1
|
Zhang Y, Lv W, Kang Z, Guo A, Li J, Dai C, Zhang M, Gao S, Li S, Miao Z, Chen S, Feng X, Li Y, Chen P, Liu BF. Drip-Dry Strategy Assisted Blu-Ray Disc Biosensor for Fast Point of Care Testing. Anal Chem 2024. [PMID: 39269278 DOI: 10.1021/acs.analchem.4c02651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Discs and numerous other consumer products have been developed for point of care testing (POCT) to replace traditional large and expensive biochemical devices in certain scenarios. Herein, we propose a drip-dry strategy (2D strategy) assisted Blu-ray disc (BD) biosensor, termed BDB, for rapid and portable POCT within 30 min with the cost of a single test < $1. The platform utilizes the covered area formed by the deposition of the substance to be measured on the activated BD surface after the evaporation of water and realizes the quantitative detection of the target through the error readout of free disc quality diagnosis software. As a proof of concept, we first demonstrated the feasibility of direct quantitative detection of substances in solution in a single system through the detection of pure proteins avoiding colorimetric reagent used in traditional optical detection. For the complex mixed systems, we then innovatively utilize the principle that soluble targets promote/inhibit the dissolution of insoluble precipitates to achieve specific detection of targets and successfully apply BDB to the indirect quantitative detection of glutathione (GSH) with LOD of 0.447 mM in the range of 2-16 mM and organophosphorus pesticides (OPs) with LOD of 2.122 × 10-7 M in the range of 1.289 × 10-7-1.289 × 10-4 M. The BDB is widely applicable, easy to operate, and less time-consuming, which is anticipated to provide an alternative method for early, on-site detection or screening.
Collapse
Affiliation(s)
- Yunhao Zhang
- 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
| | - Wenjie Lv
- 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
| | - Zixin Kang
- 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
| | - Anxin Guo
- 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
| | - Junming 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
| | - Mingyu Zhang
- 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
| | - 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
| | - 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
| | - Zeyu Miao
- 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
| | - Sihan 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
| | - 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
|
2
|
Sripada SA, Hosseini M, Ramesh S, Wang J, Ritola K, Menegatti S, Daniele MA. Advances and opportunities in process analytical technologies for viral vector manufacturing. Biotechnol Adv 2024; 74:108391. [PMID: 38848795 DOI: 10.1016/j.biotechadv.2024.108391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/14/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024]
Abstract
Viral vectors are an emerging, exciting class of biologics whose application in vaccines, oncology, and gene therapy has grown exponentially in recent years. Following first regulatory approval, this class of therapeutics has been vigorously pursued to treat monogenic disorders including orphan diseases, entering hundreds of new products into pipelines. Viral vector manufacturing supporting clinical efforts has spurred the introduction of a broad swath of analytical techniques dedicated to assessing the diverse and evolving panel of Critical Quality Attributes (CQAs) of these products. Herein, we provide an overview of the current state of analytics enabling measurement of CQAs such as capsid and vector identities, product titer, transduction efficiency, impurity clearance etc. We highlight orthogonal methods and discuss the advantages and limitations of these techniques while evaluating their adaptation as process analytical technologies. Finally, we identify gaps and propose opportunities in enabling existing technologies for real-time monitoring from hardware, software, and data analysis viewpoints for technology development within viral vector biomanufacturing.
Collapse
Affiliation(s)
- Sobhana A Sripada
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Mahshid Hosseini
- Joint Department of Biomedical Engineering, North Carolina State University, and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA
| | - Srivatsan Ramesh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Junhyeong Wang
- Joint Department of Biomedical Engineering, North Carolina State University, and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA
| | - Kimberly Ritola
- North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA; Neuroscience Center, Brain Initiative Neurotools Vector Core, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA; North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA; Biomanufacturing Training and Education Center, North Carolina State University, 890 Main Campus Dr, Raleigh, NC 27695, USA.
| | - Michael A Daniele
- Joint Department of Biomedical Engineering, North Carolina State University, and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA; North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA; Department of Electrical and Computer Engineering, North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA.
| |
Collapse
|
3
|
Barzegar F, Kamankesh M, Mohammadi A. An efficient microchip electromembrane extraction online with high-performance liquid chromatography for the measurement of nicotine in high consumption vegetables. PHYTOCHEMICAL ANALYSIS : PCA 2024. [PMID: 39031170 DOI: 10.1002/pca.3418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/22/2024]
Abstract
INTRODUCTION Nicotine, a highly addictive substance, is naturally produced in the Solanaceae family of plants, particularly tobacco. The presence of nicotine in plant foods has adverse effects on the lungs, kidneys, heart, and reproductive system. OBJECTIVE A novel three-phase microchip flat electromembrane coupled with online high-performance liquid chromatography (HPLC) was developed to analyze nicotine in tomato, mushroom, eggplant, bell pepper, and red pepper. METHODS The microchip was connected to the HPLC in online mode. All effective variables were optimized to achieve the best extraction response. The use of electric potential and 2-nitrophenyl octyl ether -5% di(2-ethylhexyl) phosphate as a modified supported liquid membrane (SLM) increased the sensitivity and selectivity. RESULTS The optimal extraction voltage, extraction time, and ion balance were 40 V, 10 min and 0, respectively. The dynamic linear range was 0.5-1000 ng g-1. The obtained recovery, relative standard deviation, and enrichment factor were 98%, 7%, and 35, respectively. The limits of detection 0.4 ng g-1 and the limits of quantification were obtained 1.3 ng g-1. The highest (105.0 ng g-1) and lowest (3.4 ng g-1) concentrations of nicotine were obtained for eggplant and tomato, respectively. CONCLUSION Selective electromembrane extraction of nicotine from the donor phase to the acceptor phase was performed by optimizing the main variables influencing the method mechanism. The new channel design in this analytical system and online injection increased efficiency, stability, and repeatability. The results revealed that this method is capable for the efficient determination of trace amount of nicotine in edible vegetables.
Collapse
Affiliation(s)
- Fatemeh Barzegar
- Department of Food Science and Technology, Faculty of Nutrition Science, Food Science and Technology/National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marzieh Kamankesh
- Food Safety Research Center (salt), Semnan University of Medical Sciences, Semnan, Iran
- School of Pharmacy, Semnan University of Medical Sciences, Semnan, Iran
| | - Abdorreza Mohammadi
- Department of Food Science and Technology, Faculty of Nutrition Science, Food Science and Technology/National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Food Safety Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Isokawa M, Nakanishi K, Kanamori T, Sekiguchi T, Funatsu T, Shoji S, Tsunoda M. Pillar Array Mixer for Postcolumn Derivatization Integrated into Liquid Chromatography-Based Microfluidic Device. Anal Chem 2024; 96:11002-11008. [PMID: 38870183 DOI: 10.1021/acs.analchem.4c01669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The chemical derivatization of target analytes can enhance the sensitivity and selectivity of separation-based methods for metabolite analysis using microfluidic devices. However, the development of chromatography-based microfluidic devices with integrated derivatization units is challenging. In this study, a novel derivatization unit with a pillar array (PA)-based mixing channel was developed for postcolumn derivatization during on-chip liquid chromatography (LC). The PA mixer enhanced mixing between the derivatization reagents and analytes in the transverse direction, while preventing analyte dispersion in the flow direction. After the concept was confirmed using computational fluid dynamics analysis, microfluidic devices with a LC column and PA mixer were fabricated on a 20 × 20 mm silicon plate. Fluid experiments were performed using a PA mixer with a pillar size of 5 or 10 μm or a hollow-channel mixer, which revealed that the PA mixer enhanced transverse mixing without increasing the width of the analyte peak. Moreover, the developed device enabled the analysis of three amino acids within 40 s by separation via hydrophilic interaction chromatography followed by postcolumn fluorogenic derivatization with naphthalene-2,3-dicarboxaldehyde and fluorescence detection. Our results demonstrate the potential of integrated derivatization units for the development of micrototal analysis systems for use in bioanalysis.
Collapse
Affiliation(s)
- Muneki Isokawa
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Kanki Nakanishi
- Department of Nanoscience and Nanoengineering, Waseda University, Tokyo 169-8555, Japan
| | - Takahiro Kanamori
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Tetsushi Sekiguchi
- Department of Nanoscience and Nanoengineering, Waseda University, Tokyo 169-8555, Japan
| | - Takashi Funatsu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Shuichi Shoji
- Department of Nanoscience and Nanoengineering, Waseda University, Tokyo 169-8555, Japan
| | - Makoto Tsunoda
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
5
|
Abeywardena SBY, Yue Z, Wallace GG, Innis PC. Novel 3D textile structures and geometries for electrofluidics. Electrophoresis 2024; 45:1171-1181. [PMID: 38837441 DOI: 10.1002/elps.202400020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/03/2024] [Accepted: 05/14/2024] [Indexed: 06/07/2024]
Abstract
The integration of microfluidics with electric field control, commonly referred to as electrofluidics, has led to new opportunities for biomedical analysis. The requirement for closed microcapillary channels in microfluidics, typically formed via complex microlithographic fabrication approaches, limits the direct accessibility to the separation processes during conventional electrofluidic devices. Textile structures provide an alternative and low-cost approach to overcome these limitations via providing open and surface-accessible capillary channels. Herein, we investigate the potential of different 3D textile structures for electrofluidics. In this study, 12 polyester yarns were braided around nylon monofilament cores of different diameters to produce functional 3D core-shell textile structures. Capillary electrophoresis performances of these 3D core-shell textile structures both before and after removing the nylon core were evaluated in terms of mobility and bandwidth of a fluorescence marker compound. It was shown that the fibre arrangement and density govern the inherent capillary formation within these textile structures which also impacts upon the solute analyte mobility and separation bandwidth during electrophoretic studies. Core-shell textile structures with a 0.47 mm nylon core exhibited the highest fluorescein mobility and presented a narrower separation bandwidth. This optimal textile structure was readily converted to different geometries via a simple heat-setting of the central nylon core. This approach can be used to fabricate an array of miniaturized devices that possess many of the basic functionalities required in electrofluidics while maintaining open surface access that is otherwise impractical in classical approaches.
Collapse
Affiliation(s)
- Sujani B Y Abeywardena
- ARC Centre of Excellence for Electromaterials Science (ACES), Intelligent Polymer Research Institute (IPRI), Australian Institute for Innovative Materials (AIIM), Innovation Campus, University of Wollongong, North Wollongong, New South Wales, Australia
| | - Zhilian Yue
- ARC Centre of Excellence for Electromaterials Science (ACES), Intelligent Polymer Research Institute (IPRI), Australian Institute for Innovative Materials (AIIM), Innovation Campus, University of Wollongong, North Wollongong, New South Wales, Australia
| | - Gordon G Wallace
- ARC Centre of Excellence for Electromaterials Science (ACES), Intelligent Polymer Research Institute (IPRI), Australian Institute for Innovative Materials (AIIM), Innovation Campus, University of Wollongong, North Wollongong, New South Wales, Australia
| | - Peter C Innis
- ARC Centre of Excellence for Electromaterials Science (ACES), Intelligent Polymer Research Institute (IPRI), Australian Institute for Innovative Materials (AIIM), Innovation Campus, University of Wollongong, North Wollongong, New South Wales, Australia
| |
Collapse
|
6
|
Xin L, Xiao X, Xiao W, Peng R, Wang H, Pan F. Screening for urothelial carcinoma cells in urine based on digital holographic flow cytometry through machine learning and deep learning methods. LAB ON A CHIP 2024; 24:2736-2746. [PMID: 38660758 DOI: 10.1039/d3lc00854a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The incidence of urothelial carcinoma continues to rise annually, particularly among the elderly. Prompt diagnosis and treatment can significantly enhance patient survival and quality of life. Urine cytology remains a widely-used early screening method for urothelial carcinoma, but it still has limitations including sensitivity, labor-intensive procedures, and elevated cost. In recent developments, microfluidic chip technology offers an effective and efficient approach for clinical urine specimen analysis. Digital holographic microscopy, a form of quantitative phase imaging technology, captures extensive data on the refractive index and thickness of cells. The combination of microfluidic chips and digital holographic microscopy facilitates high-throughput imaging of live cells without staining. In this study, digital holographic flow cytometry was employed to rapidly capture images of diverse cell types present in urine and to reconstruct high-precision quantitative phase images for each cell type. Then, various machine learning algorithms and deep learning models were applied to categorize these cell images, and remarkable accuracy in cancer cell identification was achieved. This research suggests that the integration of digital holographic flow cytometry with artificial intelligence algorithms offers a promising, precise, and convenient approach for early screening of urothelial carcinoma.
Collapse
Affiliation(s)
- Lu Xin
- Key Laboratory of Precision Opto-mechatronics Technology, Beihang University, Beijing 100191, China.
| | - Xi Xiao
- Peking University Third Hospital, Department of Radiation Oncology, Beijing 100191, China.
| | - Wen Xiao
- Key Laboratory of Precision Opto-mechatronics Technology, Beihang University, Beijing 100191, China.
| | - Ran Peng
- Peking University Third Hospital, Department of Radiation Oncology, Beijing 100191, China.
| | - Hao Wang
- Peking University Third Hospital, Department of Radiation Oncology, Beijing 100191, China.
- Peking University Third Hospital, Cancer Center, Beijing 100191, China
| | - Feng Pan
- Key Laboratory of Precision Opto-mechatronics Technology, Beihang University, Beijing 100191, China.
| |
Collapse
|
7
|
Lu S, Tseng J, Chuang L, Chang N, Chen S, Hsu C, Chien J, Lin C, Lee E. Electrophoresis of a weakly charged dielectric fluid droplet in a cylindrical pore. Electrophoresis 2024. [PMID: 38613523 DOI: 10.1002/elps.202300276] [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: 11/29/2023] [Revised: 03/14/2024] [Accepted: 04/02/2024] [Indexed: 04/15/2024]
Abstract
Electrophoresis of a weakly charged dielectric droplet with constant surface charge density in a chargeless cylindrical pore is investigated theoretically in this study, focusing on the boundary confinement effect of the double layer, which in turn determines the ultimate motion of the droplet. A patched pseudo-spectral method based on the Chebyshev polynomial is adopted to solve the resulting governing fundamental electrokinetic equations. Mobility reversal, among other interesting phenomena, is observed when the droplet is in a narrow cylindrical pore. No such observation was made in the corresponding motion of a rigid particle. The droplet with a thick double layer may even move against the prediction based on the Coulomb electrostatic law, for instance, a positively charged droplet may move against the electric field. The significant enhancement of the motion-deterring double layer polarization due to the severe steric boundary confinement within a narrow cylindrical pore is found to be responsible for this seemingly peculiar phenomenon. Moreover, smaller droplets may move in the opposite direction of the larger ones. The results are useful in capillary electrophoresis involving droplets in particular and migration of droplets through narrow channels in general.
Collapse
Affiliation(s)
- Shirley Lu
- Department of Chemical Engineering, National Taiwan University, Taipei, Taiwan
| | - Jessica Tseng
- Department of Chemical Engineering, National Taiwan University, Taipei, Taiwan
| | - Lily Chuang
- Department of Chemical Engineering, National Taiwan University, Taipei, Taiwan
| | - Nemo Chang
- Department of Chemical Engineering, National Taiwan University, Taipei, Taiwan
| | - Sunny Chen
- Department of Chemical Engineering, National Taiwan University, Taipei, Taiwan
| | - Celia Hsu
- Department of Chemical Engineering, National Taiwan University, Taipei, Taiwan
| | - Jean Chien
- Department of Chemical Engineering, National Taiwan University, Taipei, Taiwan
| | - Carol Lin
- Department of Chemical Engineering, National Taiwan University, Taipei, Taiwan
| | - Eric Lee
- Department of Chemical Engineering, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
8
|
Bhattacharya A, Chakraborty S. Modulating selective ionic enrichment and depletion zones in straight nanochannels via the interplay of surface charge modulation and electric field mediated fluid-thickening. Electrophoresis 2024; 45:752-763. [PMID: 38143284 DOI: 10.1002/elps.202300189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/08/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023]
Abstract
We report the possibilities of achieving highly controlled segregation of ion-enriched and ion-depleted regions in straight nanochannels. This is achieved via harnessing the interplay of an axial gradient of the induced transverse electric field on account of electrical double layer phenomenon and the localized thickening of the fluid because of intensified electric fields due to the large spatial gradients of the electrical potential in extreme confinements. By considering alternate surface patches of different charge densities over pre-designed axial spans, we illustrate how these effects can be exploited to realize selectively ion-enriched and ion-depleted zones. Physically, this is attributed to setting up of an axial concentration gradient that delves on the ionic advection due to the combined effect of an externally applied electric field and induced back-pressure gradient along the channel axis and electro-migration due to the combinatorial influences of the applied and the induced electrostatic fields. With an explicit handle on the pertinent parameters, our results offer insights on the possible means of imposing delicate controls on the solute-enrichment and depletion phenomena, a paradigm that remained unexplored thus far.
Collapse
Affiliation(s)
- Anindita Bhattacharya
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Suman Chakraborty
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
9
|
Bu Y, Wang J, Ni S, Lu Z, Guo Y, Yobas L. High-Performance Gel-Free and Label-Free Size Fractionation of Extracellular Vesicles with Two-Dimensional Electrophoresis in a Microfluidic Artificial Sieve. Anal Chem 2024; 96:3508-3516. [PMID: 38364051 DOI: 10.1021/acs.analchem.3c05290] [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: 02/18/2024]
Abstract
Extracellular vesicles (EVs) are cell-derived particles that exhibit diverse sizes, molecular contents, and clinical implications for various diseases depending on their specific subpopulations. However, fractionation of EV subpopulations with high resolution, efficiency, purity, and yield remains an elusive goal due to their diminutive sizes. In this study, we introduce a novel strategy that effectively separates EV subpopulations in a gel-free and label-free manner, using two-dimensional (2D) electrophoresis in a microfluidic artificial sieve. The microfabricated artificial sieve consists of periodically arranged micro-slit-well structures in a 2D array and generates an anisotropic electric field pattern to size fractionate EVs into discrete streams and steer the subpopulations into designated outlets for collection within a minute. Along with fractionating EV subpopulations, contaminants such as free proteins and short nucleic acids can be simultaneously directed to waste outlets, thus accomplishing both size fractionation and purification of EVs with high performance. Our platform offers a simple, rapid, and versatile solution for EV subpopulation isolation, which can potentially facilitate the discovery of biomarkers for specific EV subtypes and the development of EV-based therapeutics.
Collapse
Affiliation(s)
- Yang Bu
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| | - Jinhui Wang
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| | - Sheng Ni
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| | - Zechen Lu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| | - Yusong Guo
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| | - Levent Yobas
- Department of Electronic and Computer Engineering, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong SAR 999077, P. R. China
| |
Collapse
|
10
|
Ren K, Xie Y, Wang C, Yan J, Shi Y, Guo J, Guo J. Application of the fuzzy proportional integral differential (PID) temperature control algorithm in a liver function test system based on a centrifugal microfluidic device. Talanta 2024; 268:125330. [PMID: 37879203 DOI: 10.1016/j.talanta.2023.125330] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 10/27/2023]
Abstract
Clinical laboratory examinations frequently include biochemical analysis of the liver. The presence of alanine aminotransferase (ALT) and aspartate transaminase (AST) in serum can be used to identify liver damage. In this study, a centrifugal microfluidic-based clinical biochemical detection system was developed for the detection of liver function markers. Using the centrifugal microfluidic chip and centrifugal force on the chip, separation of blood cells and serum was performed. The extraction and mixing of quantitative serum and diluent were completed under the chip design of microchannels and microchambers. The lyophilized reagent beads in the chip interacted with the combined solution. The Fuzzy PID algorithm regulates the power of the heating film to deliver the ideal reaction temperature. In accordance with Beer-Lambert, the rate of change in the absorbance of the reaction solution at 340 nm of the light source was measured and a standard curve for the relationship between concentration and rate of change in absorbance was constructed. The system is portable, quick, and simple to use because it uses a centrifugal microfluidic chip instead of the conventional detection and analysis approach. In the future, it is anticipated that the system will have several applications in the detection of highly integrated on-chip point-of-care devices.
Collapse
Affiliation(s)
- Keyi Ren
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, China
| | - Yiweng Xie
- School of Computer Science, Fudan University, Shanghai, 200433, China
| | - Chuang Wang
- University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Jiasheng Yan
- University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yuxing Shi
- University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Jiuchuan Guo
- University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Jinhong Guo
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, China; University of Electronic Science and Technology of China, Chengdu, 611731, China.
| |
Collapse
|
11
|
Anand G, Safaripour S, Snoeyink C. Anomalous, dielectrophoretic transport of molecules in non-electrolytes. J Sep Sci 2024; 47:e2300719. [PMID: 38066389 DOI: 10.1002/jssc.202300719] [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: 09/27/2023] [Revised: 11/08/2023] [Accepted: 11/27/2023] [Indexed: 01/19/2024]
Abstract
The electric field (E-field) dielectric polarization-based separations mechanism represents a novel method for separating solutions at small length scales. An E-field gradient with a maximum strength of 0.4 MV/m applied across a 10 μm deep channel is shown to increase the concentration inside the low E-field region by ≈ 40% relative to the high E-field region. This concentration change is two orders of magnitude higher than the estimated change predicted using the classical equilibrium thermodynamics for the same E-field. The deviation between the predicted and the experimental results suggests that the change in volumetric E-field energy with solute concentration is insufficient to explain this phenomenon. The study also explores the effect of varying strength of E-field and frequency of supplied voltage on the dielectric polarization-based separation efficiency. While the increase in the former increases the separation efficiency, the increase in the latter reduces the degree of concentration change due to ineffective charging of the electrodes.
Collapse
Affiliation(s)
- Gaurav Anand
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, New York, USA
| | - Samira Safaripour
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, New York, USA
| | - Craig Snoeyink
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
12
|
Dortaj H, Azarpira N, Pakbaz S. Insight to Biofabrication of Liver Microtissues for Disease Modeling: Challenges and Opportunities. Curr Stem Cell Res Ther 2024; 19:1303-1311. [PMID: 37846577 DOI: 10.2174/011574888x257744231009071810] [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: 04/13/2023] [Revised: 08/26/2023] [Accepted: 09/13/2023] [Indexed: 10/18/2023]
Abstract
In the last decade, liver diseases with high mortality rates have become one of the most important health problems in the world. Organ transplantation is currently considered the most effective treatment for compensatory liver failure. An increasing number of patients and shortage of donors has led to the attention of reconstructive medicine methods researchers. The biggest challenge in the development of drugs effective in chronic liver disease is the lack of a suitable preclinical model that can mimic the microenvironment of liver problems. Organoid technology is a rapidly evolving field that enables researchers to reconstruct, evaluate, and manipulate intricate biological processes in vitro. These systems provide a biomimetic model for studying the intercellular interactions necessary for proper organ function and architecture in vivo. Liver organoids, formed by the self-assembly of hepatocytes, are microtissues and can exhibit specific liver characteristics for a long time in vitro. Hepatic organoids are identified as an impressive tool for evaluating potential cures and modeling liver diseases. Modeling various liver diseases, including tumors, fibrosis, non-alcoholic fatty liver, etc., allows the study of the effects of various drugs on these diseases in personalized medicine. Here, we summarize the literature relating to the hepatic stem cell microenvironment and the formation of liver Organoids.
Collapse
Affiliation(s)
- Hengameh Dortaj
- Department of Tissue Engineering and Applied Cell Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Pakbaz
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
13
|
Zeng Z, Tian J, Ren Z, Yang Y, Gong Q, Sun R, Zhang X, Liu W, Chen C. Digital droplet immunoassay based on a microfluidic chip with magnetic beads for the detection of prostate-specific antigen. J Sep Sci 2023; 46:e2300471. [PMID: 37905470 DOI: 10.1002/jssc.202300471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 11/02/2023]
Abstract
Sensitive biomarker detection techniques are beneficial for both disease diagnosis and postoperative examinations. In this study, we report an integrated microfluidic chip designed for the immunodetection of prostate-specific antigens (PSAs). The microfluidic chip is based on the three-dimensional structure of quartz capillaries. The outlet channel extends to 1.8 cm, effectively facilitating the generation of uniform droplets ranging in size from 3 to 50 μm. Furthermore, we successfully immobilized the captured antibodies onto the surface of magnetic beads using an activator, and we constructed an immunosandwich complex by employing biotinylated antibodies. A key feature of this microfluidic chip is its integration of microfluidic droplet technology advantages, such as high-throughput parallelism, enzymatic signal amplification, and small droplet size. This integration results in an exceptionally sensitive PSA detection capability, with the detection limit reduced to 7.00 ± 0.62 pg/mL.
Collapse
Affiliation(s)
- Zhaokui Zeng
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Jingjing Tian
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zixuan Ren
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Ying Yang
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Qian Gong
- Department of Pharmacy, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruowei Sun
- Hunan Zaochen Nanorobot Co.Ltd, Liuyang, China
| | - Xun Zhang
- Hunan Zaochen Nanorobot Co.Ltd, Liuyang, China
| | - Wenfang Liu
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Chuanpin Chen
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|
14
|
Mai Z, Xiao S, Zhang W, Wang K. A multi-ion interference decoupling model based on ion-selective electrode arrays. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:5038-5049. [PMID: 37740373 DOI: 10.1039/d3ay00888f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
Compared to traditional liquid-junction ion-selective electrodes, solid-contact ion-selective electrodes (SC-ISEs) have attracted much attention and undergone rapid development due to their compactness and ease of integration. However, the application and widespread use of SC-ISEs are limited by their non-ideal selectivity and susceptibility to signal drift. Although the principles of artificial neural network (ANN) methods have shown significant progress in partially resolving the selectivity issue, they generally require extensive calibration steps and computational resources to implement. As a result, numerical computation models are more practical and economical, but existing approaches often overlook experimental phenomena and have relatively complex modeling principles. In this study, we propose a proportional factor model based on the trend of SC-ISEs affected by the multiple ions, along with a scalable dynamic correction procedure to improve its robustness. This model utilizes an estimated response surface method to solve nonlinear equations, requiring fewer calibration experiments. It accurately extracts the concentrations of multiple target ions in the presence of multi-ion interference and dynamically adjusts the model parameters for different types of ISEs. Additionally, we design a multi-channel SC-ISE array as a carrier for theoretical validation. In a case study, we demonstrate the feasibility of decoupling multiple ions using the SC-ISE array, obtaining concentrations of calcium, magnesium, sodium, and potassium ions, and verify the accuracy of the multi-ion detection system for real-world water samples.
Collapse
Affiliation(s)
- Zhancheng Mai
- School of Electronics and Information Technology, Sun Yat sen University, Panyu District, Guangzhou City, Guangdong Province, China.
| | - Shaoqiu Xiao
- School of Electronics and Information Technology, Sun Yat sen University, Panyu District, Guangzhou City, Guangdong Province, China.
| | - Wei Zhang
- Guangke Chipwey Sensing Technologies Co., Ltd, Foshan, China
| | - Kai Wang
- School of Electronics and Information Technology, Sun Yat sen University, Panyu District, Guangzhou City, Guangdong Province, China.
| |
Collapse
|
15
|
Sena-Torralba A, Banguera-Ordoñez YD, Mira-Pascual L, Maquieira Á, Morais S. Exploring the potential of paper-based electrokinetic phenomena in PoC biosensing. Trends Biotechnol 2023; 41:1299-1313. [PMID: 37150668 DOI: 10.1016/j.tibtech.2023.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 05/09/2023]
Abstract
In order to decentralize health care, the development of point-of-care (PoC) assays has gained significant attention in recent decades. The lateral flow immunoassay (LFIA) has emerged as a promising bioanalytical method due to its low cost and single-step detection process. However, its limited sensitivity and inability to detect disease biomarkers at clinically relevant levels have hindered its application for early diagnosis. This review explores the potential of merging different electrokinetic phenomena into paper-based assays to enhance their analytical performance, offering a versatile and affordable approach for PoC testing. The review exposes the challenges faced in integrating electrokinetic phenomena with paper-based biosensing and concludes by discussing the issues that need to be improved to maximize the potential of this technology for early diagnosis.
Collapse
Affiliation(s)
- Amadeo Sena-Torralba
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camino de Vera s/n, 46022, Valencia, Spain; Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
| | - Yulieth D Banguera-Ordoñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camino de Vera s/n, 46022, Valencia, Spain
| | - Laia Mira-Pascual
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camino de Vera s/n, 46022, Valencia, Spain
| | - Ángel Maquieira
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camino de Vera s/n, 46022, Valencia, Spain; Departamento de Química, Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain
| | - Sergi Morais
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Camino de Vera s/n, 46022, Valencia, Spain; Departamento de Química, Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain.
| |
Collapse
|
16
|
Aghajanloo B, Ejeian F, Frascella F, Marasso SL, Cocuzza M, Tehrani AF, Nasr Esfahani MH, Inglis DW. Pumpless deterministic lateral displacement separation using a paper capillary wick. LAB ON A CHIP 2023; 23:2106-2112. [PMID: 36943724 DOI: 10.1039/d3lc00039g] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Deterministic lateral displacement (DLD) is a passive separation method that separates particles by hydrodynamic size. This label-free method is a promising technique for cell separation because of its high size resolution and insensitivity to flow rate. Development of capillary-driven microfluidic technologies allows microfluidic devices to be operated without any external power for fluid pumping, lowering their total cost and complexity. Herein, we develop and test a DLD-based particle and cell sorting method that is driven entirely by capillary pressure. We show microchip self-filling, flow focusing, flow stability, and capture of separated particles. We achieve separation efficiency of 92% for particle-particle separation and more than 99% efficiency for cell-particle separation. The high performance of driven flow and separation along with simplicity of the operation and setup make it a valuable candidate for point-of-care devices.
Collapse
Affiliation(s)
- Behrouz Aghajanloo
- Department of Mechanical Engineering, Isfahan University of Technology, Isfahan, Iran
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- DISAT, Politecnico di Torino, Turin, Italy
- School of Engineering, Macquarie University, Sydney, Australia.
| | - Fatemeh Ejeian
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | | | - Simone L Marasso
- DISAT, Politecnico di Torino, Turin, Italy
- CNR-IMEM, Parma, Italy
| | - Matteo Cocuzza
- DISAT, Politecnico di Torino, Turin, Italy
- CNR-IMEM, Parma, Italy
| | | | - Mohammad Hossein Nasr Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - David W Inglis
- School of Engineering, Macquarie University, Sydney, Australia.
| |
Collapse
|
17
|
Sharbati P, Sadaghiani AK, Koşar A. New Generation Dielectrophoretic-Based Microfluidic Device for Multi-Type Cell Separation. BIOSENSORS 2023; 13:bios13040418. [PMID: 37185493 PMCID: PMC10135750 DOI: 10.3390/bios13040418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/11/2023] [Accepted: 03/22/2023] [Indexed: 05/17/2023]
Abstract
This study introduces a new generation of dielectrophoretic-based microfluidic device for the precise separation of multiple particle/cell types. The device features two sets of 3D electrodes, namely cylindrical and sidewall electrodes. The main channel of the device terminates with three outlets: one in the middle for particles that sense negative dielectrophoresis force and two others at the right and left sides for particles that sense positive dielectrophoresis force. To evaluate the device performance, we used red blood cells (RBCs), T-cells, U937-MC cells, and Clostridium difficile bacteria as our test subjects. Our results demonstrate that the proposed microfluidic device could accurately separate bioparticles in two steps, with sidewall electrodes of 200 µm proving optimal for efficient separation. Applying different voltages for each separation step, we found that the device performed most effectively at 6 Vp-p applied to the 3D electrodes, and at 20 Vp-p and 11 Vp-p applied to the sidewall electrodes for separating RBCs from bacteria and T-cells from U937-MC cells, respectively. Notably, the device's maximum electric fields remained below the cell electroporation threshold, and we achieved a separation efficiency of 95.5% for multi-type particle separation. Our findings proved the device's capacity for separating multiple particle types with high accuracy, without limitation for particle variety.
Collapse
Affiliation(s)
- Pouya Sharbati
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
- Sabanci University Nanotechnology and Applications Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
| | - Abdolali K Sadaghiani
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
- Sabanci University Nanotechnology and Applications Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
| | - Ali Koşar
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
- Sabanci University Nanotechnology and Applications Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
| |
Collapse
|
18
|
Hettiarachchi S, Cha H, Ouyang L, Mudugamuwa A, An H, Kijanka G, Kashaninejad N, Nguyen NT, Zhang J. Recent microfluidic advances in submicron to nanoparticle manipulation and separation. LAB ON A CHIP 2023; 23:982-1010. [PMID: 36367456 DOI: 10.1039/d2lc00793b] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Manipulation and separation of submicron and nanoparticles are indispensable in many chemical, biological, medical, and environmental applications. Conventional technologies such as ultracentrifugation, ultrafiltration, size exclusion chromatography, precipitation and immunoaffinity capture are limited by high cost, low resolution, low purity or the risk of damage to biological particles. Microfluidics can accurately control fluid flow in channels with dimensions of tens of micrometres. Rapid microfluidics advancement has enabled precise sorting and isolating of nanoparticles with better resolution and efficiency than conventional technologies. This paper comprehensively studies the latest progress in microfluidic technology for submicron and nanoparticle manipulation. We first summarise the principles of the traditional techniques for manipulating nanoparticles. Following the classification of microfluidic techniques as active, passive, and hybrid approaches, we elaborate on the physics, device design, working mechanism and applications of each technique. We also compare the merits and demerits of different microfluidic techniques and benchmark them with conventional technologies. Concurrently, we summarise seven standard post-separation detection techniques for nanoparticles. Finally, we discuss current challenges and future perspectives on microfluidic technology for nanoparticle manipulation and separation.
Collapse
Affiliation(s)
- Samith Hettiarachchi
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Haotian Cha
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Lingxi Ouyang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | | | - Hongjie An
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Gregor Kijanka
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Navid Kashaninejad
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| | - Jun Zhang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland 4111, Australia.
| |
Collapse
|
19
|
Lambiase G, Klottrup-Rees K, Lovelady C, Ali S, Shepherd S, Muroni M, Lindo V, James DC, Dickman MJ. An automated, low volume, and high-throughput analytical platform for aggregate quantitation from cell culture media. J Chromatogr A 2023; 1691:463809. [PMID: 36731329 DOI: 10.1016/j.chroma.2023.463809] [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: 10/07/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
High throughput screening methods have driven a paradigm shift in biopharmaceutical development by reducing the costs of good manufactured (COGM) and accelerate the launch to market of novel drug products. Scale-down cell culture systems such as shaken 24- and 96-deep-well plates (DWPs) are used for initial screening of hundreds of recombinant mammalian clonal cell lines to quickly and efficiently select the best producing strains expressing product quality attributes that fit to industry platform. A common modification monitored from early-stage product development is protein aggregation due to its impact on safety and efficacy. This study aims to integrate high-throughput analysis of aggregation-prone therapeutic proteins with 96-deep well plate screening to rank clones based on the aggregation levels of the expressed proteins. Here we present an automated, small-scale analytical platform workflow combining the purification and subsequent aggregation analysis of protein biopharmaceuticals expressed in 96-DWP cell cultures. Product purification was achieved by small-scale solid-phase extraction using dual flow chromatography (DFC) automated on a robotic liquid handler for the parallel processing of up to 96 samples at a time. At-line coupling of size-exclusion chromatography (SEC) using a 2.1 mm ID column enabled the detection of aggregates with sub-2 µg sensitivity and a 3.5 min run time. The entire workflow was designed as an application to aggregation-prone mAbs and "mAb-like" next generation biopharmaceuticals, such as bispecific antibodies (BsAbs). Application of the high-throughput analytical workflow to a shake plate overgrow (SPOG) screen, enabled the screening of 384 different clonal cell lines in 32 h, requiring < 2 μg of protein per sample. Aggregation levels expressed by the clones varied between 9 and 76%. This high-throughput analytical workflow allowed for the early elimination of clonal cell lines with high aggregation, demonstrating the advantage of integrating analytical testing for critical quality attributes (CQAs) earlier in product development to drive better decision making.
Collapse
Affiliation(s)
- Giulia Lambiase
- Department of Chemical and Biological Engineering, University of Sheffield, Mappin St., Sheffield, UK; Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Kerensa Klottrup-Rees
- Cell Culture and Fermentation Sciences, BioPharmaceuticals Development, AstraZeneca, Cambridge, UK
| | - Clare Lovelady
- Cell Culture and Fermentation Sciences, BioPharmaceuticals Development, AstraZeneca, Cambridge, UK
| | - Salma Ali
- Cell Culture and Fermentation Sciences, BioPharmaceuticals Development, AstraZeneca, Cambridge, UK
| | - Samuel Shepherd
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Maurizio Muroni
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Vivian Lindo
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK.
| | - David C James
- Department of Chemical and Biological Engineering, University of Sheffield, Mappin St., Sheffield, UK.
| | - Mark J Dickman
- Department of Chemical and Biological Engineering, University of Sheffield, Mappin St., Sheffield, UK.
| |
Collapse
|
20
|
Li Z, Liu H, Wang D, Zhang M, Yang Y, Ren TL. Recent advances in microfluidic sensors for nutrients detection in water. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2022.116790] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
21
|
Naghdi E, Moran GE, Reinau ME, De Malsche W, Neusüß C. Concepts and recent advances in microchip electrophoresis coupled to mass spectrometry: Technologies and applications. Electrophoresis 2023; 44:246-267. [PMID: 35977423 DOI: 10.1002/elps.202200179] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/11/2022] [Accepted: 08/13/2022] [Indexed: 02/01/2023]
Abstract
The online coupling of microchip electrophoresis (ME) as a fast, highly efficient, and low-cost miniaturized separation technique to mass spectrometry (MS) as an information-rich and sensitive characterization technique results in ME-MS an attractive tool for various applications. In this paper, we review the basic concepts and latest advances in technology for ME coupled to MS during the period of 2016-2021, covering microchip materials, structures, fabrication techniques, and interfacing to electrospray ionization (ESI)-MS and matrix-assisted laser desorption/ionization-MS. Two critical issues in coupling ME and ESI-MS include the electrical connection used to define the electrophoretic field strength along the separation channel and the generation of the electrospray for MS detection, as well as, a miniaturized ESI-tip. The recent commercialization of ME-MS in zone electrophoresis and isoelectric focusing modes has led to the widespread application of these techniques in academia and industry. Here we summarize recent applications of ME-MS for the separation and detection of antibodies, proteins, peptides, carbohydrates, metabolites, and so on. Throughout the paper these applications are discussed in the context of benefits and limitations of ME-MS in comparison to alternative techniques.
Collapse
Affiliation(s)
- Elahe Naghdi
- Department of Chemistry, Aalen University, Aalen, Germany
| | - Griffin E Moran
- Novo Nordisk A/S, Global Research Technologies, Maaloev, Denmark
| | | | - Wim De Malsche
- µFlow group, Department of Chemical Engineering, Vrije Universiteit Brussel, Brussels, Belgium
| | | |
Collapse
|
22
|
Shao X, Huang Y, Wang G. Microfluidic devices for protein analysis using intact and top‐down mass spectrometry. VIEW 2022. [DOI: 10.1002/viw.20220032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Xinyang Shao
- Institute for Cell Analysis Shenzhen Bay Laboratory Shenzhen China
- Biomedical Pioneering Innovation Center Peking University Beijing China
- Peking‐Tsinghua Center for Life Sciences Peking University Beijing China
| | - Yanyi Huang
- Institute for Cell Analysis Shenzhen Bay Laboratory Shenzhen China
- Biomedical Pioneering Innovation Center Peking University Beijing China
- Peking‐Tsinghua Center for Life Sciences Peking University Beijing China
- College of Chemistry and Molecular Engineering and Beijing National Laboratory for Molecular Sciences Peking University Beijing China
| | - Guanbo Wang
- Institute for Cell Analysis Shenzhen Bay Laboratory Shenzhen China
- Biomedical Pioneering Innovation Center Peking University Beijing China
| |
Collapse
|
23
|
Kim H, Zhbanov A, Yang S. Microfluidic Systems for Blood and Blood Cell Characterization. BIOSENSORS 2022; 13:13. [PMID: 36671848 PMCID: PMC9856090 DOI: 10.3390/bios13010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 06/17/2023]
Abstract
A laboratory blood test is vital for assessing a patient's health and disease status. Advances in microfluidic technology have opened the door for on-chip blood analysis. Currently, microfluidic devices can reproduce myriad routine laboratory blood tests. Considerable progress has been made in microfluidic cytometry, blood cell separation, and characterization. Along with the usual clinical parameters, microfluidics makes it possible to determine the physical properties of blood and blood cells. We review recent advances in microfluidic systems for measuring the physical properties and biophysical characteristics of blood and blood cells. Added emphasis is placed on multifunctional platforms that combine several microfluidic technologies for effective cell characterization. The combination of hydrodynamic, optical, electromagnetic, and/or acoustic methods in a microfluidic device facilitates the precise determination of various physical properties of blood and blood cells. We analyzed the physical quantities that are measured by microfluidic devices and the parameters that are determined through these measurements. We discuss unexplored problems and present our perspectives on the long-term challenges and trends associated with the application of microfluidics in clinical laboratories. We expect the characterization of the physical properties of blood and blood cells in a microfluidic environment to be considered a standard blood test in the future.
Collapse
Affiliation(s)
- Hojin Kim
- Department of Mechatronics Engineering, Dongseo University, Busan 47011, Republic of Korea
| | - Alexander Zhbanov
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Sung Yang
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
24
|
Qing LS, Wang TT, Luo HY, Du JL, Wang RY, Luo P. Microfluidic strategies for natural products in drug discovery: Current status and future perspectives. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Zhang S, Deng J, Li J, Tian F, Liu C, Fang L, Sun J. Advanced microfluidic technologies for isolating extracellular vesicles. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
26
|
Wang Y, Weng C, Sun H, Deng Z, Jiang B. Effect of Interfacial Interaction on the Demolding Deformation of Injection Molded Microfluidic Chips. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3416. [PMID: 36234545 PMCID: PMC9565601 DOI: 10.3390/nano12193416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 06/16/2023]
Abstract
During the demolding process, the interfacial interaction between the polymer and the metal mold insert will lead to the deformation of the micro-structure, which will directly affect the molding quality and performance of injection molded microfluidic chips. In this study, the demolding quality of micro-channels and micro-mixing structures of polycarbonate (PC), polymethyl methacrylate (PMMA), cyclic olefin copolymer (COC), and polystyrene (PS) microfluidic chips for heavy metal detection were investigated by molding experiments. The experimental results showed that the structures of microfluidic chips could be completely replicated. However, tensile deformation and fracture defects were observed at the edges of the micro-structures after demolding. Compared to the Ni mold insert, the calculation of the relative deviation percentages showed that the width of the micro-channel became larger and the depth became smaller, while the dimensions of the micro-mixing structure changes in the opposite direction. Subsequently, a molecular dynamics (MD) simulation model of polymer/nickel (Ni) mold insert for injection molding was established. The changes of adhesion work, demolding resistance and potential energy during demolding were analyzed. The simulation results showed that the polymer structures had some deformations such as necking, molecular chain stretching and voids under the action of adhesion work and demolding resistance. The difference in the contact area with the mold insert directly brought different interfacial interactions. In addition, the potential energy change of the polymer system could be used to quantitatively characterize the demolding deformation of the structure. Overall, the MD method is able to effectively explain the internal mechanisms of interfacial interactions, leading to the demolding deformation of polymer structures from the molecular/atomic scale.
Collapse
Affiliation(s)
- Yilei Wang
- College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Can Weng
- College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
- State Key Laboratory of High Performance Complex Manufacturing, Central South University, Changsha 410083, China
| | - Huijie Sun
- College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Zijian Deng
- College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Bingyan Jiang
- College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
- State Key Laboratory of High Performance Complex Manufacturing, Central South University, Changsha 410083, China
| |
Collapse
|
27
|
Van Volkenburg T, Benzing JS, Craft KL, Ohiri K, Kilhefner A, Irons K, Bradburne C. Microfluidic Chromatography for Enhanced Amino Acid Detection at Ocean Worlds. ASTROBIOLOGY 2022; 22:1116-1128. [PMID: 35984944 PMCID: PMC9508454 DOI: 10.1089/ast.2021.0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/18/2022] [Indexed: 06/15/2023]
Abstract
Increasing interest in the detection of biogenic signatures, such as amino acids, on icy moons and bodies within our solar system has led to the development of compact in situ instruments. Given the expected dilute biosignatures and high salinities of these extreme environments, purification of icy samples before analysis enables increased detection sensitivity. Herein, we outline a novel compact cation exchange method to desalinate proteinogenic amino acids in solution, independent of the type and concentration of salts in the sample. Using a modular microfluidic device, initial experiments explored operational limits of binding capacity with phenylalanine and three model cations, Na+, Mg2+, and Ca2+. Phenylalanine recovery (94-17%) with reduced conductivity (30-200 times) was seen at high salt-to-amino-acid ratios between 25:1 and 500:1. Later experiments tested competition between mixtures of 17 amino acids and other chemistries present in a terrestrial ocean sample. Recoveries ranged from 11% to 85% depending on side chain chemistry and cation competition, with concentration shown for select high affinity amino acids. This work outlines a nondestructive amino acid purification device capable of coupling to multiple downstream analytical techniques for improved characterization of icy samples at remote ocean worlds.
Collapse
Affiliation(s)
| | | | - Kathleen L. Craft
- Johns Hopkins University Applied Physics Laboratory, Laurel, Maryland, USA
| | - Korine Ohiri
- Johns Hopkins University Applied Physics Laboratory, Laurel, Maryland, USA
| | - Ashley Kilhefner
- Johns Hopkins University Applied Physics Laboratory, Laurel, Maryland, USA
| | - Kristen Irons
- University of North Carolina at Chapel Hill College of Arts and Sciences, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
28
|
Pinheiro KMP, Duarte LM, Rodrigues MF, Vaz BG, Junior IM, Carvalho RM, Coltro WKT. Determination of naphthenic acids in produced water by using microchip electrophoresis with integrated contactless conductivity detection. J Chromatogr A 2022; 1677:463307. [PMID: 35834889 DOI: 10.1016/j.chroma.2022.463307] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/16/2022] [Accepted: 07/04/2022] [Indexed: 10/17/2022]
Abstract
This study reports for the first time the use of a microchip electrophoresis (ME) device with integrated capacitively coupled contactless conductivity detection (C4D) to analyze naphthenic acids in produced water. A mixture containing 9-anthracenecarboxylic, 1-naphthoic, and benzoic acids was separated and detected using a running buffer composed of 10 mmol L-1 carbonate buffer (pH = 10.2). The separation was achieved within ca. 140 s with baseline resolution greater than 2 and efficiency values ranging from 1.9 × 105 to 2.4 × 105 plates m-1. The developed methodology provided linear correlation with determination coefficients greater than 0.992 for the concentration ranges between 50 and 250 µmol L-1 for benzoic and 9-anthracenecarboxylic acids, and between 50 and 200 µmol L-1 for 1-naphthoic acid. The achieved limit of detection values varied between 4.7 and 7.7 µmol L-1. The proposed methodology revealed satisfactory repeatability with RSD values for a sequence of eight injections between 5.5 and 7.7% for peak areas and lower than 1% for migration times. In addition, inter-day precision was evaluated for sixteen injections (a sequence of four injections performed during four days), and the RSD values were lower than 11.5 and 4.9% for peak areas and migration time, respectively. Five produced water samples were analyzed, and it was possible to detect and quantify 9-anthracenecarboxylic acid. The concentrations ranged from 1.05 to 2.24 mmol L-1 with recovery values between 90.8 and 96.0%. ME-C4D demonstrated satisfactory analytical performance for determining naphthenic acids in produced water for the first time, which is useful for petroleum or oil industry investigation.
Collapse
Affiliation(s)
- Kemilly M P Pinheiro
- Instituto de Química, Universidade Federal de Goiás, Goiânia, GO 74690-900, Brazil
| | - Lucas M Duarte
- Instituto de Química, Universidade Federal de Goiás, Goiânia, GO 74690-900, Brazil; Instituto de Química, Universidade Federal Fluminense, Niterói, RJ 24020-141, Brazil
| | - Marcella F Rodrigues
- Instituto de Química, Universidade Federal de Goiás, Goiânia, GO 74690-900, Brazil
| | - Boniek G Vaz
- Instituto de Química, Universidade Federal de Goiás, Goiânia, GO 74690-900, Brazil
| | - Iris Medeiros Junior
- Centro de Pesquisas e Desenvolvimento Leopoldo Américo Miguez de Mello (CENPES), Rio de Janeiro, RJ 21040-000, Brazil
| | - Rogerio M Carvalho
- Centro de Pesquisas e Desenvolvimento Leopoldo Américo Miguez de Mello (CENPES), Rio de Janeiro, RJ 21040-000, Brazil
| | - Wendell K T Coltro
- Instituto de Química, Universidade Federal de Goiás, Goiânia, GO 74690-900, Brazil; Instituto Nacional de Ciência e Tecnologia de Bioanalítica, Campinas, SP 13084-971, Brazil.
| |
Collapse
|
29
|
Wang B, Park B. Microfluidic Sampling and Biosensing Systems for Foodborne Escherichia coli and Salmonella. Foodborne Pathog Dis 2022; 19:359-375. [PMID: 35713922 DOI: 10.1089/fpd.2021.0087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Developments of portable biosensors for field-deployable detections have been increasingly important to control foodborne pathogens in regulatory environment and in early stage of outbreaks. Conventional cultivation and gene amplification methods require sophisticated instruments and highly skilled professionals; while portable biosensing devices provide more freedom for rapid detections not only in research laboratories but also in the field; however, their sensitivity and specificity are limited. Microfluidic methods have the advantage of miniaturizing instrumental size while integrating multiple functions and high-throughput capability into one streamlined system at low cost. Minimal sample consumption is another advantage to detect samples in different sizes and concentrations, which is important for the close monitoring of pathogens at consumer end. They improve measurement or manipulation of bacteria by increasing the ratio of functional interface of the device to the targeted biospecies and in turn reducing background interference. This article introduces the major active and passive microfluidic devices that have been used for bacteria sampling and biosensing. The emphasis is on particle-based sorting/enrichment methods with or without external physical fields applied to the microfluidic devices and on various biosensing applications reported for bacteria sampling. Three major fabrication methods for microfluidics are briefly discussed with their advantages and limitations. The applications of these active and passive microfluidic sampling methods in the past 5 years have been summarized, with the focus on Escherichia coli and Salmonella. The current challenges to microfluidic bacteria sampling are caused by the small size and nonspherical shape of various bacterial cells, which can induce unpredictable deviations in sampling and biosensing processes. Future studies are needed to develop rapid prototyping methods for device manufacturing, which can facilitate rapid response to various foodborne pathogen outbreaks.
Collapse
Affiliation(s)
- Bin Wang
- U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, Georgia, USA
| | - Bosoon Park
- U.S. National Poultry Research Center, Agricultural Research Service, U.S. Department of Agriculture, Athens, Georgia, USA
| |
Collapse
|
30
|
Li W, Chaihu L, Jiang J, Wu B, Zheng X, Dai R, Tian Y, Huang Y, Wang G, Men Y. Microfluidic Platform for Time-Resolved Characterization of Protein Higher-Order Structures and Dynamics Using Top-Down Mass Spectrometry. Anal Chem 2022; 94:7520-7527. [PMID: 35584038 DOI: 10.1021/acs.analchem.2c00077] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Characterization of protein higher-order structures and dynamics is essential for understanding the biological functions of proteins and revealing the underlying mechanisms. Top-down mass spectrometry (MS) accesses structural information at both the intact protein level and the peptide fragment level. Native top-down MS allows analysis of a protein complex's architecture and subunits' identity and modifications. Top-down hydrogen/deuterium exchange (HDX) MS offers high spatial resolution for conformational or binding interface analysis and enables conformer-specific characterization. A microfluidic chip can provide superior performance for front-end reactions useful for these MS workflows, such as flexibility in manipulating multiple reactant flows, integrating various functional modules, and automation. However, most microchip-MS devices are designed for bottom-up approaches or top-down proteomics. Here, we demonstrate a strategy for designing a microchip for top-down MS analysis of protein higher-order structures and dynamics. It is suitable for time-resolved native MS and HDX MS, with designs aiming for efficient ionization of intact protein complexes, flexible manipulation of multiple reactant flows, and precise control of reaction times over a broad range of flow rates on the submicroliter per minute scale. The performance of the prototype device is demonstrated by measurements of systems including monoclonal antibodies, antibody-antigen complexes, and coexisting protein conformers. This strategy may benefit elaborate structural analysis of biomacromolecules and inspire method development using the microchip-MS approach.
Collapse
Affiliation(s)
- Wen Li
- Research Center for Biomedical Optics and Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lingxiao Chaihu
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.,Institute of Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Jialu Jiang
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Bizhu Wu
- Research Center for Biomedical Optics and Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xuan Zheng
- Research Center for Biomedical Optics and Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Rongrong Dai
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ye Tian
- Institute of Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yanyi Huang
- Institute of Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China.,Biomedical Pioneering Innovation Centre, Peking University, Beijing 100871, China
| | - Guanbo Wang
- Institute of Cell Analysis, Shenzhen Bay Laboratory, Shenzhen 518132, China.,Biomedical Pioneering Innovation Centre, Peking University, Beijing 100871, China
| | - Yongfan Men
- Research Center for Biomedical Optics and Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
31
|
Ezrre S, Reyna MA, Anguiano C, Avitia RL, Márquez H. Lab-on-a-Chip Platforms for Airborne Particulate Matter Applications: A Review of Current Perspectives. BIOSENSORS 2022; 12:191. [PMID: 35448251 PMCID: PMC9024784 DOI: 10.3390/bios12040191] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
Lab-on-a-Chip (LoC) devices are described as versatile, fast, accurate, and low-cost platforms for the handling, detection, characterization, and analysis of a wide range of suspended particles in water-based environments. However, for gas-based applications, particularly in atmospheric aerosols science, LoC platforms are rarely developed. This review summarizes emerging LoC devices for the classification, measurement, and identification of airborne particles, especially those known as Particulate Matter (PM), which are linked to increased morbidity and mortality levels from cardiovascular and respiratory diseases. For these devices, their operating principles and performance parameters are introduced and compared while highlighting their advantages and disadvantages. Discussing the current applications will allow us to identify challenges and determine future directions for developing more robust LoC devices to monitor and analyze airborne PM.
Collapse
Affiliation(s)
- Sharon Ezrre
- Instituto de Ingeniería, Universidad Autónoma de Baja California (UABC), Mexicali 21100, Mexico;
| | - Marco A. Reyna
- Instituto de Ingeniería, Universidad Autónoma de Baja California (UABC), Mexicali 21100, Mexico;
| | - Citlalli Anguiano
- Facultad de Ingeniería, Universidad Autónoma de Baja California (UABC), Mexicali 21280, Mexico; (C.A.); (R.L.A.)
| | - Roberto L. Avitia
- Facultad de Ingeniería, Universidad Autónoma de Baja California (UABC), Mexicali 21280, Mexico; (C.A.); (R.L.A.)
| | - Heriberto Márquez
- Departamento de Óptica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Ensenada 22860, Mexico;
| |
Collapse
|
32
|
Mikkonen S, Josefsson L, Mäkinen MEL, Chotteau V, Emmer Å. Capillary and microchip electrophoresis method development for amino acid monitoring during biopharmaceutical cultivation. Biotechnol J 2022; 17:e2100325. [PMID: 35320618 DOI: 10.1002/biot.202100325] [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: 06/28/2021] [Revised: 02/19/2022] [Accepted: 03/19/2022] [Indexed: 11/12/2022]
Abstract
The increased use of biopharmaceuticals calls for improved means of bioprocess monitoring. In this work, capillary electrophoresis (CE) and microchip electrophoresis (MCE) methods were developed and applied for the analysis of amino acids (AAs) in cell culture supernatant. In samples from different days of a Chinese hamster ovary cell cultivation process, all 19 proteinogenic AAs containing primary amine groups could be detected using CE, and 17 out of 19 AAs using MCE. The relative concentration changes in different samples agreed well with those measured by high-performance liquid chromatography (HPLC). Compared to the more commonly employed HPLC analysis, the CE and MCE methods resulted in faster analysis, while significantly lowering both the sample and reagent consumption, and the cost per analysis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Saara Mikkonen
- KTH Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Chemistry, Division of Applied Physical Chemistry, Stockholm, Sweden
| | - Leila Josefsson
- KTH Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Chemistry, Division of Applied Physical Chemistry, Stockholm, Sweden
| | - Meeri E-L Mäkinen
- KTH Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Industrial Biotechnology, Stockholm, Sweden
| | - Veronique Chotteau
- KTH Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Industrial Biotechnology, Stockholm, Sweden.,AdBIOPRO, Competence Centre for Advanced BioProduction by Continuous Processing, KTH, Stockholm, Sweden
| | - Åsa Emmer
- KTH Royal Institute of Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Chemistry, Division of Applied Physical Chemistry, Stockholm, Sweden
| |
Collapse
|
33
|
A Novel Planar Grounded Capacitively Coupled Contactless Conductivity Detector for Microchip Electrophoresis. MICROMACHINES 2022; 13:mi13030394. [PMID: 35334684 PMCID: PMC8953769 DOI: 10.3390/mi13030394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/19/2022] [Accepted: 02/25/2022] [Indexed: 11/30/2022]
Abstract
In the microchip electrophoresis with capacitively coupled contactless conductivity detection, the stray capacitance of the detector causes high background noise, which seriously affects the sensitivity and stability of the detection system. To reduce the effect, a novel design of planar grounded capacitively coupled contactless conductivity detector (PG-C4D) based on printed circuit board (PCB) is proposed. The entire circuit plane except the sensing electrodes is covered by the ground electrode, greatly reducing the stray capacitance. The efficacy of the design has been verified by the electrical field simulation and the electrophoresis detection experiments of inorganic ions. The baseline intensity of the PG-C4D was less than 1/6 of that of the traditional C4D. The PG-C4D with the new design also demonstrated a good repeatability of migration time, peak area, and peak height (n = 5, relative standard deviation, RSD ≤ 0.3%, 3%, and 4%, respectively), and good linear coefficients within the range of 0.05–0.75 mM (R2 ≥ 0.986). The detection sensitivity of K+, Na+, and Li+ reached 0.05, 0.1, and 0.1 mM respectively. Those results prove that the new design is an effective and economical approach which can improve sensitivity and repeatability of a PCB based PG-C4D, which indicate a great application potential in agricultural and environmental monitoring.
Collapse
|
34
|
Verma N, Walia S, Pandya A. Micro/nanofluidic devices for DNA/RNA detection and separation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 186:85-107. [PMID: 35033291 DOI: 10.1016/bs.pmbts.2021.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The development and research have ramped up at a greater speed than ever in the field of diseases diagnosis. Still there is struggle in developing early detection techniques which uses complex biomolecules like RNA, DNA and proteins in order to detect diseases caused by bacteria, viruses or fungi. Until now separation techniques used before detection rely on traditional techniques like electrophoresis etc. which often require centralized services. Although efforts are made in developing devices that is capable enough on carrying out separation and detection based on microfluidic (MF) and nanofluidic (NF) or lab on chip. Hence, in this chapter, we have discussed about the advancement, limitations and future steps that needs to be taken to flourish the field of NF and MF for the detection and separation of nucleic acid.
Collapse
Affiliation(s)
- Nidhi Verma
- Department of Engineering and Physical Sciences, Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Sakshi Walia
- Department of Biological Sciences and Biotechnology, Institute of Advanced Research, Gandhinagar, India
| | - Alok Pandya
- Department of Engineering and Physical Sciences, Institute of Advanced Research, Gandhinagar, Gujarat, India.
| |
Collapse
|
35
|
Jia X, Yang X, Luo G, Liang Q. Recent progress of microfluidic technology for pharmaceutical analysis. J Pharm Biomed Anal 2021; 209:114534. [PMID: 34929566 DOI: 10.1016/j.jpba.2021.114534] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022]
Abstract
In recent years, the progress of microfluidic technology has provided new tools for pharmaceutical analysis and the proposal of pharm-lab-on-a-chip is appealing for its great potential to integrate pharmaceutical test and pharmacological test in a single chip system. Here, we summarize and highlight recent advances of chip-based principles, techniques and devices for pharmaceutical test and pharmacological/toxicological test focusing on the separation and analysis of drug molecules on a chip and the construction of pharmacological models on a chip as well as their demonstrative applications in quality control, drug screening and precision medicine. The trend and challenge of microfluidic technology for pharmaceutical analysis are also discussed and prospected. We hope this review would update the insight and development of pharm-lab-on-a-chip.
Collapse
Affiliation(s)
- Xiaomeng Jia
- Center for Synthetic and Systems Biology, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, PR China
| | - Xiaoping Yang
- Center for Synthetic and Systems Biology, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, PR China
| | - Guoan Luo
- Center for Synthetic and Systems Biology, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, PR China.
| | - Qionglin Liang
- Center for Synthetic and Systems Biology, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, PR China.
| |
Collapse
|
36
|
Tang M, Zhang C, Ta L, Tan L, Zhang M, Xu D. Fully Automatic Multi-Class Multi-Residue Analysis of Veterinary Drugs Simultaneously in an Integrated Chip-MS Platform. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:14320-14329. [PMID: 34779203 DOI: 10.1021/acs.jafc.1c05235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Microfluidic chip analysis has great potential advantages such as high integration, fast speed analysis, and automatic operation and is widely used not only in biological fields but also in many other analytical areas such as agriculture and food safety. Herein, a fully automatic multi-class multi-residue analysis of veterinary drugs simultaneously in an integrated chip-mass spectrometry (chip-MS) platform was developed. The developed microfluidic chip platform integrated three modules including the extraction and filtration module, "pass-through" clean-up module, and online evaporation module. The resulting chip has been coupled to a MS detector successfully, in which 23 kinds of residues in five classes were simultaneously qualitatively and quantitatively detected without chromatographic separation, obtaining the limits of detection of the spiked milk sample in the range of 0.23-4.13 ng/mL and the recovery rate in the range from 71.7 to 118.0% under optimized conditions. The microfluidic chip system developed in this study provided a new idea for the development of detection chips and exhibited considerable potential in the point-of-care testing in milk.
Collapse
Affiliation(s)
- Minmin Tang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Jiangsu Key Laboratory of Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Chenchen Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - La Ta
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Li Tan
- NMPA Key Laboratory for Impurity Profile of Chemical Drugs, Jiangsu Institute for Food and Drug Control, Nanjing 210008, China
| | - Mei Zhang
- NMPA Key Laboratory for Impurity Profile of Chemical Drugs, Jiangsu Institute for Food and Drug Control, Nanjing 210008, China
| | - Danke Xu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
37
|
Hong T, Liu X, Zhou Q, Liu Y, Guo J, Zhou W, Tan S, Cai Z. What the Microscale Systems "See" In Biological Assemblies: Cells and Viruses? Anal Chem 2021; 94:59-74. [PMID: 34812604 DOI: 10.1021/acs.analchem.1c04244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Tingting Hong
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Xing Liu
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Qi Zhou
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Yilian Liu
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Jing Guo
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, 172 Tongzipo Road, Changsha, Hunan 410013, China
| | - Songwen Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, 172 Tongzipo Road, Changsha, Hunan 410013, China.,Jiangsu Dawning Pharmaceutical Co., Ltd., Changzhou, Jiangsu 213100, China
| | - Zhiqiang Cai
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China.,Jiangsu Dawning Pharmaceutical Co., Ltd., Changzhou, Jiangsu 213100, China
| |
Collapse
|
38
|
Hassanpour Tamrin S, Sanati Nezhad A, Sen A. Label-Free Isolation of Exosomes Using Microfluidic Technologies. ACS NANO 2021; 15:17047-17079. [PMID: 34723478 DOI: 10.1021/acsnano.1c03469] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Exosomes are cell-derived structures packaged with lipids, proteins, and nucleic acids. They exist in diverse bodily fluids and are involved in physiological and pathological processes. Although their potential for clinical application as diagnostic and therapeutic tools has been revealed, a huge bottleneck impeding the development of applications in the rapidly burgeoning field of exosome research is an inability to efficiently isolate pure exosomes from other unwanted components present in bodily fluids. To date, several approaches have been proposed and investigated for exosome separation, with the leading candidate being microfluidic technology due to its relative simplicity, cost-effectiveness, precise and fast processing at the microscale, and amenability to automation. Notably, avoiding the need for exosome labeling represents a significant advance in terms of process simplicity, time, and cost as well as protecting the biological activities of exosomes. Despite the exciting progress in microfluidic strategies for exosome isolation and the countless benefits of label-free approaches for clinical applications, current microfluidic platforms for isolation of exosomes are still facing a series of problems and challenges that prevent their use for clinical sample processing. This review focuses on the recent microfluidic platforms developed for label-free isolation of exosomes including those based on sieving, deterministic lateral displacement, field flow, and pinched flow fractionation as well as viscoelastic, acoustic, inertial, electrical, and centrifugal forces. Further, we discuss advantages and disadvantages of these strategies with highlights of current challenges and outlook of label-free microfluidics toward the clinical utility of exosomes.
Collapse
Affiliation(s)
- Sara Hassanpour Tamrin
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, CCIT 125, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
| | - Amir Sanati Nezhad
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, CCIT 125, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- Center for Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
| | - Arindom Sen
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
- Center for Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive N.W., Calgary, Alberta T2N 1N4, Canada
| |
Collapse
|
39
|
Abbasgholi N Asbaghi B, Alsadig A, Cabrera H. Online electrophoretic nanoanalysis using miniaturized gel electrophoresis and thermal lens microscopy detection. J Chromatogr A 2021; 1657:462596. [PMID: 34689905 DOI: 10.1016/j.chroma.2021.462596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 09/13/2021] [Accepted: 09/28/2021] [Indexed: 10/20/2022]
Abstract
Online thermal lens microscopy (TLM) coupled with gel electrophoresis (GE) can represent a powerful tool for separating and detecting a wide range of biomaterials. Unlike slab gel electrophoresis (SGE), the proposed method does not require prolonged procedure between separation and detection. In this work, we developed an online monitoring GE system to separate and detect nanosized materials. The design is based on a homemade and cost-effective miniaturized GE chip (MGEC) integrated with real-time TLM detection through microcontroller-based digitization board platform. To validate the feasibility and practicability of the proposed approach, we evaluated its separation capability via employing synthesized Fe3O4-Au core-shell nanoparticles (NPs) which served remarkably for the proof-of-concept. The optimum conditions for the separation process were achieved through optimization of the excitation power as 30 mW, detection position at 24 mm, the concentration of agarose gel 0.5 % w/v, and 37.5 V/cm as the effective electric field strength. The findings showed that two populations of Fe3O4-Au, core-shell, and uncapped Fe3O4 NPs, were effectively separated in less than eleven minutes, demonstrating rapid assessment of the nanomaterial production quality. Moreover, other characterization techniques such as HRTEM and EDX were employed to confirm the presence of the two dissimilar kinds of NPs separated using MGEC-TLM. The sensitivity of the method was demonstrated by determining the limit of detection (23 pM) for 10 nm AuNPs. It is envisaged that our presented system enables rapid, economical, low volume of reagents consumption and high potential analysis for quality test in various bioanalytical and nanotechnological applications.
Collapse
Affiliation(s)
| | - Ahmed Alsadig
- PhD School in Nanotechnology, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; NanoInnovation Lab, Elettra-Sincrotrone Trieste S.C.p.A., 34149 Basovizza, Trieste, Italy
| | - Humberto Cabrera
- Optics Lab, STI Unit, The Abdus Salam International Centre for Theoretical Physics, Trieste 34151, Italy.
| |
Collapse
|
40
|
Mahapatra B, Bandopadhyay A. Effect of skimming layer in an electroosmotically driven viscoelastic fluid flow over charge modulated walls. Electrophoresis 2021; 43:724-731. [PMID: 34748645 DOI: 10.1002/elps.202100221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 02/04/2023]
Abstract
We report a numerical study on the effect of the skimming layer in an EOF of Oldroyd-B fluid over charge modulated walls. Three types of flow conditions were identified on the basis of the relative thickness of the skimming layer and the electrical double layer. We observe maximum slip velocity magnitude when the skimming layer thickness is very less than the thickness of the electrical double layer. For higher skimming layer thickness compared to the thickness of electrical double layer, slip velocity magnitude attenuates, and the polymeric stress inside the skimming layer becomes zero. Enhanced fluid elasticity generates asymmetric flow structures inside the microchannel, which can also be achieved by imposing an asymmetric surface charge along the channel walls. Our present analysis highlights the complex flow dynamics of the EOF of biofluids/polymeric fluids with a near-wall region depleted of macro-molecules.
Collapse
Affiliation(s)
- Bimalendu Mahapatra
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Aditya Bandopadhyay
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| |
Collapse
|
41
|
Semi-Automatic Lab-on-PCB System for Agarose Gel Preparation and Electrophoresis for Biomedical Applications. MICROMACHINES 2021; 12:mi12091071. [PMID: 34577715 PMCID: PMC8467303 DOI: 10.3390/mi12091071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 11/19/2022]
Abstract
In this paper, a prototype of a semi-automatic lab-on-PCB for agarose gel preparation and electrophoresis is developed. The dimensions of the device are 38 × 34 mm2 and it includes a conductivity sensor for detecting the TAE buffer (Tris-acetate-EDTA buffer), a microheater for increasing the solubility of the agarose, a negative temperature coefficient (NTC) thermistor for controlling the temperature, a light dependent resistor (LDR) sensor for measuring the transparency of the mixture, and two electrodes for performing the electrophoresis. The agarose preparation functions are governed by a microcontroller. The device requires a PMMA structure to define the wells of the agarose gel, and to release the electrodes from the agarose. The maximum voltage and current that the system requires are 40 V to perform the electrophoresis, and 1 A for activating the microheater. The chosen temperature for mixing is 80 ∘C, with a mixing time of 10 min. In addition, the curing time is about 30 min. This device is intended to be integrated as a part of a larger lab-on-PCB system for DNA amplification and detection. However, it can be used to migrate DNA amplified in conventional thermocyclers. Moreover, the device can be modified for preparing larger agarose gels and performing electrophoresis.
Collapse
|
42
|
Shi Y, Ye P, Yang K, Meng J, Guo J, Pan Z, Bayin Q, Zhao W. Application of Microfluidics in Immunoassay: Recent Advancements. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:2959843. [PMID: 34326976 PMCID: PMC8302407 DOI: 10.1155/2021/2959843] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022]
Abstract
In recent years, point-of-care testing has played an important role in immunoassay, biochemical analysis, and molecular diagnosis, especially in low-resource settings. Among various point-of-care-testing platforms, microfluidic chips have many outstanding advantages. Microfluidic chip applies the technology of miniaturizing conventional laboratory which enables the whole biochemical process including reagent loading, reaction, separation, and detection on the microchip. As a result, microfluidic platform has become a hotspot of research in the fields of food safety, health care, and environmental monitoring in the past few decades. Here, the state-of-the-art application of microfluidics in immunoassay in the past decade will be reviewed. According to different driving forces of fluid, microfluidic platform is divided into two parts: passive manipulation and active manipulation. In passive manipulation, we focus on the capillary-driven microfluidics, while in active manipulation, we introduce pressure microfluidics, centrifugal microfluidics, electric microfluidics, optofluidics, magnetic microfluidics, and digital microfluidics. Additionally, within the introduction of each platform, innovation of the methods used and their corresponding performance improvement will be discussed. Ultimately, the shortcomings of different platforms and approaches for improvement will be proposed.
Collapse
Affiliation(s)
- Yuxing Shi
- School of Automation Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Peng Ye
- School of Automation Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Kuojun Yang
- School of Automation Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jie Meng
- School of Automation Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jiuchuan Guo
- School of Automation Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Zhixiang Pan
- School of Automation Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Qiaoge Bayin
- School of Automation Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Wenhao Zhao
- School of Automation Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| |
Collapse
|
43
|
Knowing more from less: miniaturization of ligand-binding assays and electrophoresis as new paradigms for at-line monitoring and control of mammalian cell bioprocesses. Curr Opin Biotechnol 2021; 71:55-64. [PMID: 34246047 DOI: 10.1016/j.copbio.2021.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/17/2021] [Accepted: 06/20/2021] [Indexed: 02/01/2023]
Abstract
Monitoring technologies for Process Analytical Technology (PAT) in mammalian cell cultures are often focusing on the same hand full parameters although a deeper knowledge and control of a larger panel of culture components would highly benefit process optimization, control and robustness. This short review highlights key advances in microfluidic affinity assays and microchip capillary electrophoresis (MCE). Aiming at the miniaturization and integration of PAT, these can detect at-line a variety of metabolites, proteins and Critical Quality Attributes (CQA's) in a bioprocess. Furthermore, discrete analytical components, which can potentially support the translation of increasingly mature microfluidic technologies towards this novel application, are also presented as a comprehensive toolbox ranging from sample preparation to signal acquisition.
Collapse
|
44
|
Rapid Production of PDMS Microdevices for Electrodriven Separations and Microfluidics by 3D-Printed Scaffold Removal. SEPARATIONS 2021. [DOI: 10.3390/separations8050067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In our work, we produced PDMS-based microfluidic devices by mechanical removal of 3D-printed scaffolds inserted in PDMS. Two setups leading to the fabrication of monolithic PDMS-based microdevices and bonded (or stamped) PDMS-based microdevices were designed. In the monolithic devices, the 3D-printed scaffolds were fully inserted in the PDMS and then carefully removed. The bonded devices were produced by forming imprints of the 3D-printed scaffolds in PDMS, followed by bonding the PDMS parts to glass slides. All these microfluidic devices were then successfully employed in three proof-of-concept applications: capture of magnetic microparticles, formation of droplets, and isotachophoresis separation of model organic dyes.
Collapse
|
45
|
Liu Y, Liu Y, Liu Z, Hill JP, Alowasheeir A, Xu Z, Xu X, Yamauchi Y. Ultra-durable, multi-template molecularly imprinted polymers for ultrasensitive monitoring and multicomponent quantification of trace sulfa antibiotics. J Mater Chem B 2021; 9:3192-3199. [PMID: 33885623 DOI: 10.1039/d1tb00091h] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Traditional analysis methods are susceptible to interference caused by the complexity of sample matrices, and detector surface fouling arising from nonspecific adsorption of microorganisms (in biological samples) which leads in particular to a gradual loss of sensitivity. Imprinted materials can be used to effectively reduce interference originating in the matrices. However, the poor reproducibility and multicomponent quantification of trace antibiotics represent significant challenges to the detection process. Meanwhile, the high biological risk presented by bacterial antibiotic immunity and the persistence of antibiotics in foodstuffs, especially meat, both caused by the overuse of sulfonamide antibiotics, remain urgent issues. Here, we present the first example of a method for the accurate quantification of trace sulfa antibiotics (SAs) based on multi-template imprinted polymers (MMIPs). Levels of multiple SAs have been simultaneously successfully quantified by applying MMIP extraction coupled with UPLC-MS/MS analysis. This method shows excellent linearity of detection in the range of 0.1-500 μg L-1, and ultrasensitivity with low limits of detection of 0.03 μg L-1. The maximum SA residue recovered from sample tissues by using MMIPs was 5.48 μg g-1. MMIP-coupled UPLC-MS/MS quantification of SAs is an accurate and repeatable method for the monitoring of SA accumulation in mouse tissue samples. It also provides an effective strategy for the tracking and quantification of drugs in other biological samples.
Collapse
Affiliation(s)
- Yuanchen Liu
- Faculty of Science, Kunming University of Science and Technology, Kunming 650500, China.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Wang J, Ma P, Kim DH, Liu BF, Demirci U. Towards Microfluidic-Based Exosome Isolation and Detection for Tumor Therapy. NANO TODAY 2021; 37:101066. [PMID: 33777166 PMCID: PMC7990116 DOI: 10.1016/j.nantod.2020.101066] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Exosomes are a class of cell-secreted, nano-sized extracellular vesicles with a bilayer membrane structure of 30-150 nm in diameter. Their discovery and application have brought breakthroughs in numerous areas, such as liquid biopsies, cancer biology, drug delivery, immunotherapy, tissue repair, and cardiovascular diseases. Isolation of exosomes is the first step in exosome-related research and its applications. Standard benchtop exosome separation and sensing techniques are tedious and challenging, as they require large sample volumes, multi-step operations that are complex and time-consuming, requiring cumbersome and expensive instruments. In contrast, microfluidic platforms have the potential to overcome some of these limitations, owing to their high-precision processing, ability to handle liquids at a microscale, and integrability with various functional units, such as mixers, actuators, reactors, separators, and sensors. These platforms can optimize the detection process on a single device, representing a robust and versatile technique for exosome separation and sensing to attain high purity and high recovery rates with a short processing time. Herein, we overview microfluidic strategies for exosome isolation based on their hydrodynamic properties, size filtration, acoustic fields, immunoaffinity, and dielectrophoretic properties. We focus especially on advances in label-free isolation of exosomes with active biological properties and intact morphological structures. Further, we introduce microfluidic techniques for the detection of exosomal proteins and RNAs with high sensitivity, high specificity, and low detection limits. We summarize the biomedical applications of exosome-mediated therapeutic delivery targeting cancer cells. To highlight the advantages of microfluidic platforms, conventional techniques are included for comparison. Future challenges and prospects of microfluidics towards exosome isolation applications are also discussed. Although the use of exosomes in clinical applications still faces biological, technical, regulatory, and market challenges, in the foreseeable future, recent developments in microfluidic technologies are expected to pave the way for tailoring exosome-related applications in precision medicine.
Collapse
Affiliation(s)
- Jie Wang
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Peng Ma
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Britton Chance Center for Biomedical Photonics 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
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Daniel H Kim
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California 95064, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Bi-Feng Liu
- Britton Chance Center for Biomedical Photonics 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
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| |
Collapse
|
47
|
Skottvoll F, Hansen FA, Harrison S, Boger IS, Mrsa A, Restan MS, Stein M, Lundanes E, Pedersen-Bjergaard S, Aizenshtadt A, Krauss S, Sullivan G, Bogen IL, Wilson SR. Electromembrane Extraction and Mass Spectrometry for Liver Organoid Drug Metabolism Studies. Anal Chem 2021; 93:3576-3585. [PMID: 33534551 PMCID: PMC8023518 DOI: 10.1021/acs.analchem.0c05082] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022]
Abstract
Liver organoids are emerging tools for precision drug development and toxicity screening. We demonstrate that electromembrane extraction (EME) based on electrophoresis across an oil membrane is suited for segregating selected organoid-derived drug metabolites prior to mass spectrometry (MS)-based measurements. EME allowed drugs and drug metabolites to be separated from cell medium components (albumin, etc.) that could interfere with subsequent measurements. Multiwell EME (parallel-EME) holding 100 μL solutions allowed for simple and repeatable monitoring of heroin phase I metabolism kinetics. Organoid parallel-EME extracts were compatible with ultrahigh-performance liquid chromatography (UHPLC) used to separate the analytes prior to detection. Taken together, liver organoids are well-matched with EME followed by MS-based measurements.
Collapse
Affiliation(s)
- Frøydis
Sved Skottvoll
- Department
of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| | - Frederik André Hansen
- Department
of Pharmacy, University of Oslo, P.O. Box 1068, Blindern, NO-0316 Oslo, Norway
| | - Sean Harrison
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| | - Ida Sneis Boger
- Department
of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| | - Ago Mrsa
- Department
of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| | - Magnus Saed Restan
- Department
of Pharmacy, University of Oslo, P.O. Box 1068, Blindern, NO-0316 Oslo, Norway
| | - Matthias Stein
- Institute
of Medicinal and Pharmaceutical Chemistry, TU Braunschweig, Beethovenstr.
55, DE-38106 Braunschweig, Germany
| | - Elsa Lundanes
- Department
of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
| | - Stig Pedersen-Bjergaard
- Department
of Pharmacy, University of Oslo, P.O. Box 1068, Blindern, NO-0316 Oslo, Norway
- Department
of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Aleksandra Aizenshtadt
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| | - Stefan Krauss
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
- Department
of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 1110, Blindern, 0317, Oslo, Norway
| | - Gareth Sullivan
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
- Department
of Pediatric Research, Oslo University Hospital
and University of Oslo, P.O. Box 1112,
Blindern, 0317 Oslo, Norway
| | - Inger Lise Bogen
- Section
for Drug Abuse Research, Department of Forensic Sciences, Oslo University Hospital, P.O. Box 4950, Nydalen, NO-0424 Oslo, Norway
- Institute
of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1103,
Blindern, NO-0317 Oslo, Norway
| | - Steven Ray Wilson
- Department
of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
- Hybrid
Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences,
Faculty of Medicine, University of Oslo, P.O. Box 1112, Blindern, NO-0317 Oslo, Norway
| |
Collapse
|
48
|
Nouwairi RL, O'Connell KC, Gunnoe LM, Landers JP. Microchip Electrophoresis for Fluorescence-Based Measurement of Polynucleic Acids: Recent Developments. Anal Chem 2020; 93:367-387. [PMID: 33351599 DOI: 10.1021/acs.analchem.0c04596] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Renna L Nouwairi
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Killian C O'Connell
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Leah M Gunnoe
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22903, United States
| | - James P Landers
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22903, United States.,Department of Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, Virginia 22903, United States.,Department of Pathology, University of Virginia Health Science Center, Charlottesville, Virginia 22903, United States
| |
Collapse
|
49
|
Xie Y, Rufo J, Zhong R, Rich J, Li P, Leong KW, Huang TJ. Microfluidic Isolation and Enrichment of Nanoparticles. ACS NANO 2020; 14:16220-16240. [PMID: 33252215 PMCID: PMC8164652 DOI: 10.1021/acsnano.0c06336] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Over the past decades, nanoparticles have increased in implementation to a variety of applications ranging from high-efficiency electronics to targeted drug delivery. Recently, microfluidic techniques have become an important tool to isolate and enrich populations of nanoparticles with uniform properties (e.g., size, shape, charge) due to their precision, versatility, and scalability. However, due to the large number of microfluidic techniques available, it can be challenging to identify the most suitable approach for isolating or enriching a nanoparticle of interest. In this review article, we survey microfluidic methods for nanoparticle isolation and enrichment based on their underlying mechanisms, including acoustofluidics, dielectrophoresis, filtration, deterministic lateral displacement, inertial microfluidics, optofluidics, electrophoresis, and affinity-based methods. We discuss the principles, applications, advantages, and limitations of each method. We also provide comparisons with bulk methods, perspectives for future developments and commercialization, and next-generation applications in chemistry, biology, and medicine.
Collapse
Affiliation(s)
- Yuliang Xie
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242, United States
| | - Joseph Rufo
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Ruoyu Zhong
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Joseph Rich
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Peng Li
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, New York 10032, United States
| | - Tony Jun Huang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
50
|
Guzman NA, Guzman DE. A Two-Dimensional Affinity Capture and Separation Mini-Platform for the Isolation, Enrichment, and Quantification of Biomarkers and Its Potential Use for Liquid Biopsy. Biomedicines 2020; 8:biomedicines8080255. [PMID: 32751506 PMCID: PMC7459796 DOI: 10.3390/biomedicines8080255] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/22/2020] [Accepted: 07/26/2020] [Indexed: 02/07/2023] Open
Abstract
Biomarker detection for disease diagnosis, prognosis, and therapeutic response is becoming increasingly reliable and accessible. Particularly, the identification of circulating cell-free chemical and biochemical substances, cellular and subcellular entities, and extracellular vesicles has demonstrated promising applications in understanding the physiologic and pathologic conditions of an individual. Traditionally, tissue biopsy has been the gold standard for the diagnosis of many diseases, especially cancer. More recently, liquid biopsy for biomarker detection has emerged as a non-invasive or minimally invasive and less costly method for diagnosis of both cancerous and non-cancerous diseases, while also offering information on the progression or improvement of disease. Unfortunately, the standardization of analytical methods to isolate and quantify circulating cells and extracellular vesicles, as well as their extracted biochemical constituents, is still cumbersome, time-consuming, and expensive. To address these limitations, we have developed a prototype of a portable, miniaturized instrument that uses immunoaffinity capillary electrophoresis (IACE) to isolate, concentrate, and analyze cell-free biomarkers and/or tissue or cell extracts present in biological fluids. Isolation and concentration of analytes is accomplished through binding to one or more biorecognition affinity ligands immobilized to a solid support, while separation and analysis are achieved by high-resolution capillary electrophoresis (CE) coupled to one or more detectors. When compared to other existing methods, the process of this affinity capture, enrichment, release, and separation of one or a panel of biomarkers can be carried out on-line with the advantages of being rapid, automated, and cost-effective. Additionally, it has the potential to demonstrate high analytical sensitivity, specificity, and selectivity. As the potential of liquid biopsy grows, so too does the demand for technical advances. In this review, we therefore discuss applications and limitations of liquid biopsy and hope to introduce the idea that our affinity capture-separation device could be used as a form of point-of-care (POC) diagnostic technology to isolate, concentrate, and analyze circulating cells, extracellular vesicles, and viruses.
Collapse
Affiliation(s)
- Norberto A. Guzman
- Princeton Biochemicals, Inc., Princeton, NJ 08816, USA
- Correspondence: ; Tel.: +1-908-510-5258
| | - Daniel E. Guzman
- Princeton Biochemicals, Inc., Princeton, NJ 08816, USA
- Department of Internal Medicine, University of California at San Francisco, San Francisco, CA 94143, USA; or
| |
Collapse
|