1
|
Ozyilmaz E, Gediz Ilis G. Design and Performance Analysis of Spiral Microchannels for Efficient Particle Separation Using Inertial Microfluidics. MICROMACHINES 2025; 16:349. [PMID: 40141959 PMCID: PMC11946254 DOI: 10.3390/mi16030349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025]
Abstract
Accurate separation in microfluidic devices is crucial for biomedical applications; however, enhancing their performance remains challenging due to computational and experimental constraints. This study aims to optimize microfluidic devices by systematically refining spiral microchannel configurations for the segregation of circulating tumor cells (CTCs) and red blood cells (RBCs) through detailed variable analysis and resource-efficient techniques. The spiral design was developed into six variations, considering loop numbers (2, 3, and 4), aspect ratios (2.333, 3.333, and 5), spiral radii (5, 6, and 7 mm), flow rates (1.5, 2, and 3 mL/min), surface roughness levels (0, 0.5, and 1 μm), and particle sizes (12, 18, and 24 μm). Simulations were conducted in COMSOL Multiphysics and evaluated using the Taguchi method to determine the optimal configuration, reducing the analysis set from 216 to 27 through an efficient experimental design approach. The results identified the optimal structure as having an aspect ratio of 3.333, four loops, a spiral radius of 6-7 mm, a flow rate of 3 mL/min, a surface roughness of 1 μm, and a particle diameter of 24 μm. Among the evaluated parameters, aspect ratio (61.2%) had the most significant impact, followed by the number of loops (13.9%) and flow rate (9.4%). The optimized design demonstrated high separation efficiency and purity, achieving 97.5% and 97.6%, respectively. The fabrication process involved 3D-printing the channel mold, followed by polydimethylsiloxane (PDMS) casting, validating the durability and scalability of the proposed design. This study integrates simulation and experimental results, providing a robust framework for developing next-generation microfluidic devices and advancing diagnostic and targeted therapeutic applications.
Collapse
Affiliation(s)
- Eda Ozyilmaz
- Department of Mechanical Engineering, Gebze Technical University, 41400 Gebze, Kocaeli, Türkiye;
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Gamze Gediz Ilis
- Department of Mechanical Engineering, Gebze Technical University, 41400 Gebze, Kocaeli, Türkiye;
| |
Collapse
|
2
|
Fan D, Liu Y, Liu Y. The Latest Advances in Microfluidic DLD Cell Sorting Technology: The Optimization of Channel Design. BIOSENSORS 2025; 15:126. [PMID: 39997028 PMCID: PMC11853672 DOI: 10.3390/bios15020126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025]
Abstract
Cell sorting plays a crucial role in both medical and biological research. As a key passive sorting technique in the field of microfluidics, deterministic lateral displacement (DLD) has been widely applied to cell separation and sorting. This review aims to summarize the latest advances in the optimization of channel design for microfluidic DLD cell sorting. First, we provide an overview of the design elements of microfluidic DLD cell sorting channels, focusing on key factors that affect separation efficiency and accuracy, including channel geometry, fluid dynamics, and the interaction between cells and channel surfaces. Subsequently, we review recent innovations and progress in channel design for microfluidic DLD technology, exploring its applications in biomedical fields and its integration with machine learning. Additionally, we discuss the challenges currently faced in optimizing channel design for microfluidic DLD cell sorting. Finally, based on existing research, we make a summary and put forward prospective views on the further development of this field.
Collapse
Affiliation(s)
- Dan Fan
- School of Engineering, Dali University, Dali 671003, China;
| | - Yi Liu
- School of Engineering, Dali University, Dali 671003, China;
| | - Yaling Liu
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Bioengineering, Lehigh University, Bethlehem, PA 18015, USA
| |
Collapse
|
3
|
Zheng K, Liu Z, Pang Y, Wang X, Zhao S, Zheng N, Cai F, Zhang C. Predictive Model for Cell Positioning during Periodic Lateral Migration in Spiral Microchannels. Anal Chem 2024; 96:18230-18238. [PMID: 39436732 DOI: 10.1021/acs.analchem.4c04456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
The periodic lateral migration of submicrometer cells is the primary factor leading to low precision in a spiral microchannel during cell isolation. In this study, a mathematical predictive model (PM) is derived for the lateral position of cells during the periodic lateral migration process. We analyze the relationship of migration period, migration width, and starting point of lateral migration with microchannel structure and flow conditions and determine the empirical coefficients in PM. Results indicate that the aspect ratio of the microchannel and the Reynolds number (Re) are key factors that influence the periodicity of the cell lateral migration. The lateral migration width is jointly affected by Re, the cell blockage ratio, and the microchannel curvature radius. The inlet structure of the microchannel and the ratio of the cell sample to the sheath flow rate are critical parameters for regulating the initial position. Moreover, the structure of the pressure field at the inlet constrains the distribution range of the starting point of the lateral migration. Regardless of whether the particles/cells undergo 0.5, 1, or multiple lateral migration cycles, the lateral positions predicted by PM align well with the experimental observations, thus verifying the accuracy of PM. This research helps to elucidate the characteristics of periodic lateral migration of cells in spiral microchannels and can provide practical guidance for the development and optimization of miniature spiral microchannel chips for precise cell isolation.
Collapse
Affiliation(s)
- Kai Zheng
- College of Mechanical and Energy Engineering, Beijing University of Technology, Beijing 100124, China
| | - Zhaomiao Liu
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing 100124, China
| | - Yan Pang
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing 100124, China
| | - Xiang Wang
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing 100124, China
| | - Siyu Zhao
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing 100124, China
| | - Nan Zheng
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing 100124, China
| | - Fanming Cai
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing 100124, China
| | - Chenchen Zhang
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
4
|
Elsemary MT, Maritz MF, Smith LE, Warkiani ME, Thierry B. Enrichment of T-lymphocytes from leukemic blood using inertial microfluidics toward improved chimeric antigen receptor-T cell manufacturing. Cytotherapy 2024; 26:1264-1274. [PMID: 38819362 DOI: 10.1016/j.jcyt.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/30/2024] [Accepted: 05/05/2024] [Indexed: 06/01/2024]
Abstract
Chimeric antigen receptor cell therapy is a successful immunotherapy for the treatment of blood cancers. However, hurdles in their manufacturing remain including efficient isolation and purification of the T-cell starting material. Herein, we describe a one-step separation based on inertial spiral microfluidics for efficient enrichment of T-cells in B-cell acute lymphoblastic leukemia (ALL) and B-cell chronic lymphocytic leukemia patient's samples. In healthy donors used to optimize the process, the lymphocyte purity was enriched from 65% (SD ± 0.2) to 91% (SD ± 0.06) and T-cell purity was enriched from 45% (SD ± 0.1) to 73% (SD ± 0.02). Leukemic samples had higher starting B-cells compared to the healthy donor samples. Efficient enrichment and recovery of lymphocytes and T-cells were achieved in ALL samples with B-cells, monocytes and leukemic blasts depleted by 80% (SD ± 0.09), 89% (SD ± 0.1) and 74% (SD ± 0.09), respectively, and a 70% (SD ± 0.1) T-cell recovery. Chronic lymphocytic leukemia samples had lower T-cell numbers, and the separation process was less efficient compared to the ALL. This study demonstrates the use of inertial microfluidics for T-cell enrichment and depletion of B-cell blasts in ALL, suggesting its potential to address a key bottleneck of the chimeric antigen receptor-T manufacturing workflow.
Collapse
MESH Headings
- Humans
- T-Lymphocytes/immunology
- Receptors, Chimeric Antigen/immunology
- Immunotherapy, Adoptive/methods
- Microfluidics/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Cell Separation/methods
- B-Lymphocytes/immunology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Mona T Elsemary
- Future Industries Institute, University of South Australia Mawson Lakes Campus, Mawson Lakes, SA, Australia
| | - Michelle F Maritz
- Future Industries Institute, University of South Australia Mawson Lakes Campus, Mawson Lakes, SA, Australia
| | - Louise E Smith
- Future Industries Institute, University of South Australia Mawson Lakes Campus, Mawson Lakes, SA, Australia
| | | | - Benjamin Thierry
- Future Industries Institute, University of South Australia Mawson Lakes Campus, Mawson Lakes, SA, Australia.
| |
Collapse
|
5
|
Norouzy N, Nikdoost A, Rezai P. Parallelization of Curved Inertial Microfluidic Channels to Increase the Throughput of Simultaneous Microparticle Separation and Washing. MICROMACHINES 2024; 15:1228. [PMID: 39459102 PMCID: PMC11509581 DOI: 10.3390/mi15101228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024]
Abstract
The rising global need for clean water highlights the importance of efficient sample preparation methods to separate and wash various contaminants such as microparticles. Microfluidic methods for these purposes have emerged but they mostly deliver either separation or washing, with very low throughputs. Here, we investigate parallelization of a curved-channel particle separation and washing device in order to increase its throughput for sample preparation. A curved microchannel applies inertial forces to focus larger 10 µm microparticles at the inner wall of the channel and separate them from smaller 5 µm microparticles at the outer wall. At the same time, Dean flow recirculation is used to exchange the carrier solution of the large microparticles to a clean buffer (washing). We increased the number of curved channels in a stepwise manner from two to four to eight channels in two different arraying designs, i.e., rectangular and polar arrays. We examined efficient separation of target 10 µm particles from 5 µm particles, while transferring the larger microparticles into a clean buffer. Dean flow recirculation studies demonstrated that the rectangular arrayed device performs better, providing solution exchange efficiencies of more than 96% on average as compared to 89% for the polar array device. Our 8-curve rectangular array device provided a particle separation efficiency of 98.93 ± 0.91%, while maintaining a sample purity of 92.83 ± 1.47% at a high working flow rate of 12.8 mL/min. Moreover, the target particles were transferred into a clean buffer with a solution exchange efficiency of 96.81 ± 0.54% in our 8-curve device. Compared to the literature, our in-plane parallelization design of curved microchannels resulted in a 13-fold increase in the working flow rate of the setup while maintaining a very high performance in particle separation and washing. Our microfluidic device offers the potential to enhance the throughput and the separation and washing efficiencies in applications for biological and environmental samples.
Collapse
Affiliation(s)
| | | | - Pouya Rezai
- Department of Mechanical Engineering, York University, BRG 433B, 4700 Keele St., Toronto, ON M3J 1P3, Canada; (N.N.); (A.N.)
| |
Collapse
|
6
|
Petruzzellis I, Martínez Vázquez R, Caragnano S, Gaudiuso C, Osellame R, Ancona A, Volpe A. Lab-on-Chip Systems for Cell Sorting: Main Features and Advantages of Inertial Focusing in Spiral Microchannels. MICROMACHINES 2024; 15:1135. [PMID: 39337795 PMCID: PMC11434521 DOI: 10.3390/mi15091135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024]
Abstract
Inertial focusing-based Lab-on-Chip systems represent a promising technology for cell sorting in various applications, thanks to their alignment with the ASSURED criteria recommended by the World Health Organization: Affordable, Sensitive, Specific, User-friendly, Rapid and Robust, Equipment-free, and Delivered. Inertial focusing techniques using spiral microchannels offer a rapid, portable, and easy-to-prototype solution for cell sorting. Various microfluidic devices have been investigated in the literature to understand how hydrodynamic forces influence particle focusing in spiral microchannels. This is crucial for the effective prototyping of devices that allow for high-throughput and efficient filtration of particles of different sizes. However, a clear, comprehensive, and organized overview of current research in this area is lacking. This review aims to fill this gap by offering a thorough summary of the existing literature, thereby guiding future experimentation and facilitating the selection of spiral geometries and materials for cell sorting in microchannels. To this end, we begin with a detailed theoretical introduction to the physical mechanisms underlying particle separation in spiral microfluidic channels. We also dedicate a section to the materials and prototyping techniques most commonly used for spiral microchannels, highlighting and discussing their respective advantages and disadvantages. Subsequently, we provide a critical examination of the key details of inertial focusing across various cross-sections (rectangular, trapezoidal, triangular, hybrid) in spiral devices as reported in the literature.
Collapse
Affiliation(s)
- Isabella Petruzzellis
- Physics Department, Università degli Studi di Bari & Politecnico di Bari, Via Orabona 4, 7016 Bari, Italy; (I.P.); (S.C.); (A.A.)
| | - Rebeca Martínez Vázquez
- Institute for Photonics and Nanotechnologies (IFN), National Research Council, Piazza L. da Vinci 32, 20133 Milan, Italy;
| | - Stefania Caragnano
- Physics Department, Università degli Studi di Bari & Politecnico di Bari, Via Orabona 4, 7016 Bari, Italy; (I.P.); (S.C.); (A.A.)
| | - Caterina Gaudiuso
- Institute for Photonics and Nanotechnologies (IFN), National Research Council, Via Amendola 173, 70125 Bari, Italy;
| | - Roberto Osellame
- Institute for Photonics and Nanotechnologies (IFN), National Research Council, Piazza L. da Vinci 32, 20133 Milan, Italy;
| | - Antonio Ancona
- Physics Department, Università degli Studi di Bari & Politecnico di Bari, Via Orabona 4, 7016 Bari, Italy; (I.P.); (S.C.); (A.A.)
- Institute for Photonics and Nanotechnologies (IFN), National Research Council, Via Amendola 173, 70125 Bari, Italy;
| | - Annalisa Volpe
- Physics Department, Università degli Studi di Bari & Politecnico di Bari, Via Orabona 4, 7016 Bari, Italy; (I.P.); (S.C.); (A.A.)
- Institute for Photonics and Nanotechnologies (IFN), National Research Council, Via Amendola 173, 70125 Bari, Italy;
| |
Collapse
|
7
|
Norouzy N, Zabihihesari A, Rezai P. Simultaneous high-throughput particle-bacteria separation and solution exchange via in-plane and out-of-plane parallelization of microfluidic centrifuges. BIOMICROFLUIDICS 2024; 18:054107. [PMID: 39345266 PMCID: PMC11435783 DOI: 10.1063/5.0215930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/20/2024] [Indexed: 10/01/2024]
Abstract
Inertial microfluidic devices have gained attention for point-of-need (PoN) sample preparation. Yet, devices capable of simultaneous particle-bacteria solution exchange and separation are low in throughput, hindering their applicability to PoN settings. This paper introduces a microfluidic centrifuge for high-throughput solution exchange and separation of microparticles, addressing the need for processing large sample volumes at elevated flow rates. The device integrates Dean flow recirculation and inertial focusing of microparticles within 24 curved microchannels assembled in a three-layer configuration via in-plane and out-of-plane parallelization. We studied solution exchange and particle migration using singleplex and duplex samples across devices with varying curve numbers (2-curve, 8-curve, and 24-curve). Processing 5 and 10 μm microparticles at flow rates up to 16.8 ml/min achieved a solution exchange efficiency of 96.69%. In singleplex solutions, 10 and 5 μm particles selectively migrated to inner and outer outlets, demonstrating separation efficiencies of 99.7% and 90.3%, respectively. With duplex samples, sample purity was measured to be 93.4% and 98.6% for 10 and 5 μm particles collected from the inner and the outer outlets, respectively. Application of our device in biological assays was shown by performing duplex experiments where 10 μm particles were isolated from Salmonella bacterial suspension with purity of 97.8% while increasing the state-of-the-art particle solution exchange and separation throughput by 16 folds. This parallelization enabled desirable combinations of high throughput, low-cost, and scalability, without compromising efficiency and purity, paving the way for sample preparation at the PoN in the future.
Collapse
Affiliation(s)
- Nima Norouzy
- Department of Mechanical Engineering, York University, BRG 433B, 4700 Keele St., Toronto, Ontario M3J 1P3, Canada
| | - Alireza Zabihihesari
- Department of Mechanical Engineering, York University, BRG 433B, 4700 Keele St., Toronto, Ontario M3J 1P3, Canada
| | - Pouya Rezai
- Department of Mechanical Engineering, York University, BRG 433B, 4700 Keele St., Toronto, Ontario M3J 1P3, Canada
| |
Collapse
|
8
|
Lee KCM, Chung BMF, Siu DMD, Ho SCK, Ng DKH, Tsia KK. Dispersion-free inertial focusing (DIF) for high-yield polydisperse micro-particle filtration and analysis. LAB ON A CHIP 2024; 24:4182-4197. [PMID: 39101363 DOI: 10.1039/d4lc00275j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
Inertial focusing excels at the precise spatial ordering and separation of microparticles by size within fluid flows. However, this advantage, resulting from its inherent size-dependent dispersion, could turn into a drawback that challenges applications requiring consistent and uniform positioning of polydisperse particles, such as microfiltration and flow cytometry. To overcome this fundamental challenge, we introduce Dispersion-Free Inertial Focusing (DIF). This new method minimizes particle size-dependent dispersion while maintaining the high throughput and precision of standard inertial focusing, even in a highly polydisperse scenario. We demonstrate a rule-of-thumb principle to reinvent an inertial focusing system and achieve an efficient focusing of particles ranging from 6 to 30 μm in diameter onto a single plane with less than 3 μm variance and over 95% focusing efficiency at highly scalable throughput (2.4-30 mL h-1) - a stark contrast to existing technologies that struggle with polydispersity. We demonstrated that DIF could be applied in a broad range of applications, particularly enabling high-yield continuous microparticle filtration and large-scale high-resolution single-cell morphological analysis of heterogeneous cell populations. This new technique is also readily compatible with the existing inertial microfluidic design and thus could unleash more diverse systems and applications.
Collapse
Affiliation(s)
- Kelvin C M Lee
- The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong.
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong
| | - Bob M F Chung
- The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong.
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong
| | - Dickson M D Siu
- The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong.
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong
| | - Sam C K Ho
- The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong.
| | - Daniel K H Ng
- The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong.
| | - Kevin K Tsia
- The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong.
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong
| |
Collapse
|
9
|
Mika T, Kalnins M, Spalvins K. The use of droplet-based microfluidic technologies for accelerated selection of Yarrowia lipolytica and Phaffia rhodozyma yeast mutants. Biol Methods Protoc 2024; 9:bpae049. [PMID: 39114747 PMCID: PMC11303513 DOI: 10.1093/biomethods/bpae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/24/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Microorganisms are widely used for the industrial production of various valuable products, such as pharmaceuticals, food and beverages, biofuels, enzymes, amino acids, vaccines, etc. Research is constantly carried out to improve their properties, mainly to increase their productivity and efficiency and reduce the cost of the processes. The selection of microorganisms with improved qualities takes a lot of time and resources (both human and material); therefore, this process itself needs optimization. In the last two decades, microfluidics technology appeared in bioengineering, which allows for manipulating small particles (from tens of microns to nanometre scale) in the flow of liquid in microchannels. The technology is based on small-volume objects (microdroplets from nano to femtolitres), which are manipulated using a microchip. The chip is made of an optically transparent inert to liquid medium material and contains a series of channels of small size (<1 mm) of certain geometry. Based on the physical and chemical properties of microparticles (like size, weight, optical density, dielectric constant, etc.), they are separated using microsensors. The idea of accelerated selection of microorganisms is the application of microfluidic technologies to separate mutants with improved qualities after mutagenesis. This article discusses the possible application and practical implementation of microfluidic separation of mutants, including yeasts like Yarrowia lipolytica and Phaffia rhodozyma after chemical mutagenesis will be discussed.
Collapse
Affiliation(s)
- Taras Mika
- Institute of Energy Systems and Environment, Riga Technical University, 12 – K1 Āzene street, Riga, LV-1048, Latvia
| | - Martins Kalnins
- Institute of Energy Systems and Environment, Riga Technical University, 12 – K1 Āzene street, Riga, LV-1048, Latvia
| | - Kriss Spalvins
- Institute of Energy Systems and Environment, Riga Technical University, 12 – K1 Āzene street, Riga, LV-1048, Latvia
| |
Collapse
|
10
|
Mortelmans T, Marty B, Kazazis D, Padeste C, Li X, Ekinci Y. Three-Dimensional Microfluidic Capillary Device for Rapid and Multiplexed Immunoassays in Whole Blood. ACS Sens 2024; 9:2455-2464. [PMID: 38687557 PMCID: PMC11129352 DOI: 10.1021/acssensors.4c00153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
In this study, we demonstrate whole blood immunoassays using a microfluidic device optimized for conducting rapid and multiplexed fluorescence-linked immunoassays. The device is capable of handling whole blood samples without any preparatory treatment. The three-dimensional channels in poly(methyl methacrylate) are designed to passively load bodily fluids and, due to their linearly tapered profile, facilitate size-dependent immobilization of biofunctionalized particles. The channel geometry is optimized to allow for the unimpeded flow of cellular constituents such as red blood cells (RBCs). Additionally, to make the device easier to operate, the biofunctionalized particles are pretrapped in a first step, and the channel is dried under vacuum, after which it can be loaded with the biological sample. This novel approach and design eliminated the need for traditionally laborious steps such as filtering, incubation, and washing steps, thereby substantially simplifying the immunoassay procedures. Moreover, by leveraging the shallow device dimensions, we show that sample loading to read-out is possible within 5 min. Our results also show that the presence of RBCs does not compromise the sensitivity of the assays when compared to those performed in a pure buffer solution. This highlights the practical adaptability of the device for simple and rapid whole-blood assays. Lastly, we demonstrate the device's multiplexing capability by pretrapping particles of different sizes, each functionalized with a different antigen, thus enabling the performance of multiplexed on-chip whole-blood immunoassays, showcasing the device's versatility and effectiveness toward low-cost, simple, and multiplexed sensing of biomarkers and pathogens directly in whole blood.
Collapse
Affiliation(s)
- Thomas Mortelmans
- Laboratory for X-ray Nanoscience and Technologies, 5232 Villigen, Switzerland
- Swiss Nanoscience Institute, University of Basel, 4056 Basel, Switzerland
| | - Balz Marty
- Laboratory for X-ray Nanoscience and Technologies, 5232 Villigen, Switzerland
| | - Dimitrios Kazazis
- Laboratory for X-ray Nanoscience and Technologies, 5232 Villigen, Switzerland
| | - Celestino Padeste
- Laboratory of Nanoscale Biology, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Xiaodan Li
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Yasin Ekinci
- Laboratory for X-ray Nanoscience and Technologies, 5232 Villigen, Switzerland
| |
Collapse
|
11
|
Li Y, Zhou S, Wu Q, Gong C. CRISPR/Cas gene editing and delivery systems for cancer therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1938. [PMID: 38456346 DOI: 10.1002/wnan.1938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 03/09/2024]
Abstract
CRISPR/Cas systems stand out because of simplicity, efficiency, and other superiorities, thus becoming attractive and brilliant gene-editing tools in biomedical field including cancer therapy. CRISPR/Cas systems bring promises for cancer therapy through manipulating and engineering on tumor cells or immune cells. However, there have been concerns about how to overcome the numerous physiological barriers and deliver CRISPR components to target cells efficiently and accurately. In this review, we introduced the mechanisms of CRISPR/Cas systems, summarized the current delivery strategies of CRISPR/Cas systems by physical methods, viral vectors, and nonviral vectors, and presented the current application of CRISPR/Cas systems in cancer clinical treatment. Furthermore, we discussed prospects related to delivery approaches of CRISPR/Cas systems. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yingjie Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shiyao Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qinjie Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Changyang Gong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Akbari Z, Raoufi MA, Mirjalali S, Aghajanloo B. A review on inertial microfluidic fabrication methods. BIOMICROFLUIDICS 2023; 17:051504. [PMID: 37869745 PMCID: PMC10589053 DOI: 10.1063/5.0163970] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/02/2023] [Indexed: 10/24/2023]
Abstract
In recent decades, there has been significant interest in inertial microfluidics due to its high throughput, ease of fabrication, and no need for external forces. The focusing efficiency of inertial microfluidic systems relies entirely on the geometrical features of microchannels because hydrodynamic forces (inertial lift forces and Dean drag forces) are the main driving forces in inertial microfluidic devices. In the past few years, novel microchannel structures have been propounded to improve particle manipulation efficiency. However, the fabrication of these unconventional structures has remained a serious challenge. Although researchers have pushed forward the frontiers of microfabrication technologies, the fabrication techniques employed for inertial microfluidics have not been discussed comprehensively. This review introduces the microfabrication approaches used for creating inertial microchannels, including photolithography, xurography, laser cutting, micromachining, microwire technique, etching, hot embossing, 3D printing, and injection molding. The advantages and disadvantages of these methods have also been discussed. Then, the techniques are reviewed regarding resolution, structures, cost, and materials. This review provides a thorough insight into the manufacturing methods of inertial microchannels, which could be helpful for future studies to improve the harvesting yield and resolution by choosing a proper fabrication technique.
Collapse
Affiliation(s)
- Zohreh Akbari
- Department of Mechanical Engineering, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - Sheyda Mirjalali
- School of Engineering, Macquarie University, Sydney, New South Wales, Australia
| | - Behrouz Aghajanloo
- School of Engineering, Macquarie University, Sydney, New South Wales, Australia
| |
Collapse
|
13
|
Esan A, Vanholsbeeck F, Swift S, McGoverin CM. Continuous separation of bacterial cells from large debris using a spiral microfluidic device. BIOMICROFLUIDICS 2023; 17:044104. [PMID: 37576440 PMCID: PMC10415021 DOI: 10.1063/5.0159254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023]
Abstract
With the global increase in food exchange, rapid identification and enumeration of bacteria has become crucial for protecting consumers from bacterial contamination. Efficient analysis requires the separation of target particles (e.g., bacterial cells) from food and/or sampling matrices to prevent matrix interference with the detection and analysis of target cells. However, studies on the separation of bacteria-sized particles and defined particles, such as bacterial cells, from heterogeneous debris, such as meat swab suspensions, are limited. In this study, we explore the use of passive-based inertial microfluidics to separate bacterial cells from debris, such as fascia, muscle tissues, and cotton fibers, extracted from ground meat and meat swabs-a novel approach demonstrated for the first time. Our objective is to evaluate the recovery efficiency of bacterial cells from large debris obtained from ground meat and meat swab suspensions using a spiral microfluidic device. In this study, we establish the optimal flow rates and Dean number for continuous bacterial cell and debris separation and a methodology to determine the percentage of debris removed from the sample suspension. Our findings demonstrate an average recovery efficiency of ∼ 80% for bacterial cells separated from debris in meat swab suspensions, while the average recovery efficiency from ground beef suspensions was ∼ 70%. Furthermore, approximately 50% of the debris in the ground meat suspension were separated from bacterial cells.
Collapse
Affiliation(s)
| | | | - Simon Swift
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
14
|
Farahinia A, Zhang W, Badea I. Recent Developments in Inertial and Centrifugal Microfluidic Systems along with the Involved Forces for Cancer Cell Separation: A Review. SENSORS (BASEL, SWITZERLAND) 2023; 23:s23115300. [PMID: 37300027 DOI: 10.3390/s23115300] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/23/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023]
Abstract
The treatment of cancers is a significant challenge in the healthcare context today. Spreading circulating tumor cells (CTCs) throughout the body will eventually lead to cancer metastasis and produce new tumors near the healthy tissues. Therefore, separating these invading cells and extracting cues from them is extremely important for determining the rate of cancer progression inside the body and for the development of individualized treatments, especially at the beginning of the metastasis process. The continuous and fast separation of CTCs has recently been achieved using numerous separation techniques, some of which involve multiple high-level operational protocols. Although a simple blood test can detect the presence of CTCs in the blood circulation system, the detection is still restricted due to the scarcity and heterogeneity of CTCs. The development of more reliable and effective techniques is thus highly desired. The technology of microfluidic devices is promising among many other bio-chemical and bio-physical technologies. This paper reviews recent developments in the two types of microfluidic devices, which are based on the size and/or density of cells, for separating cancer cells. The goal of this review is to identify knowledge or technology gaps and to suggest future works.
Collapse
Affiliation(s)
- Alireza Farahinia
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Wenjun Zhang
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Ildiko Badea
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
15
|
Schellenberg J, Dehne M, Lange F, Scheper T, Solle D, Bahnemann J. Establishment of a Perfusion Process with Antibody-Producing CHO Cells Using a 3D-Printed Microfluidic Spiral Separator with Web-Based Flow Control. Bioengineering (Basel) 2023; 10:656. [PMID: 37370588 PMCID: PMC10295792 DOI: 10.3390/bioengineering10060656] [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: 04/24/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Monoclonal antibodies are increasingly dominating the market for human therapeutic and diagnostic agents. For this reason, continuous methods-such as perfusion processes-are being explored and optimized in an ongoing effort to increase product yields. Unfortunately, many established cell retention devices-such as tangential flow filtration-rely on membranes that are prone to clogging, fouling, and undesirable product retention at high cell densities. To circumvent these problems, in this work, we have developed a 3D-printed microfluidic spiral separator for cell retention, which can readily be adapted and replaced according to process conditions (i.e., a plug-and-play system) due to the fast and flexible 3D printing technique. In addition, this system was also expanded to include automatic flushing, web-based control, and notification via a cellphone application. This set-up constitutes a proof of concept that was successful at inducing a stable process operation at a viable cell concentration of 10-17 × 106 cells/mL in a hybrid mode (with alternating cell retention and cell bleed phases) while significantly reducing both shear stress and channel blockage. In addition to increasing efficiency to nearly 100%, this microfluidic device also improved production conditions by successfully separating dead cells and cell debris and increasing cell viability within the bioreactor.
Collapse
Affiliation(s)
- Jana Schellenberg
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany; (J.S.); (M.D.); (F.L.); (T.S.)
| | - Michaela Dehne
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany; (J.S.); (M.D.); (F.L.); (T.S.)
- Institute of Physics, University of Augsburg, Universitätsstr. 1, 86159 Augsburg, Germany
| | - Ferdinand Lange
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany; (J.S.); (M.D.); (F.L.); (T.S.)
| | - Thomas Scheper
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany; (J.S.); (M.D.); (F.L.); (T.S.)
| | - Dörte Solle
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany; (J.S.); (M.D.); (F.L.); (T.S.)
| | - Janina Bahnemann
- Institute of Physics, University of Augsburg, Universitätsstr. 1, 86159 Augsburg, Germany
| |
Collapse
|
16
|
Abstract
The advancement of microfluidics has enabled numerous discoveries and technologies in life sciences. However, due to the lack of industry standards and configurability, the design and fabrication of microfluidic devices require highly skilled technicians. The diversity of microfluidic devices discourages biologists and chemists from applying this technique in their laboratories. Modular microfluidics, which integrates the standardized microfluidic modules into a whole, complex platform, brings the capability of configurability to conventional microfluidics. The exciting features, including portability, on-site deployability, and high customization motivate us to review the state-of-the-art modular microfluidics and discuss future perspectives. In this review, we first introduce the working mechanisms of the basic microfluidic modules and evaluate their feasibility as modular microfluidic components. Next, we explain the connection approaches among these microfluidic modules, and summarize the advantages of modular microfluidics over integrated microfluidics in biological applications. Finally, we discuss the challenge and future perspectives of modular microfluidics.
Collapse
Affiliation(s)
- Jialin Wu
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, China
- Nanophotonics Research Center, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, China
| | - Hui Fang
- Nanophotonics Research Center, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen, China
| | - Jun Zhang
- Queensland Micro and Nanotechnology Centre, Griffith University, Brisbane, QLD, 4111, Australia
| | - Sheng Yan
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
17
|
Analysis of circulating tumour cells separation in a curved microchannel under a high gravitational field. Chem Eng Sci 2023. [DOI: 10.1016/j.ces.2022.118235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
18
|
Shrestha J, Razavi Bazaz S, Ding L, Vasilescu S, Idrees S, Söderström B, Hansbro PM, Ghadiri M, Ebrahimi Warkiani M. Rapid separation of bacteria from primary nasal samples using inertial microfluidics. LAB ON A CHIP 2022; 23:146-156. [PMID: 36484411 DOI: 10.1039/d2lc00794k] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Microbial populations play a crucial role in human health and the development of many diseases. These diseases often arise from the explosive proliferation of opportunistic bacteria, such as those in the nasal cavity. Recently, there have been increases in the prevalence of these opportunistic pathogens displaying antibiotic resistance. Thus, the study of the nasal microbiota and its bacterial diversity is critical in understanding pathogenesis and developing microbial-based therapies for well-known and emerging diseases. However, the isolation and analysis of these populations for clinical study complicates the already challenging task of identifying and profiling potentially harmful bacteria. Existing methods are limited by low sample throughput, expensive labeling, and low recovery of bacteria with ineffective removal of cells and debris. In this study, we propose a novel microfluidic channel with a zigzag configuration for enhanced isolation and detection of bacteria from human clinical nasal swabs. This microfluidic zigzag channel separates the bacteria from epithelial cells and debris by size differential focusing. As such, pure bacterial cell fractions devoid of large contaminating debris or epithelial cells are obtained. DNA sequencing performed on the separated bacteria defines the diversity and species present. This novel method of bacterial separation is simple, robust, rapid, and cost-effective and has the potential to be used for the rapid identification of bacterial cell populations from clinical samples.
Collapse
Affiliation(s)
- Jesus Shrestha
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
- Woolcock Institute of Medical Research, Respiratory Technology Group, University of Sydney, Sydney, New South Wales 2037, Australia
| | - Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
| | - Lin Ding
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
| | - Steven Vasilescu
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
| | - Sobia Idrees
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney 2007, Australia
| | - Bill Söderström
- Australian Institute for Microbiology and Infection, Faculty of Science, University of Technology Sydney, New South Wales 2007, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney 2007, Australia
| | - Maliheh Ghadiri
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
- Woolcock Institute of Medical Research, Respiratory Technology Group, University of Sydney, Sydney, New South Wales 2037, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, New South Wales 2007, Australia
- Institute of Molecular Medicine, Sechenov First Moscow State University, Moscow 119991, Russia
| |
Collapse
|
19
|
Minh CN, Peerhossaini H, Jarrahi M. Phototactic microswimmers in pulsatile flow: Toward a novel harvesting method. BIOMICROFLUIDICS 2022; 16:054103. [PMID: 36186758 PMCID: PMC9519186 DOI: 10.1063/5.0097580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/19/2022] [Indexed: 06/16/2023]
Abstract
Phototactic behavior is coupled with pulsatile flow features to reveal the advantages of pulsation for separating motile algae cells in a double Y-microchannel. The underlying mechanism is as follows: during half of the pulsation cycle, when the flow rate is low, the phototactic microswimmers are mainly redirected by the external stimulation (light); while, during the rest of the cycle, the flow effects become dominant and the microswimmers are driven toward the desired outlet. The results show that in the absence of light source, the pulsatile flow has no advantage over the steady flow for separation, and the microswimmers have no preference between the exit channels; the separation index (SI) is around 50%. However, when the light is on, SI increases to 65% and 75% in the steady and pulsatile flows, respectively. Although the experiments are conducted on the well-known model alga, Chlamydomonas reinhardtii, a numerical simulation based on a simple model demonstrates that the idea can be extended to other active particles stimulated by an attractive or repulsive external field. Thus, the potential applications can go beyond algae harvesting to the control and enhancement of separation processes without using any mechanical component or chemical substance.
Collapse
|
20
|
Xu X, Huang X, Sun J, Chen J, Wu G, Yao Y, Zhou N, Wang S, Sun L. 3D-Stacked Multistage Inertial Microfluidic Chip for High-Throughput Enrichment of Circulating Tumor Cells. CYBORG AND BIONIC SYSTEMS 2022; 2022:9829287. [PMID: 38645277 PMCID: PMC11030111 DOI: 10.34133/2022/9829287] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 06/26/2022] [Indexed: 02/05/2023] Open
Abstract
Whether for cancer diagnosis or single-cell analysis, it remains a major challenge to isolate the target sample cells from a large background cell for high-efficiency downstream detection and analysis in an integrated chip. Therefore, in this paper, we propose a 3D-stacked multistage inertial microfluidic sorting chip for high-throughput enrichment of circulating tumor cells (CTCs) and convenient downstream analysis. In this chip, the first stage is a spiral channel with a trapezoidal cross-section, which has better separation performance than a spiral channel with a rectangular cross-section. The second and third stages adopt symmetrical square serpentine channels with different rectangular cross-section widths for further separation and enrichment of sample cells reducing the outlet flow rate for easier downstream detection and analysis. The multistage channel can separate 5 μm and 15 μm particles with a separation efficiency of 92.37% and purity of 98.10% at a high inlet flow rate of 1.3 mL/min. Meanwhile, it can separate tumor cells (SW480, A549, and Caki-1) from massive red blood cells (RBCs) with a separation efficiency of >80%, separation purity of >90%, and a concentration fold of ~20. The proposed work is aimed at providing a high-throughput sample processing system that can be easily integrated with flowing sample detection methods for rapid CTC analysis.
Collapse
Affiliation(s)
- X. Xu
- Ministry of Education Key Laboratory of RF Circuits and Systems, Hangzhou Dianzi University, Hangzhou, 310018 Zhejiang, China
| | - X. Huang
- Ministry of Education Key Laboratory of RF Circuits and Systems, Hangzhou Dianzi University, Hangzhou, 310018 Zhejiang, China
| | - J. Sun
- Ministry of Education Key Laboratory of RF Circuits and Systems, Hangzhou Dianzi University, Hangzhou, 310018 Zhejiang, China
| | - J. Chen
- Ministry of Education Key Laboratory of RF Circuits and Systems, Hangzhou Dianzi University, Hangzhou, 310018 Zhejiang, China
| | - G. Wu
- Institute for Translational Medicine, Zhejiang University, Hangzhou, 310029 Zhejiang, China
| | - Y. Yao
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Westlake University, Hangzhou, 310024 Zhejiang, China
| | - N. Zhou
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Westlake University, Hangzhou, 310024 Zhejiang, China
| | - S. Wang
- Institute for Translational Medicine, Zhejiang University, Hangzhou, 310029 Zhejiang, China
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
- Institute for Advanced Study, Chengdu University, Chengdu 610106, China
| | - L. Sun
- Ministry of Education Key Laboratory of RF Circuits and Systems, Hangzhou Dianzi University, Hangzhou, 310018 Zhejiang, China
| |
Collapse
|
21
|
Fu T, Li S, Wei L, Huang S, Xue Y, Cui K. Optimal whole blood dilution protocol for preprocessing samples prior to circulating tumor cell capture by size-based isolation. J Clin Lab Anal 2022; 36:e24524. [PMID: 35657145 PMCID: PMC9279958 DOI: 10.1002/jcla.24524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study compared whole blood dilution versus density gradient centrifugation for pre-processing blood samples prior to circulating tumor cell (CTC) capture on the efficiency of CTC separation by size-based isolation. MATERIALS AND METHODS Whole blood from a healthy volunteer spiked with SKBR3 cells was used to optimize the whole blood dilution protocol for sample volume, dilution ratio, and paraformaldehyde (PFA) concentration. Whole blood from healthy volunteers spiked with SKBR3, A549, or PC3 cells, and whole blood from patients with advanced gastric, esophageal, or liver cancer, was used to compare pre-processing by the optimal whole blood dilution protocol with density-gradient centrifugation. All statistical evaluations were performed using Student t test of the Statistical Package for Social Sciences (SPSS version 17.0). RESULTS In blood samples from healthy volunteers, spiked SKBR3 cell recovery rates were highest in 5 ml of whole blood, diluted with 2.5 ml buffer, and fixed with 0.2% PFA, and spiked SKBR3, A549, and PC3 cell recovery rates from 5 ml whole blood were significantly greater when using the optimized whole blood dilution protocol (87.67% ± 1.76%, 79.50% ± 0.50% and 71.83% ± 1.04%, respectively) compared to density-gradient centrifugation (46.83 ± 1.76%, 37.00 ± 1.50% and 41.00 ± 1.50%, respectively).
Collapse
Affiliation(s)
- Tianhao Fu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Sheng Li
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Liang Wei
- Wuhan YZY Medical Science & Technology Co., LtdWuhanChina
| | - Shaoyi Huang
- Wuhan YZY Medical Science & Technology Co., LtdWuhanChina
| | - Yan Xue
- Jinan Children's HospitalJinanChina
| | - Kai Cui
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| |
Collapse
|
22
|
Sierra-Agudelo J, Rodriguez-Trujillo R, Samitier J. Microfluidics for the Isolation and Detection of Circulating Tumor Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:389-412. [PMID: 35761001 DOI: 10.1007/978-3-031-04039-9_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nowadays, liquid biopsy represents one of the most promising techniques for early diagnosis, monitoring, and therapy screening of cancer. This novel methodology includes, among other techniques, the isolation, capture, and analysis of circulating tumor cells (CTCs). Nonetheless, the identification of CTC from whole blood is challenging due to their extremely low concentration (1-100 per ml of whole blood), and traditional methods result insufficient in terms of purity, recovery, throughput and/or viability of the processed sample. In this context, the development of microfluidic devices for detecting and isolating CTCs offers a wide range of new opportunities due to their excellent properties for cell manipulation and the advantages to integrate and bring different laboratory processes into the microscale improving the sensitivity, portability, reducing cost and time. This chapter explores current and recent microfluidic approaches that have been developed for the analysis and detection of CTCs, which involve cell capture methods based on affinity binding and label-free methods and detection based on electrical, chemical, and optical sensors. All the exposed technologies seek to overcome the limitations of commercial systems for the analysis and isolation of CTCs, as well as to provide extended analysis that will allow the development of novel and more efficient diagnostic tools.
Collapse
Affiliation(s)
- Jessica Sierra-Agudelo
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Romen Rodriguez-Trujillo
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain. .,Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain.
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| |
Collapse
|
23
|
Jeon H, Cremers C, Le D, Abell J, Han J. Multi-dimensional-double-spiral (MDDS) inertial microfluidic platform for sperm isolation directly from the raw semen sample. Sci Rep 2022; 12:4212. [PMID: 35273303 PMCID: PMC8913683 DOI: 10.1038/s41598-022-08042-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/01/2022] [Indexed: 12/19/2022] Open
Abstract
Here, we propose a fully-automated platform using a spiral inertial microfluidic device for standardized semen preparation that can process patient-derived semen samples with diverse fluidic conditions without any pre-washing steps. We utilized the multi-dimensional double spiral (MDDS) device to effectively isolate sperm cells from other non-sperm seminal cells (e.g., leukocytes) in the semen sample. The recirculation platform was employed to minimize sample dependency and achieve highly purified and concentrated (up to tenfold) sperm cells in a rapid and fully-automated manner (~ 10 min processing time for 50 mL of diluted semen sample). The clinical (raw) semen samples obtained from healthy donors were directly used without any pre-washing step to evaluate the developed separation platform, which showed excellent performance with ~ 80% of sperm cell recovery, and > 99.95% and > 98% removal of 10-μm beads (a surrogate for leukocytes) from low-viscosity and high-viscosity semen samples, respectively. We expect that the novel platform will be an efficient and automated tool to achieve purified sperm cells directly from raw semen samples for assisted reproductive technologies (ARTs) as an alternative to density centrifugation or swim-up methods, which often suffer from the low recovery of sperm cells and labor-intensive steps.
Collapse
Affiliation(s)
- Hyungkook Jeon
- Research Laboratory of Electronics, Massachusetts Institute of Technology (MIT), Cambridge, MA, 02139, USA
| | - Claudia Cremers
- Ohana Biosciences, 20 Acorn Park Dr, Cambridge, MA, 02140, USA
| | - Doris Le
- Ohana Biosciences, 20 Acorn Park Dr, Cambridge, MA, 02140, USA
| | - Justin Abell
- Ohana Biosciences, 20 Acorn Park Dr, Cambridge, MA, 02140, USA
| | - Jongyoon Han
- Research Laboratory of Electronics, Massachusetts Institute of Technology (MIT), Cambridge, MA, 02139, USA. .,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, 02139, USA. .,Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, 02139, USA.
| |
Collapse
|
24
|
Abdollahzadeh L, Seyfi Mazraeno M, Hosseini SN, Fazlali A, Khatami M. Application of a 3D printed miniaturized hydrocyclone in biopharmaceutical industry-numerical and experimental studies of yeast separation from fermentation culture media. Prep Biochem Biotechnol 2022; 53:31-39. [PMID: 35225162 DOI: 10.1080/10826068.2022.2035746] [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] [Indexed: 01/05/2023]
Abstract
Various industries ranging from water purification to pharmaceutical production have experienced multi separation steps that impose more process time and contamination possibility by batch operation. We propose a developed microfluidic particle sorter (miniaturized hydrocyclone) that adopts centrifugal force as it has ability to decline the number of separation steps and the risk of extrinsic contamination in continuous process. While biological industries have not relied on mini hydrocyclones considerably because of low efficiency and microfabrication difficulties, current work has been planned to conquer these obstacles. In this research, biomass separation from fermentation broth by 3 mm hydrocyclones was investigated. The effect of apex size, feed flow rate, hydrocyclone geometry were analyzed numerically in four mini-hydrocyclones. The most efficient mini-hydrocyclone was chosen to be made by elegant additive manufacturing technology and studied experimentally. The separation efficiency was achieved up to 90% while the concentration ratio of heavy stream (apex) to dilute stream (vortex finder) was reached more than twofold. The mini hydrocyclone performance in view of energy target was studied by Euler-Reynolds-Efficiency plots. The 4 μm cut size was achieved that is promising high throughput separation for biological particles.
Collapse
Affiliation(s)
- Laleh Abdollahzadeh
- Department of Hepatitis B Vaccine Production, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | | | - Seyed Nezamedin Hosseini
- Department of Hepatitis B Vaccine Production, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Fazlali
- Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, Iran
| | - Maryam Khatami
- Department of Hepatitis B Vaccine Production, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
25
|
Bordhan P, Razavi Bazaz S, Jin D, Ebrahimi Warkiani M. Advances and enabling technologies for phase-specific cell cycle synchronisation. LAB ON A CHIP 2022; 22:445-462. [PMID: 35076046 DOI: 10.1039/d1lc00724f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cell cycle synchronisation is the process of isolating cell populations at specific phases of the cell cycle from heterogeneous, asynchronous cell cultures. The process has important implications in targeted gene-editing and drug efficacy of cells and in studying cell cycle events and regulatory mechanisms involved in the cell cycle progression of multiple cell species. Ideally, cell cycle synchrony techniques should be applicable for all cell types, maintain synchrony across multiple cell cycle events, maintain cell viability and be robust against metabolic and physiological perturbations. In this review, we categorize cell cycle synchronisation approaches and discuss their operational principles and performance efficiencies. We highlight the advances and technological development trends from conventional methods to the more recent microfluidics-based systems. Furthermore, we discuss the opportunities and challenges for implementing high throughput cell synchronisation and provide future perspectives on synchronisation platforms, specifically hybrid cell synchrony modalities, to allow the highest level of phase-specific synchrony possible with minimal alterations in diverse types of cell cultures.
Collapse
Affiliation(s)
- Pritam Bordhan
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
- Institute for Biomedical Materials & Devices, Faculty of Science, University of Technology Sydney, New South Wales 2007, Australia
| | - Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
- Institute for Biomedical Materials & Devices, Faculty of Science, University of Technology Sydney, New South Wales 2007, Australia
| | - Dayong Jin
- Institute for Biomedical Materials & Devices, Faculty of Science, University of Technology Sydney, New South Wales 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia.
- Institute for Biomedical Materials & Devices, Faculty of Science, University of Technology Sydney, New South Wales 2007, Australia
| |
Collapse
|
26
|
Jeon H, Kwon T, Yoon J, Han J. Engineering a deformation-free plastic spiral inertial microfluidic system for CHO cell clarification in biomanufacturing. LAB ON A CHIP 2022; 22:272-285. [PMID: 34931631 DOI: 10.1039/d1lc00995h] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Inertial microfluidics has enabled many impactful high throughput applications. However, devices fabricated in soft elastomer (i.e., polydimethylsiloxane (PDMS)) suffer reliability issues due to significant deformation generated by the high pressure and flow rates in inertial microfluidics. In this paper, we demonstrated deformation-free and mass-producible plastic spiral inertial microfluidic devices for high-throughput cell separation applications. The design of deformable PDMS spiral devices was translated to their plastic version by compensating for the channel deformation in the PDMS devices, analyzed by numerical simulation and confocal imaging methods. The developed plastic spiral devices showed similar performance to their original PDMS devices for blood separation and Chinese hamster ovary (CHO) cell retention. Furthermore, using a multiplexed plastic spiral unit containing 100 spirals, we successfully demonstrated ultra-high-throughput cell clarification (at a processing rate of 1 L min-1) with a high cell-clarification efficiency of ∼99% (at the cell density changing from ∼2 to ∼10 × 106 cells mL-1). Benefitting from the continuous and clogging-free separation with an industry-level throughput, the cell clarification device could be a critical breakthrough for the production of therapeutic biologics such as antibodies or vaccines, impacting biomanufacturing in general.
Collapse
Affiliation(s)
- Hyungkook Jeon
- Research Laboratory of Electronics, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA.
| | - Taehong Kwon
- Research Laboratory of Electronics, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA.
| | - Junghyo Yoon
- Research Laboratory of Electronics, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA.
| | - Jongyoon Han
- Research Laboratory of Electronics, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| |
Collapse
|
27
|
Li X, Duan J, Qu Z, Wang J, Ji M, Zhang B. Continuous Particle Separation Driven by 3D Ag-PDMS Electrodes with Dielectric Electrophoretic Force Coupled with Inertia Force. MICROMACHINES 2022; 13:mi13010117. [PMID: 35056282 PMCID: PMC8780234 DOI: 10.3390/mi13010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/07/2022] [Accepted: 01/07/2022] [Indexed: 02/04/2023]
Abstract
Cell separation has become @important in biological and medical applications. Dielectrophoresis (DEP) is widely used due to the advantages it offers, such as the lack of a requirement for biological markers and the fact that it involves no damage to cells or particles. This study aimed to report a novel approach combining 3D sidewall electrodes and contraction/expansion (CEA) structures to separate three kinds of particles with different sizes or dielectric properties continuously. The separation was achieved through the interaction between electrophoretic forces and inertia forces. The CEA channel was capable of sorting particles with different sizes due to inertial forces, and also enhanced the nonuniformity of the electric field. The 3D electrodes generated a non-uniform electric field at the same height as the channels, which increased the action range of the DEP force. Finite element simulations using the commercial software, COMSOL Multiphysics 5.4, were performed to determine the flow field distributions, electric field distributions, and particle trajectories. The separation experiments were assessed by separating 4 µm polystyrene (PS) particles from 20 µm PS particles at different flow rates by experiencing positive and negative DEP. Subsequently, the sorting performances of the 4 µm PS particles, 20 µm PS particles, and 4 µm silica particles with different solution conductivities were observed. Both the numerical simulations and the practical particle separation displayed high separating efficiency (separation of 4 µm PS particles, 94.2%; separation of 20 µm PS particles, 92.1%; separation of 4 µm Silica particles, 95.3%). The proposed approach is expected to open a new approach to cell sorting and separating.
Collapse
Affiliation(s)
- Xiaohong Li
- Key Laboratory of Instrumentation Science & Dynamic Measurement Ministry of Education, Micro Nano Technology Research Center, North University of China, Taiyuan 030051, China; (X.L.); (J.D.); (Z.Q.); (J.W.); (M.J.)
- Taiyuan Institute of Technology, Taiyuan 030051, China
| | - Junping Duan
- Key Laboratory of Instrumentation Science & Dynamic Measurement Ministry of Education, Micro Nano Technology Research Center, North University of China, Taiyuan 030051, China; (X.L.); (J.D.); (Z.Q.); (J.W.); (M.J.)
| | - Zeng Qu
- Key Laboratory of Instrumentation Science & Dynamic Measurement Ministry of Education, Micro Nano Technology Research Center, North University of China, Taiyuan 030051, China; (X.L.); (J.D.); (Z.Q.); (J.W.); (M.J.)
| | - Jiayun Wang
- Key Laboratory of Instrumentation Science & Dynamic Measurement Ministry of Education, Micro Nano Technology Research Center, North University of China, Taiyuan 030051, China; (X.L.); (J.D.); (Z.Q.); (J.W.); (M.J.)
| | - Miaomiao Ji
- Key Laboratory of Instrumentation Science & Dynamic Measurement Ministry of Education, Micro Nano Technology Research Center, North University of China, Taiyuan 030051, China; (X.L.); (J.D.); (Z.Q.); (J.W.); (M.J.)
| | - Binzhen Zhang
- Key Laboratory of Instrumentation Science & Dynamic Measurement Ministry of Education, Micro Nano Technology Research Center, North University of China, Taiyuan 030051, China; (X.L.); (J.D.); (Z.Q.); (J.W.); (M.J.)
- Correspondence:
| |
Collapse
|
28
|
Tang H, Niu J, Jin H, Lin S, Cui D. Geometric structure design of passive label-free microfluidic systems for biological micro-object separation. MICROSYSTEMS & NANOENGINEERING 2022; 8:62. [PMID: 35685963 PMCID: PMC9170746 DOI: 10.1038/s41378-022-00386-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/27/2022] [Accepted: 03/18/2022] [Indexed: 05/05/2023]
Abstract
Passive and label-free microfluidic devices have no complex external accessories or detection-interfering label particles. These devices are now widely used in medical and bioresearch applications, including cell focusing and cell separation. Geometric structure plays the most essential role when designing a passive and label-free microfluidic chip. An exquisitely designed geometric structure can change particle trajectories and improve chip performance. However, the geometric design principles of passive and label-free microfluidics have not been comprehensively acknowledged. Here, we review the geometric innovations of several microfluidic schemes, including deterministic lateral displacement (DLD), inertial microfluidics (IMF), and viscoelastic microfluidics (VEM), and summarize the most creative innovations and design principles of passive and label-free microfluidics. We aim to provide a guideline for researchers who have an interest in geometric innovations of passive label-free microfluidics.
Collapse
Affiliation(s)
- Hao Tang
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai, 200240 China
| | - Jiaqi Niu
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai, 200240 China
| | - Han Jin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai, 200240 China
- National Engineering Research Center for Nanotechnology, Shanghai Jiao Tong University, 28 Jiangchuan Easternroad, Shanghai, 200241 China
| | - Shujing Lin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai, 200240 China
- National Engineering Research Center for Nanotechnology, Shanghai Jiao Tong University, 28 Jiangchuan Easternroad, Shanghai, 200241 China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai, 200240 China
- National Engineering Research Center for Nanotechnology, Shanghai Jiao Tong University, 28 Jiangchuan Easternroad, Shanghai, 200241 China
| |
Collapse
|
29
|
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: 15.5] [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
|
30
|
Xu X, Huang X, Sun J, Wang R, Yao J, Han W, Wei M, Chen J, Guo J, Sun L, Yin M. Recent progress of inertial microfluidic-based cell separation. Analyst 2021; 146:7070-7086. [PMID: 34761757 DOI: 10.1039/d1an01160j] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cell separation has consistently been a pivotal technology of sample preparation in biomedical research. Compared with conventional bulky cell separation technologies applied in the clinic, cell separation based on microfluidics can accurately manipulate the displacement of liquid or cells at the microscale, which has great potential in point-of-care testing (POCT) applications due to small device size, low cost, low sample consumption, and high operating accuracy. Among various microfluidic cell separation technologies, inertial microfluidics has attracted great attention due to its simple structure and high throughput. In recent years, many researchers have explored the principles and applications of inertial microfluidics and developed different channel structures, including straight channels, curved channels, and multistage channels. However, the recently developed multistage channels have not been discussed and classified in detail compared with more widely discussed straight and curved channels. Therefore, in this review, a comprehensive and detailed review of recent progress in the multistage channel is presented. According to the channel structure, the inertial microfluidic separation technology is divided into (i) straight channel, (ii) curved channel, (iii) composite channel, and (iv) integrated device. The structural development of straight and curved channels is discussed in detail. And based on straight and curved channels, the multistage cell separation structures are reviewed, with a special focus on a variety of latest structures and related innovations of composite and integrated channels. Finally, the future prospects for the existing challenges in the development of inertial microfluidic cell separation technology are presented.
Collapse
Affiliation(s)
- Xuefeng Xu
- Key Laboratory of RF Circuits and Systems, Ministry of Education, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Xiwei Huang
- Key Laboratory of RF Circuits and Systems, Ministry of Education, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Jingjing Sun
- Key Laboratory of RF Circuits and Systems, Ministry of Education, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Renjie Wang
- Key Laboratory of RF Circuits and Systems, Ministry of Education, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Jiangfan Yao
- Key Laboratory of RF Circuits and Systems, Ministry of Education, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Wentao Han
- Key Laboratory of RF Circuits and Systems, Ministry of Education, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Maoyu Wei
- Key Laboratory of RF Circuits and Systems, Ministry of Education, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Jin Chen
- Key Laboratory of RF Circuits and Systems, Ministry of Education, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Jinhong Guo
- School of Communication and Information Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Lingling Sun
- Key Laboratory of RF Circuits and Systems, Ministry of Education, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Ming Yin
- The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
31
|
Choe SW, Kim B, Kim M. Progress of Microfluidic Continuous Separation Techniques for Micro-/Nanoscale Bioparticles. BIOSENSORS 2021; 11:464. [PMID: 34821680 PMCID: PMC8615634 DOI: 10.3390/bios11110464] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 05/03/2023]
Abstract
Separation of micro- and nano-sized biological particles, such as cells, proteins, and nucleotides, is at the heart of most biochemical sensing/analysis, including in vitro biosensing, diagnostics, drug development, proteomics, and genomics. However, most of the conventional particle separation techniques are based on membrane filtration techniques, whose efficiency is limited by membrane characteristics, such as pore size, porosity, surface charge density, or biocompatibility, which results in a reduction in the separation efficiency of bioparticles of various sizes and types. In addition, since other conventional separation methods, such as centrifugation, chromatography, and precipitation, are difficult to perform in a continuous manner, requiring multiple preparation steps with a relatively large minimum sample volume is necessary for stable bioprocessing. Recently, microfluidic engineering enables more efficient separation in a continuous flow with rapid processing of small volumes of rare biological samples, such as DNA, proteins, viruses, exosomes, and even cells. In this paper, we present a comprehensive review of the recent advances in microfluidic separation of micro-/nano-sized bioparticles by summarizing the physical principles behind the separation system and practical examples of biomedical applications.
Collapse
Affiliation(s)
- Se-woon Choe
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea;
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea
| | - Bumjoo Kim
- Department of Mechanical Engineering and Automotive Engineering, Kongju National University, Cheonan 1223-24, Korea;
- Department of Future Convergence Engineering, Kongju National University, Cheonan 1223-24, Korea
| | - Minseok Kim
- Department of Mechanical System Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
- Department of Aeronautics, Mechanical and Electronic Convergence Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
| |
Collapse
|
32
|
Ding L, Moloudi R, Warkiani ME. Bioreactor-Based Adherent Cells Harvesting from Microcarriers with 3D Printed Inertial Microfluidics. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2436:257-266. [PMID: 34724188 DOI: 10.1007/7651_2021_444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Harvesting adherent cells from microcarriers has become one of the major challenges of the downstream bioprocessing at large scale the current method has high maintenance and operation cost, which are the results of frequent clogging, due to cell lysing effect and microcarrier cake formation on membrane-based technology. These problems hugely impede the adaptation of microcarriers technologies in large-scale cell culture and hampered the supply of cells to the clinical need. Here, we describe two 3D printing-based methods to fabricate inertial microfluidic devices for separating adherent cells from microcarriers which overcome the above-mentioned limitations. The spiral devices are employed to separate mesenchymal stem cells from the microcarriers with 99% microcarrier removal rate and 77% cell recovery rate in one round of separation.
Collapse
Affiliation(s)
- Lin Ding
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Reza Moloudi
- Skeletal Biology and Engineering Research Centre, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
33
|
Zhou Z, Chen Y, Zhu S, Liu L, Ni Z, Xiang N. Inertial microfluidics for high-throughput cell analysis and detection: a review. Analyst 2021; 146:6064-6083. [PMID: 34490431 DOI: 10.1039/d1an00983d] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Since it was first proposed in 2007, inertial microfluidics has been extensively studied in terms of theory, design, fabrication, and application. In recent years, with the rapid development of microfabrication technologies, a variety of channel structures that can focus, concentrate, separate, and capture bioparticles or fluids have been designed and manufactured to extend the range of potential biomedical applications of inertial microfluidics. Due to the advantages of high throughput, simplicity, and low device cost, inertial microfluidics is a promising candidate for rapid sample processing, especially for large-volume samples with low-abundance targets. As an approach to cellular sample pretreatment, inertial microfluidics has been widely employed to ensure downstream cell analysis and detection. In this review, a comprehensive summary of the application of inertial microfluidics for high-throughput cell analysis and detection is presented. According to application areas, the recent advances can be sorted into label-free cell mechanical phenotyping, sheathless flow cytometric counting, electrical impedance cytometer, high-throughput cellular image analysis, and other methods. Finally, the challenges and prospects of inertial microfluidics for cell analysis and detection are summarized.
Collapse
Affiliation(s)
- Zheng Zhou
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| | - Yao Chen
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| | - Shu Zhu
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| | - Linbo Liu
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| | - Zhonghua Ni
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| | - Nan Xiang
- School of Mechanical Engineering, and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
34
|
Kwon T, Choi K, Han J. Separation of Ultra-High-Density Cell Suspension via Elasto-Inertial Microfluidics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101880. [PMID: 34396694 DOI: 10.1002/smll.202101880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/24/2021] [Indexed: 05/20/2023]
Abstract
Separation of high-density suspension particles at high throughput is crucial for many chemical, biomedical, and environmental applications. In this study, elasto-inertial microfluidics is used to manipulate ultra-high-density cells to achieve stable equilibrium positions in microchannels, aided by the inherent viscoelasticity of high-density cell suspension. It is demonstrated that ultra-high-density Chinese hamster ovary cell suspension (>26 packed cell volume% (PCV%), >95 million cells mL-1 ) can be focused at distinct lateral equilibrium positions under high-flow-rate conditions (up to 10 mL min-1 ). The effect of flow rates, channel dimensions, and cell densities on this unique focusing behavior is studied. Cell clarification is further demonstrated using this phenomenon, from 29.7 PCV% (108.1 million cells mL-1 ) to 8.3 PCV% (33.2 million cells mL-1 ) with overall 72.1% reduction efficiency and 10 mL min-1 processing rate. This work explores an extreme case of elasto-inertial particle focusing where ultra-high-density culture suspension is efficiently manipulated at high throughput. This result opens up new opportunities for practical applications of high-particle-density suspension manipulation.
Collapse
Affiliation(s)
- Taehong Kwon
- Research Laboratory of Electronics, Massachusetts Institute of Technology, 50 Vassar Street, Cambridge, MA, 02142, USA
| | - Kyungyong Choi
- Research Laboratory of Electronics, Massachusetts Institute of Technology, 50 Vassar Street, Cambridge, MA, 02142, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 50 Vassar Street, Cambridge, MA, 02142, USA
| | - Jongyoon Han
- Research Laboratory of Electronics, Massachusetts Institute of Technology, 50 Vassar Street, Cambridge, MA, 02142, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 50 Vassar Street, Cambridge, MA, 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 50 Vassar Street, Cambridge, MA, 02142, USA
| |
Collapse
|
35
|
3D Printed Microfluidic Spiral Separation Device for Continuous, Pulsation-Free and Controllable CHO Cell Retention. MICROMACHINES 2021; 12:mi12091060. [PMID: 34577708 PMCID: PMC8470376 DOI: 10.3390/mi12091060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/20/2021] [Accepted: 08/27/2021] [Indexed: 11/16/2022]
Abstract
The development of continuous bioprocesses-which require cell retention systems in order to enable longer cultivation durations-is a primary focus in the field of modern process development. The flow environment of microfluidic systems enables the granular manipulation of particles (to allow for greater focusing in specific channel regions), which in turn facilitates the development of small continuous cell separation systems. However, previously published systems did not allow for separation control. Additionally, the focusing effect of these systems requires constant, pulsation-free flow for optimal operation, which cannot be achieved using ordinary peristaltic pumps. As described in this paper, a 3D printed cell separation spiral for CHO-K1 (Chinese hamster ovary) cells was developed and evaluated optically and with cell experiments. It demonstrated a high separation efficiency of over 95% at up to 20 × 106 cells mL-1. Control over inlet and outlet flow rates allowed the operator to adjust the separation efficiency of the device while in use-thereby enabling fine control over cell concentration in the attached bioreactors. In addition, miniaturized 3D printed buffer devices were developed that can be easily attached directly to the separation unit for usage with peristaltic pumps while simultaneously almost eradicating pump pulsations. These custom pulsation dampeners were closely integrated with the separator spiral lowering the overall dead volume of the system. The entire device can be flexibly connected directly to bioreactors, allowing continuous, pulsation-free cell retention and process operation.
Collapse
|
36
|
Combination of inertial focusing and magnetoporetic separation in a novel microdevice. KOREAN J CHEM ENG 2021. [DOI: 10.1007/s11814-021-0795-3] [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]
|
37
|
Liu Z, Screven R, Yu D, Boxer L, Myers MJ, Han J, Devireddy LR. Microfluidic Separation of Canine Adipose-Derived Mesenchymal Stromal Cells. Tissue Eng Part C Methods 2021; 27:445-461. [PMID: 34155926 DOI: 10.1089/ten.tec.2021.0082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are potential treatments for a variety of veterinary medical conditions. However, clinical trials have often fallen short of expectations, due in part to heterogeneity and lack of characterization of the MSCs. Identification and characterization of subpopulations within MSC cultures may improve those outcomes. Therefore, the functional heterogeneity of different-sized subpopulations of MSCs was evaluated. A high-throughput, biophysical, label-free microfluidic sorting approach was used to separate subpopulations of canine adipose-derived MSCs (Ad-MSCs) based on size for subsequent characterization, as well as to evaluate the impact of culture conditions on their functional heterogeneity. We found that culture-expanded canine Ad-MSCs comprise distinct subpopulations: larger MSCs (mean diameter of 18.6 ± 0.2 μm), smaller MSCs (mean diameter of 15.3 ± 0.2 μm), and intermediate MSCs (mean diameter of 16.9 ± 0.1 μm). In addition, proliferation characteristics, senescence, and differentiation potential of canine Ad-MSCs are also dependent on cell size. We observed that larger MSCs proliferate more slowly, senesce at earlier passages, and are inclined to differentiate into adipocytes compared with smaller MSCs. Most importantly, these size-dependent functions are also affected by the presence of serum in the culture medium, as well as time in culture. Cell surface staining for MSC-specific CD44 and CD90 antigens showed that all subpopulations of MSCs are indistinguishable, suggesting that this criterion is not relevant to define subpopulations of MSCs. Finally, transcriptome analysis showed differential gene expression between larger and smaller subpopulations of MSCs. Larger MSCs expressed genes involved in cellular senescence such as cyclin-dependent kinase inhibitor 1A and smaller MSCs expressed genes that promote cell growth [mechanistic target of rapamycin 1 (mTORC1) pathway] and cell proliferation [myelocytomatosis (myc), e2f targets]. These results suggest that different subpopulations of MSCs have specific properties. Impact statement Clinical trials of mesenchymal stromal cells (MSCs) from veterinary species have often fallen short of expectations, due in part to heterogeneity and lack of characterization of the MSCs. A high-throughput, biophysical, label-free microfluidic sorting approach was used to separate subpopulations of canine adipose-derived MSCs (Ad-MSCs) based on size for subsequent characterization. Proliferation characteristics, senescence, and differentiation potential of canine Ad-MSCs are also dependent on cell size. Cell surface staining for MSC-specific cell surface markers showed that all subpopulations of MSCs are indistinguishable, suggesting that this criterion is not relevant to define subpopulations of MSCs.
Collapse
Affiliation(s)
- Zhuoming Liu
- Division of Applied Veterinary Research, Center for Veterinary Medicine, U.S. Food and Drug Administration, Laurel, Maryland, USA
| | - Rudell Screven
- Division of Applied Veterinary Research, Center for Veterinary Medicine, U.S. Food and Drug Administration, Laurel, Maryland, USA
| | - Debbie Yu
- Micro/Nanofluidic BioMEMS Group, Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Lynne Boxer
- Office of New Animal Drug Evaluation, Center for Veterinary Medicine, U.S. Food and Drug Administration, Rockville, Maryland, USA
| | - Michael J Myers
- Division of Applied Veterinary Research, Center for Veterinary Medicine, U.S. Food and Drug Administration, Laurel, Maryland, USA
| | - Jongyoon Han
- Micro/Nanofluidic BioMEMS Group, Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Laxminarayana R Devireddy
- Division of Applied Veterinary Research, Center for Veterinary Medicine, U.S. Food and Drug Administration, Laurel, Maryland, USA
| |
Collapse
|
38
|
Wang Y, Nunna BB, Talukder N, Etienne EE, Lee ES. Blood Plasma Self-Separation Technologies during the Self-Driven Flow in Microfluidic Platforms. Bioengineering (Basel) 2021; 8:94. [PMID: 34356201 PMCID: PMC8301051 DOI: 10.3390/bioengineering8070094] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/19/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023] Open
Abstract
Blood plasma is the most commonly used biofluid in disease diagnostic and biomedical analysis due to it contains various biomarkers. The majority of the blood plasma separation is still handled with centrifugation, which is off-chip and time-consuming. Therefore, in the Lab-on-a-chip (LOC) field, an effective microfluidic blood plasma separation platform attracts researchers' attention globally. Blood plasma self-separation technologies are usually divided into two categories: active self-separation and passive self-separation. Passive self-separation technologies, in contrast with active self-separation, only rely on microchannel geometry, microfluidic phenomena and hydrodynamic forces. Passive self-separation devices are driven by the capillary flow, which is generated due to the characteristics of the surface of the channel and its interaction with the fluid. Comparing to the active plasma separation techniques, passive plasma separation methods are more considered in the microfluidic platform, owing to their ease of fabrication, portable, user-friendly features. We propose an extensive review of mechanisms of passive self-separation technologies and enumerate some experimental details and devices to exploit these effects. The performances, limitations and challenges of these technologies and devices are also compared and discussed.
Collapse
Affiliation(s)
- Yudong Wang
- Advanced Energy Systems and Microdevices Laboratory, Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA; (Y.W.); (B.B.N.); (N.T.); (E.E.E.)
| | - Bharath Babu Nunna
- Advanced Energy Systems and Microdevices Laboratory, Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA; (Y.W.); (B.B.N.); (N.T.); (E.E.E.)
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Cambridge, MA 02139, USA
| | - Niladri Talukder
- Advanced Energy Systems and Microdevices Laboratory, Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA; (Y.W.); (B.B.N.); (N.T.); (E.E.E.)
| | - Ernst Emmanuel Etienne
- Advanced Energy Systems and Microdevices Laboratory, Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA; (Y.W.); (B.B.N.); (N.T.); (E.E.E.)
| | - Eon Soo Lee
- Advanced Energy Systems and Microdevices Laboratory, Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA; (Y.W.); (B.B.N.); (N.T.); (E.E.E.)
| |
Collapse
|
39
|
Rodriguez-Mateos P, Ngamsom B, Dyer CE, Iles A, Pamme N. Inertial focusing of microparticles, bacteria, and blood in serpentine glass channels. Electrophoresis 2021; 42:2246-2255. [PMID: 34031893 DOI: 10.1002/elps.202100083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/11/2021] [Accepted: 05/18/2021] [Indexed: 11/08/2022]
Abstract
Early detection of pathogenic microorganisms is pivotal to diagnosis and prevention of health and safety crises. Standard methods for pathogen detection often rely on lengthy culturing procedures, confirmed by biochemical assays, leading to >24 h for a diagnosis. The main challenge for pathogen detection is their low concentration within complex matrices. Detection of blood-borne pathogens via techniques such as PCR requires an initial positive blood culture and removal of inhibitory blood components, reducing its potential as a diagnostic tool. Among different label-free microfluidic techniques, inertial focusing on microscale channels holds great promise for automation, parallelization, and passive continuous separation of particles and cells. This work presents inertial microfluidic manipulation of small particles and cells (1-10 μm) in curved serpentine glass channels etched at different depths (deep and shallow designs) that can be exploited for (1) bacteria preconcentration from biological samples and (2) bacteria-blood cell separation. In our shallow device, the ability to focus Escherichia coli into the channel side streams with high recovery (89% at 2.2× preconcentration factor) could be applied for bacteria preconcentration in urine for diagnosis of urinary tract infections. Relying on differential equilibrium positions of red blood cells and E. coli inside the deep device, 97% red blood cells were depleted from 1:50 diluted blood with 54% E. coli recovered at a throughput of 0.7 mL/min. Parallelization of such devices could process relevant volumes of 7 mL whole blood in 10 min, allowing faster sample preparation for downstream molecular diagnostics of bacteria present in bloodstream.
Collapse
Affiliation(s)
| | - Bongkot Ngamsom
- Department of Chemistry and Biochemistry, University of Hull, Hull, UK
| | | | - Alexander Iles
- Department of Chemistry and Biochemistry, University of Hull, Hull, UK
| | - Nicole Pamme
- Department of Chemistry and Biochemistry, University of Hull, Hull, UK
| |
Collapse
|
40
|
Kumar ARK, Shou Y, Chan B, L K, Tay A. Materials for Improving Immune Cell Transfection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007421. [PMID: 33860598 DOI: 10.1002/adma.202007421] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/08/2020] [Indexed: 06/12/2023]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy holds great promise for preventing and treating deadly diseases such as cancer. However, it remains challenging to transfect and engineer primary immune cells for clinical cell manufacturing. Conventional tools using viral vectors and bulk electroporation suffer from low efficiency while posing risks like viral transgene integration and excessive biological perturbations. Emerging techniques using microfluidics, nanoparticles, and high-aspect-ratio nanostructures can overcome these challenges, and on top of that, provide universal and high-throughput cargo delivery. Herein, the strengths and limitations of traditional and emerging materials for immune cell transfection, and commercial development of these tools, are discussed. To enhance the characterization of transfection techniques and uptake by the clinical community, a list of in vitro and in vivo assays to perform, along with relevant protocols, is recommended. The overall aim, herein, is to motivate the development of novel materials to meet rising demand in transfection for clinical CAR-T cell manufacturing.
Collapse
Affiliation(s)
- Arun R K Kumar
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Brian Chan
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Krishaa L
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| |
Collapse
|
41
|
Yin L, Au WY, Yu CC, Kwon T, Lai Z, Shang M, Warkiani ME, Rosche R, Lim CT, Han J. Miniature auto-perfusion bioreactor system with spiral microfluidic cell retention device. Biotechnol Bioeng 2021; 118:1951-1961. [PMID: 33559879 DOI: 10.1002/bit.27709] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/15/2021] [Accepted: 02/05/2021] [Indexed: 12/26/2022]
Abstract
Medium perfusion is critical in maintaining high cell concentration in cultures. The conventional membrane filtration method for medium exchange has been challenged by the fouling and clogging of the membrane filters in long-term cultures. In this study, we present a miniature auto-perfusion system that can be operated inside a common-size laboratory incubator. The system is equipped with a spiral microfluidic chip for cell retention to replace conventional membrane filters, which fundamentally overcomes the clogging and fouling problem. We showed that the system supported continuous perfusion culture of Chinese hamster ovary (CHO) cells in suspension up to 14 days without cell retention chip replacement. Compared to daily manual medium change, 25% higher CHO cell concentration can be maintained at an average auto-perfusion rate of 196 ml/day in spinner flask at 70 ml working volume (2.8 VVD). The auto-perfusion system also resulted in better cell quality at high concentrations, in terms of higher viability, more uniform and regular morphology, and fewer aggregates. We also demonstrated the potential application of the system for culturing mesenchymal stem cells on microcarriers. This miniature auto-perfusion system provides an excellent solution to maintain cell-favorable conditions and high cell concentration in small-scale cultures for research and clinical uses.
Collapse
Affiliation(s)
- Lu Yin
- Critical Analytics for Manufacturing of Personalized Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, Singapore, Singapore.,Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Wen Y Au
- Astute Water Pte. Ltd., Singapore, Singapore
| | - Chia C Yu
- Departments of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Taehong Kwon
- Departments of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Zhangxing Lai
- Department of Mechanical Engineering, National University of Singapore, Singapore, Singapore
| | - Menglin Shang
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Majid E Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Broadway, New South Wales, Australia
| | | | - Chwee T Lim
- Critical Analytics for Manufacturing of Personalized Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, Singapore, Singapore.,Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Mechanobology Institute, National University of Singapore, Singapore, Singapore.,Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, Singapore
| | - Jongyoon Han
- Critical Analytics for Manufacturing of Personalized Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, Singapore, Singapore.,Departments of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, Singapore, Singapore.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
42
|
|
43
|
|
44
|
Huang D, Man J, Jiang D, Zhao J, Xiang N. Inertial microfluidics: Recent advances. Electrophoresis 2020; 41:2166-2187. [PMID: 33027533 DOI: 10.1002/elps.202000134] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/19/2020] [Accepted: 10/02/2020] [Indexed: 02/24/2024]
Abstract
Inertial microfluidics has attracted significant attentions in last decade due to its superior advantages of high throughput, label- and external field-free operation, simplicity, and low cost. A wide variety of channel geometry designs were demonstrated for focusing, concentrating, isolating, or separating of various bioparticles such as blood components, circulating tumor cells, bacteria, and microalgae. In this review, we first briefly introduce the physics of inertial migration and Dean flow for allowing the readers with diverse backgrounds to have a better understanding of the fundamental mechanisms of inertial microfluidics. Then, we present a comprehensive review of the recent advances and applications of inertial microfluidic devices according to different channel geometries ranging from straight channels, curved channels to contraction-expansion-array channels. Finally, the challenges and future perspective of inertial microfluidics are discussed. Owing to its superior benefit for particle manipulation, the inertial microfluidics will play a more important role in biology and medicine applications.
Collapse
Affiliation(s)
- Di Huang
- College of Mechanical and Electrical Engineering, China University of Mining and Technology, Xuzhou, P. R. China
- Jiangsu Province and Education Ministry Co-sponsored Collaborative Innovation Center of Intelligent Mining Equipment, China University of Mining and Technology, Xuzhou, P. R. China
| | - Jiaxiang Man
- College of Mechanical and Electrical Engineering, China University of Mining and Technology, Xuzhou, P. R. China
- Jiangsu Province and Education Ministry Co-sponsored Collaborative Innovation Center of Intelligent Mining Equipment, China University of Mining and Technology, Xuzhou, P. R. China
| | - Di Jiang
- School of Mechanical and Electronic Engineering, Nanjing Forestry University, Nanjing, P. R. China
| | - Jiyun Zhao
- College of Mechanical and Electrical Engineering, China University of Mining and Technology, Xuzhou, P. R. China
- Jiangsu Province and Education Ministry Co-sponsored Collaborative Innovation Center of Intelligent Mining Equipment, China University of Mining and Technology, Xuzhou, P. R. China
| | - Nan Xiang
- School of Mechanical Engineering, Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing, P. R. China
| |
Collapse
|
45
|
Tsou PH, Chiang PH, Lin ZT, Yang HC, Song HL, Li BR. Rapid purification of lung cancer cells in pleural effusion through spiral microfluidic channels for diagnosis improvement. LAB ON A CHIP 2020; 20:4007-4015. [PMID: 32966477 DOI: 10.1039/d0lc00663g] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Lung cancer is one of the leading causes of death worldwide. Fifteen percent of lung cancer patients will present with malignant pleural effusion initially, and up to 50% will have malignant pleural effusion throughout the course of the disease. In this study, we developed a spiral microfluidic device that can rapidly isolate cancer cells and improve their purity through fluid dynamics. This label-free, high-throughput device continuously isolates cancer cells and other unrelated molecules from pleural effusion. Most of the background cells that affect interpretation are flushed to outlets 1 to 3, and cancer cells are hydrodynamically concentrated to outlet 4, with 90% of lung cancer cells flowing to this outlet. After processing, the purity of cancer cells identified in pleural effusion by CD45 and epithelial cell adhesion molecule (EpCAM) antibodies in flow cytometry will be increased by 6 to 24 times. The microfluidic device presented here has the advantages of rapid processing and low cost, which are conducive to rapid and accurate clinical diagnosis.
Collapse
Affiliation(s)
- Ping-Hsien Tsou
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | | | | | | | | | | |
Collapse
|
46
|
Syed MS, Mirakhorli F, Marquis C, Taylor RA, Warkiani ME. Particle movement and fluid behavior visualization using an optically transparent 3D-printed micro-hydrocyclone. BIOMICROFLUIDICS 2020; 14:064106. [PMID: 33269035 PMCID: PMC7679180 DOI: 10.1063/5.0025391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/04/2020] [Indexed: 05/03/2023]
Abstract
A hydrocyclone is a macroscale separation device employed in various industries, with many advantages, including high-throughput and low operational costs. Translating these advantages to microscale has been a challenge due to the microscale fabrication limitations that can be surmounted using 3D printing technology. Additionally, it is difficult to simulate the performance of real 3D-printed micro-hydrocyclones because of turbulent eddies and the deviations from the design due to printing resolution. To address these issues, we propose a new experimental method for the direct observation of particle motion in 3D printed micro-hydrocyclones. To do so, wax 3D printing and soft lithography were used in combination to construct a transparent micro-hydrocyclone in a single block of polydimethylsiloxane. A high-speed camera and fluorescent particles were employed to obtain clear in situ images and to confirm the presence of the vortex core. To showcase the use of this method, we demonstrate that a well-designed device can achieve a 95% separation efficiency for a sample containing a mixture of (desired) stem cells and (undesired) microcarriers. Overall, we hope that the proposed method for the direct visualization of particle trajectories in micro-hydrocyclones will serve as a tool, which can be leveraged to accelerate the development of micro-hydrocyclones for biomedical applications.
Collapse
Affiliation(s)
- Maira Shakeel Syed
- School of Mechanical and Manufacturing Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Fateme Mirakhorli
- School of Biomedical Engineering, University of Technology Sydney, NSW 2007, Australia
| | - Christopher Marquis
- School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney, NSW 2052, Australia
| | | | | |
Collapse
|
47
|
Tang W, Zhu S, Jiang D, Zhu L, Yang J, Xiang N. Channel innovations for inertial microfluidics. LAB ON A CHIP 2020; 20:3485-3502. [PMID: 32910129 DOI: 10.1039/d0lc00714e] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Inertial microfluidics has gained significant attention since first being proposed in 2007 owing to the advantages of simplicity, high throughput, precise manipulation, and freedom from an external field. Superior performance in particle focusing, filtering, concentrating, and separating has been demonstrated. As a passive technology, inertial microfluidics technology relies on the unconventional use of fluid inertia in an intermediate Reynolds number range to induce inertial migration and secondary flow, which depend directly on the channel structure, leading to particle migration to the lateral equilibrium position or trapping in a specific cavity. With the advances in micromachining technology, many channel structures have been designed and fabricated in the past decade to explore the fundamentals and applications of inertial microfluidics. However, the channel innovations for inertial microfluidics have not been discussed comprehensively. In this review, the inertial particle manipulations and underlying physics in conventional channels, including straight, spiral, sinusoidal, and expansion-contraction channels, are briefly described. Then, recent innovations in channel structure for inertial microfluidics, especially channel pattern modification and unconventional cross-sectional shape, are reviewed. Finally, the prospects for future channel innovations in inertial microfluidic chips are also discussed. The purpose of this review is to provide guidance for the continued study of innovative channel designs to improve further the accuracy and throughput of inertial microfluidics.
Collapse
Affiliation(s)
- Wenlai Tang
- School of Electrical and Automation Engineering, Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing, Nanjing Normal University, Nanjing, 210023, China.
| | | | | | | | | | | |
Collapse
|
48
|
Thurgood P, Suarez SA, Pirogova E, Jex AR, Peter K, Baratchi S, Khoshmanesh K. Tunable Harmonic Flow Patterns in Microfluidic Systems through Simple Tube Oscillation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003612. [PMID: 33006247 DOI: 10.1002/smll.202003612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/29/2020] [Indexed: 06/11/2023]
Abstract
Generation of tunable harmonic flows at low cost in microfluidic systems is a persistent and significant obstacle to this field, substantially limiting its potential to address major scientific questions and applications. This work introduces a simple and elegant way to overcome this obstacle. Harmonic flow patterns can be generated in microfluidic structures by simply oscillating the inlet tubes. Complex rib and vortex patterns can be dynamically modulated by changing the frequency and magnitude of tube oscillation and the viscosity of liquid. Highly complex rib patterns and synchronous vortices can be generated in serially connected microfluidic chambers. Similar dynamic patterns can be generated using whole or diluted blood samples without damaging the sample. This method offers unique opportunities for studying complex fluids and soft materials, chemical synthesis of various compounds, and mimicking harmonic flows in biological systems using compact, tunable, and low-cost devices.
Collapse
Affiliation(s)
- Peter Thurgood
- School of Engineering, RMIT University, Melbourne, VIC, 3000, Australia
| | | | - Elena Pirogova
- School of Engineering, RMIT University, Melbourne, VIC, 3000, Australia
| | - Aaron R Jex
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia and Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia
| | | |
Collapse
|
49
|
Jeon H, Jundi B, Choi K, Ryu H, Levy BD, Lim G, Han J. Fully-automated and field-deployable blood leukocyte separation platform using multi-dimensional double spiral (MDDS) inertial microfluidics. LAB ON A CHIP 2020; 20:3612-3624. [PMID: 32990714 DOI: 10.1039/d0lc00675k] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A fully-automated and portable leukocyte separation platform was developed based on a new type of inertial microfluidic device, multi-dimensional double spiral (MDDS) device, as an alternative to centrifugation. By combining key innovations in inertial microfluidic device designs and check-valve-based recirculation processes, highly purified and concentrated WBCs (up to >99.99% RBC removal, ∼80% WBC recovery, >85% WBC purity, and ∼12-fold concentrated WBCs compared to the input sample) were achieved in less than 5 minutes, with high reliability and repeatability (coefficient of variation, CV < 5%). Using this, one can harvest up to 0.4 million of intact WBCs from 50 μL of human peripheral blood (50 μL), without any cell damage or phenotypic changes in a fully-automated operation. Alternatively, hand-powered operation is demonstrated with comparable separation efficiency and speed, which eliminates the need for electricity altogether for truly field-friendly sample preparation. The proposed platform is therefore highly deployable for various point-of-care applications, including bedside assessment of the host immune response and blood sample processing in resource-limited environments.
Collapse
Affiliation(s)
- Hyungkook Jeon
- Research Laboratory of Electronics, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. and Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Bakr Jundi
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kyungyong Choi
- Research Laboratory of Electronics, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. and Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Hyunryul Ryu
- Research Laboratory of Electronics, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA.
| | - Bruce D Levy
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Geunbae Lim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Jongyoon Han
- Research Laboratory of Electronics, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. and Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA and Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| |
Collapse
|
50
|
Lee D, Choi YH, Lee W. Enhancement of inflection point focusing and rare-cell separations from untreated whole blood. LAB ON A CHIP 2020; 20:2861-2871. [PMID: 32647850 DOI: 10.1039/d0lc00309c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Inertial microfluidic systems have been widely used for particle or cell separation applications, especially for rare-cell enrichment and separation from blood due to the high throughput and simplicity of the systems. However, most of the separation techniques using inertial microfluidic systems require dilution of blood samples or RBC lysis to achieve a high separation efficiency, which can adversely affect the throughput and/or analysis of the collected sample. We developed a cell separation technique compatible with untreated whole blood by inflection point focusing, that is, an inertial focusing phenomenon toward inflection points of the velocity profile. We generated a strong shear-gradient lift force by varying the velocity profile in a channel cross-section with the combined effect of the channel geometry and the co-flows of two liquids with different viscosities. The natural viscosity of blood leads to a lagging flow region in the middle of the channel where larger particles and cells can be extracted to the side flow region, which enables a highly efficient separation scheme with an unprecedented high throughput.
Collapse
Affiliation(s)
- Dongwoo Lee
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| | | | | |
Collapse
|