable diagnostic testing because they are easy to use, portable, and user-friendly. However, LFA design has always faced challenges regarding sensitivity, accuracy, and complexity of the operation. By integrating new technologies and reagents, the sensitivity and accuracy of LFAs can be improved while minimizing the complexity and potential for false positives. Surface enhanced Raman spectroscopy (SERS), photoacoustic techniques, fluorescence resonance energy transfer (FRET), and the integration of smartphones and thermal readers can improve LFA accuracy and sensitivity. To ensure reliable and accurate results, careful assay design and validation, appropriate controls, and optimization of assay conditions are necessary. Continued innovation in LFA technology is crucial to improving the reliability and accuracy of rapid diagnostic testing and expanding its applications to various areas, such as food testing, water quality monitoring, and environmental testing.
Collapse
Affiliation(s)
- Mohan Kumar Dey
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Maria Iftesum
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Ram Devireddy
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
49
|
Soto J, Linsley C, Song Y, Chen B, Fang J, Neyyan J, Davila R, Lee B, Wu B, Li S. Engineering Materials and Devices for the Prevention, Diagnosis, and Treatment of COVID-19 and Infectious Diseases. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2455. [PMID: 37686965 PMCID: PMC10490511 DOI: 10.3390/nano13172455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023]
Abstract
Following the global spread of COVID-19, scientists and engineers have adapted technologies and developed new tools to aid in the fight against COVID-19. This review discusses various approaches to engineering biomaterials, devices, and therapeutics, especially at micro and nano levels, for the prevention, diagnosis, and treatment of infectious diseases, such as COVID-19, serving as a resource for scientists to identify specific tools that can be applicable for infectious-disease-related research, technology development, and treatment. From the design and production of equipment critical to first responders and patients using three-dimensional (3D) printing technology to point-of-care devices for rapid diagnosis, these technologies and tools have been essential to address current global needs for the prevention and detection of diseases. Moreover, advancements in organ-on-a-chip platforms provide a valuable platform to not only study infections and disease development in humans but also allow for the screening of more effective therapeutics. In addition, vaccines, the repurposing of approved drugs, biomaterials, drug delivery, and cell therapy are promising approaches for the prevention and treatment of infectious diseases. Following a comprehensive review of all these topics, we discuss unsolved problems and future directions.
Collapse
Affiliation(s)
- Jennifer Soto
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Chase Linsley
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yang Song
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Binru Chen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jun Fang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Josephine Neyyan
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Raul Davila
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Brandon Lee
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Benjamin Wu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Song Li
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
50
|
Lu N, Chen J, Rao Z, Guo B, Xu Y. Recent Advances of Biosensors for Detection of Multiple Antibiotics. BIOSENSORS 2023; 13:850. [PMID: 37754084 PMCID: PMC10526323 DOI: 10.3390/bios13090850] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023]
Abstract
The abuse of antibiotics has caused a serious threat to human life and health. It is urgent to develop sensors that can detect multiple antibiotics quickly and efficiently. Biosensors are widely used in the field of antibiotic detection because of their high specificity. Advanced artificial intelligence/machine learning algorithms have allowed for remarkable achievements in image analysis and face recognition, but have not yet been widely used in the field of biosensors. Herein, this paper reviews the biosensors that have been widely used in the simultaneous detection of multiple antibiotics based on different detection mechanisms and biorecognition elements in recent years, and compares and analyzes their characteristics and specific applications. In particular, this review summarizes some AI/ML algorithms with excellent performance in the field of antibiotic detection, and which provide a platform for the intelligence of sensors and terminal apps portability. Furthermore, this review gives a short review of biosensors for the detection of multiple antibiotics.
Collapse
Affiliation(s)
| | | | | | | | - Ying Xu
- School of Automation, Hangzhou Dianzi University, Hangzhou 310018, China
| |
Collapse
|