1
|
Tian Z, Wang X, Chen J. On-chip dielectrophoretic single-cell manipulation. MICROSYSTEMS & NANOENGINEERING 2024; 10:117. [PMID: 39187499 PMCID: PMC11347631 DOI: 10.1038/s41378-024-00750-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/07/2024] [Accepted: 07/07/2024] [Indexed: 08/28/2024]
Abstract
Bioanalysis at a single-cell level has yielded unparalleled insight into the heterogeneity of complex biological samples. Combined with Lab-on-a-Chip concepts, various simultaneous and high-frequency techniques and microfluidic platforms have led to the development of high-throughput platforms for single-cell analysis. Dielectrophoresis (DEP), an electrical approach based on the dielectric property of target cells, makes it possible to efficiently manipulate individual cells without labeling. This review focusses on the engineering designs of recent advanced microfluidic designs that utilize DEP techniques for multiple single-cell analyses. On-chip DEP is primarily effectuated by the induced dipole of dielectric particles, (i.e., cells) in a non-uniform electric field. In addition to simply capturing and releasing particles, DEP can also aid in more complex manipulations, such as rotation and moving along arbitrary predefined routes for numerous applications. Correspondingly, DEP electrodes can be designed with different patterns to achieve different geometric boundaries of the electric fields. Since many single-cell analyses require isolation and compartmentalization of individual cells, specific microstructures can also be incorporated into DEP devices. This article discusses common electrical and physical designs of single-cell DEP microfluidic devices as well as different categories of electrodes and microstructures. In addition, an up-to-date summary of achievements and challenges in current designs, together with prospects for future design direction, is provided.
Collapse
Affiliation(s)
- Zuyuan Tian
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB, T6G 1H9, Canada
| | - Xihua Wang
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB, T6G 1H9, Canada
| | - Jie Chen
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB, T6G 1H9, Canada.
- Academy for Engineering & Technology, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
2
|
Yang X, Liang Z, Luo Y, Yuan X, Cai Y, Yu D, Xing X. Single-cell impedance cytometry of anticancer drug-treated tumor cells exhibiting mitotic arrest state to apoptosis using low-cost silver-PDMS microelectrodes. LAB ON A CHIP 2023; 23:4848-4859. [PMID: 37860975 DOI: 10.1039/d3lc00459g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Chemotherapeutic drugs such as paclitaxel and vinblastine interact with microtubules and thus induce complex cell states of mitosis arrest at the G2/M phase followed by apoptosis dependent on drug exposure time and concentration. Microfluidic impedance cytometry (MIC), as a label-free and high-throughput technology for single-cell analysis, has been applied for viability assay of cancer cells post drug exposure at fixed time and dosage, yet verification of this technique for varied tumor cell states after anticancer drug treatment remains a challenge. Here we present a novel MIC device and for the first time perform impedance cytometry on carcinoma cells exhibiting progressive states of G2/M arrest followed by apoptosis related to drug concentration and exposure time, after treatments with paclitaxel and vinblastine, respectively. Our results from impedance cytometry reveal increased amplitude and negative phase shift at low frequency as well as higher opacity for HeLa cells under G2/M mitotic arrest compared to untreated cells. The cells under apoptosis, on the other hand, exhibit opposite changes in these electrical parameters. Therefore, the impedance features differentiate the HeLa cells under progressive states post anticancer drug treatment. We also demonstrate that vinblastine poses a more potent drug effect than paclitaxel especially at low concentrations. Our device is fabricated using a unique sacrificial layer-free soft lithography process as compared to the existing MIC device, which gives rise to readily aligned parallel microelectrodes made of silver-PDMS embedded in PDMS channel sidewalls with one molding step. Our results uncover the potential of the MIC device, with a fairly simple and low-cost fabrication process, for cellular state screening in anticancer drug therapy.
Collapse
Affiliation(s)
- Xinlong Yang
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| | - Ziheng Liang
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| | - Yuan Luo
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xueyuan Yuan
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| | - Yao Cai
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| | - Duli Yu
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| | - Xiaoxing Xing
- College of Information Science and Technology, Beijing University of Chemical Technology, No. 15 North 3rd Ring Rd., Beijing, 100029, China.
| |
Collapse
|
3
|
Rahimi A, Sharifi H, Li PCH. Cytosolic Calcium Measurement Utilizing a Single-Cell Biochip to Study the Effect of Curcumin and Resveratrol on a Single Glioma Cell. Methods Mol Biol 2023; 2689:13-25. [PMID: 37430043 DOI: 10.1007/978-1-0716-3323-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
A microfluidic method has been developed for real-time measurement of the effects of curcumin on the intracellular calcium concentration in a single glioma cell (U87-MG). This method is based on quantitative fluorescence measurement of intracellular calcium in a cell selected in a single-cell biochip. This biochip consists of three reservoirs, three channels, and a V-shaped cell retention structure. Because of the adherent nature of glioma cells, a single cell can adhere within the aforementioned V-shaped structure. The single-cell calcium measurement will minimize cell damage caused by conventional cell calcium assay methods. Previous studies have shown that curcumin increased cytosolic calcium in glioma cells using the fluorescent dye: Fluo-4. So in this study, the effects of 5 μM and 10 μM solutions of curcumin on the increases of cytosolic calcium in a single glioma cell have been measured. Moreover, the effects of 100 μM and 200 μM of resveratrol are measured. At the final stage of the experiments, ionomycin was used to increase the intracellular calcium to the highest possible level due to dye saturation. It has been demonstrated that microfluidic cell calcium measurement is a real-time cytosolic assay that requires small quantities of reagent, which will have potential uses for drug discovery.
Collapse
Affiliation(s)
- Abolfazl Rahimi
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Hamide Sharifi
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Paul C H Li
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada.
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
4
|
Cai J, Cao YB, Leung ELH. Detection of Single Non-small Cell Lung Cancer Cell Multidrug Resistance with Single-Cell Bioanalyzer. Methods Mol Biol 2023; 2689:1-11. [PMID: 37430042 DOI: 10.1007/978-1-0716-3323-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer death in the world. Despite the development of various lung cancer treatment methods, including surgery, radiation therapy, endocrine therapy, immunotherapy, and gene therapy, chemotherapy remains the most common approach for treating cancer. The risk of tumors acquiring resistance to chemotherapy remains a significant hurdle to the use of this approach for the successful treatment of various types of cancer. The majority of cancer-related deaths are related to metastasis. Circulating tumor cells (CTCs) are cells that have been detached from the primary tumor or have metastasized and entered the circulation. CTCs can cause metastases in various organs by reaching them through the bloodstream. The CTCs exist in peripheral blood as single cells or as oligoclonal clusters of tumor cells along with platelets and lymphocytes. The detection of CTCs is an important component of liquid biopsy which aids in the diagnosis, treatment, and prognosis of cancer. Here, we describe a method for extracting CTCs from the tumor of patients and using the microfluidic single-cell technique to study the inhibition of multidrug resistance due to drug efflux on a single cancer cell, to propose novel methods that can provide clinicians with more appropriate choices in their diagnostic and treatment approaches.
Collapse
Affiliation(s)
- Jun Cai
- Faculty of Pharmacy and Food Science, Zhuhai College of Science and Technology, Zhuhai, China
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, China
| | - Ya-Bing Cao
- Department of Oncology, Kiang Wu Hospital, Macau, SAR, China
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Science, University of Macau, Macau, SAR, China.
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau, SAR, China.
| |
Collapse
|
5
|
Wang H, Zhang R, Yang D, Wang X. Discrimination of Multidrug Resistance in Cancer Cells Achieved Using Single-Cell Analysis. Methods Mol Biol 2023; 2689:95-106. [PMID: 37430049 DOI: 10.1007/978-1-0716-3323-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
The biophysical signatures of single cells, such as multidrug resistance (MDR), may easily change during their various disease states. Therefore, there is an ever-growing need for advanced methods to study and analyze the response of cancer cells to therapeutic intervention. To determine the cancer cells and responses to various cancer therapies, from a cell mortality perspective, we report a label-free and real-time method to monitor the in situ responses of ovarian cancer cells using a single-cell bioanalyzer (SCB). The SCB instrument was used to detect different ovarian cancer cells, such as NCI/ADR-RES cells, which are multidrug resistant (MDR), and non-MDR OVCAR-8 cells. The discrimination of ovarian cells has been achieved at the single-cell level by measuring drug accumulation quantitatively in real time, in which the accumulation is high in non-MDR single cells without drug efflux but is low in MDR single cells which are not efflux-free. The SCB was constructed as an inverted microscope for optical imaging and fluorescent measurement of a single cell that was retained in a microfluidic chip. The single ovarian cancer cell retained in the chip offered sufficient fluorescent signals for the SCB to measure the accumulation of daunorubicin (DNR) in the single cell in the absence of cyclosporine A (CsA). The same cell allows us to detect the enhanced drug accumulation due to MDR modulation in the presence of CsA, which is the MDR inhibitor. The measurement of drug accumulation in a cell was achieved after it was captured in the chip for one hour, with the correction of background interference. The detection of accumulation enhancement due to MDR modulation by CsA was determined in terms of either the accumulation rate or enhanced concentration of DNR in the single cell (same cell, p < 0.01). It showed that with the effectiveness of efflux blocking by CsA, the intracellular DNR concentration in a single cell increased by threefold against its same cell control. This single-cell bioanalyzer instrument has the ability to discriminate MDR in different ovarian cells due to drug efflux in them by eliminating the interference of background fluorescence and by using the same cell control.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Chemistry and Chemical Engineering, Shanxi Datong University, Datong, Shanxi, China.
| | - Runxuan Zhang
- Department of Chemistry and Chemical Engineering, Shanxi Datong University, Datong, Shanxi, China
| | - Di Yang
- Department of Chemistry and Chemical Engineering, Shanxi Datong University, Datong, Shanxi, China
| | - Xin Wang
- Department of Chemistry and Chemical Engineering, Shanxi Datong University, Datong, Shanxi, China
| |
Collapse
|
6
|
Liu Y, Fan Z, Qiao L, Liu B. Advances in microfluidic strategies for single-cell research. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
7
|
Dozzo A, Galvin A, Shin JW, Scalia S, O'Driscoll CM, Ryan KB. Modelling acute myeloid leukemia (AML): What's new? A transition from the classical to the modern. Drug Deliv Transl Res 2022:10.1007/s13346-022-01189-4. [PMID: 35930221 DOI: 10.1007/s13346-022-01189-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 11/24/2022]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous malignancy affecting myeloid cells in the bone marrow (BM) but can spread giving rise to impaired hematopoiesis. AML incidence increases with age and is associated with poor prognostic outcomes. There has been a disconnect between the success of novel drug compounds observed in preclinical studies of hematological malignancy and less than exceptional therapeutic responses in clinical trials. This review aims to provide a state-of-the-art overview on the different preclinical models of AML available to expand insights into disease pathology and as preclinical screening tools. Deciphering the complex physiological and pathological processes and developing predictive preclinical models are key to understanding disease progression and fundamental in the development and testing of new effective drug treatments. Standard scaffold-free suspension models fail to recapitulate the complex environment where AML occurs. To this end, we review advances in scaffold/matrix-based 3D models and outline the most recent advances in on-chip technology. We also provide an overview of clinically relevant animal models and review the expanding use of patient-derived samples, which offer the prospect to create more "patient specific" screening tools either in the guise of 3D matrix models, microphysiological "organ-on-chip" tools or xenograft models and discuss representative examples.
Collapse
Affiliation(s)
| | - Aoife Galvin
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, 909 S. Wolcott Ave, Chicago, IL, 5091 COMRB, USA
| | - Santo Scalia
- Università degli Studi di Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Caitriona M O'Driscoll
- School of Pharmacy, University College Cork, Cork, Ireland.,SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, Cork, Ireland
| | - Katie B Ryan
- School of Pharmacy, University College Cork, Cork, Ireland. .,SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, Cork, Ireland.
| |
Collapse
|
8
|
Wang C, Hu W, Guan L, Yang X, Liang Q. Single-cell metabolite analysis on a microfluidic chip. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
9
|
Sharifi Noghabi H, Ahmed AQ, Li PCH. Intracellular Calcium Increases Due to Curcumin Measured Using a Single-Cell Biochip. ANAL LETT 2021. [DOI: 10.1080/00032719.2021.1888967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Hamideh Sharifi Noghabi
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Chemistry, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Abdul Q. Ahmed
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Paul C. H. Li
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
10
|
CUI J, LIU L, LI D, PIAO X. [Research progress in the application of external field separation technology and microfluidic technology in the separation of micro/nanoscales]. Se Pu 2021; 39:1157-1170. [PMID: 34677011 PMCID: PMC9404220 DOI: 10.3724/sp.j.1123.2020.12032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Indexed: 11/25/2022] Open
Abstract
The micro/nanoscales concerns interactions of entities with sizes in the range of 0.1-100 μm, such as biological cells, proteins, and particles. The separation of micro/nanoscales has been of immense significance for drug development, early-stage cancer detection, and customized precision therapy. For example, in recent years, rapid advances in the field of cell therapy have necessitated the development of simple and effective cell separation techniques. The isolation technique allows the collection of the required stem cells from complex samples. With the development of materials science and precision medicine, the separation of particles is also critical. The key physicochemical properties of micro/nanoscales are highly dependent on their specific size, shape, functional group, and mobility (based on the charged characteristics), which control their performance in the separation system. The current demand has made the simultaneous innovation of a separation system and an on-line detection platform imperative. Accordingly, various analytical methods involving the use of external forces, such as the flow field, magnetic field, electric field, and acoustic field, have been used for micro/nanoscales separation. Based on the physical and chemical parameters of the separation materials, these analytical methods can select different external force fields for micro/nanoscales separation, enabling real-time, accurate, efficient, and selective separation. However, at present, most of the applied field separation technologies require complex equipment and a large sample amount. This makes it crucial to miniaturize and integrate separation technologies for low-cost, rapid, and accurate micro/nanoscales separation. Microfluidic technology is a representative micro/nanoscales separation technology. It requires only a small volume of liquid, making it cost-effective; its high throughput enables continuous separation and analysis; its fast response in a microchip can allow many reactions; and finally, the miniaturization of the device allows the coupling of multiple detectors with the microchip. With the continuous growth and progress of microfluidic technology, some microfluidic platforms are now able to achieve the non-destructive separation of cells. They also enable on-line detection, offer high separation efficiency, and allow rapid separation for different biological samples. This review primarily summarizes recent advances in microfluidic chips based on flow field, electric field, magnetic field, acoustic field, and field separation technologies to improve the micro/nanoscales separation efficiency. This review also discusses the various external force fields of micro/nanoscales, such as a microparticle, single cell separation of substances classified introduction, and summarizes the advantages and disadvantages of their application and development. Finally, the prospect of the combined application of external field separation technology and microfluidic technology in the early screening of cancer cells and for precise micro/nanoscales separation is discussed, and the advantages and potential applications of the combined technology are proposed.
Collapse
|
11
|
Recent advances in single-cell analysis: Encapsulation materials, analysis methods and integrative platform for microfluidic technology. Talanta 2021; 234:122671. [PMID: 34364472 DOI: 10.1016/j.talanta.2021.122671] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 12/27/2022]
Abstract
Traditional cell biology researches on cell populations by their origin, tissue, morphology, and secretions. Because of the heterogeneity of cells, research at the single-cell level can obtain more accurate and comprehensive information that reflects the physiological state and process of the cell, increasing the significance of single-cell analysis. The application of single-cell analysis is faced with the problem of contaminated or damaged cells caused by cell sample transportation. Reversible encapsulation of a single cell can protect cells from the external environment and open the encapsulation shell to release cells, thus preserving cell integrity and improving extraction efficiency of analytes. Meanwhile, microfluidic single cell analysis (MSCA) exhibits integration, miniaturization, and high throughput, which can considerably improve the efficiency of single-cell analysis. The researches on single-cell reversible encapsulation materials, single-cell analysis methods, and the MSCA integration platform are analyzed and summarized in this review. The problems of single-cell viability, network of single-cell signal, and simultaneous detection of multiple biotoxins in food based on single-cell are proposed for future research.
Collapse
|
12
|
Demircan Yalçın Y, Töral TB, Sukas S, Yıldırım E, Zorlu Ö, Gündüz U, Külah H. A microfluidic device enabling drug resistance analysis of leukemia cells via coupled dielectrophoretic detection and impedimetric counting. Sci Rep 2021; 11:13193. [PMID: 34162990 PMCID: PMC8222334 DOI: 10.1038/s41598-021-92647-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/14/2021] [Indexed: 12/02/2022] Open
Abstract
We report the development of a lab-on-a-chip system, that facilitates coupled dielectrophoretic detection (DEP-D) and impedimetric counting (IM-C), for investigating drug resistance in K562 and CCRF-CEM leukemia cells without (immuno) labeling. Two IM-C units were placed upstream and downstream of the DEP-D unit for enumeration, respectively, before and after the cells were treated in DEP-D unit, where the difference in cell count gave the total number of trapped cells based on their DEP characteristics. Conductivity of the running buffer was matched the conductivity of cytoplasm of wild type K562 and CCRF-CEM cells. Results showed that DEP responses of drug resistant and wild type K562 cells were statistically discriminative (at p = 0.05 level) at 200 mS/m buffer conductivity and at 8.6 MHz working frequency of DEP-D unit. For CCRF-CEM cells, conductivity and frequency values were 160 mS/m and 6.2 MHz, respectively. Our approach enabled discrimination of resistant cells in a group by setting up a threshold provided by the conductivity of running buffer. Subsequent selection of drug resistant cells can be applied to investigate variations in gene expressions and occurrence of mutations related to drug resistance.
Collapse
Affiliation(s)
- Yağmur Demircan Yalçın
- Electrical and Electronics Engineering Department, Middle East Technical University, Ankara, Turkey. .,Mikro Biyosistemler A.Ş., Ankara, Turkey. .,Neuro-Nanoscale Engineering, Mechanical Engineering Department, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | | | - Sertan Sukas
- Mikro Biyosistemler A.Ş., Ankara, Turkey.,Mechanical Engineering Department, Microsystems Section, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Ender Yıldırım
- Mikro Biyosistemler A.Ş., Ankara, Turkey.,Mechanical Engineering Department, Middle East Technical University, Ankara, Turkey
| | - Özge Zorlu
- Mikro Biyosistemler A.Ş., Ankara, Turkey
| | - Ufuk Gündüz
- Biology Department, Middle East Technical University, Ankara, Turkey
| | - Haluk Külah
- Electrical and Electronics Engineering Department, Middle East Technical University, Ankara, Turkey.,Mikro Biyosistemler A.Ş., Ankara, Turkey.,METU MEMS Center, Ankara, Turkey
| |
Collapse
|
13
|
Ma Z, Zhao H, Shi L, Yu D, Guo X. Automatic medium exchange for micro-volume cell samples based on dielectrophoresis. Electrophoresis 2021; 42:1507-1515. [PMID: 33990980 DOI: 10.1002/elps.202000195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 03/27/2021] [Accepted: 04/20/2021] [Indexed: 11/06/2022]
Abstract
Cell medium exchange is a crucial step for life science and medicine. However, conventional cell medium exchange methods, including centrifuging and filtering, show limited ability for micro-volume cell samples such as circulating tumor cell (CTC) and circulating fetal cell (CFC). In this paper, we proposed an automatic medium exchange method for micro-volume cell samples based on dielectrophoresis (DEP) in microfluidic chip. Fresh medium and cell suspension were introduced into the microfluidic channel as the laminar flow. Plane stair-shaped interdigital electrodes were employed to drive the cells from the cell suspension to fresh media directly by DEP force. Additionally, we characterized and optimized the cell medium exchange according to both the theory and experiments. In the end, we achieved a 96.9% harvest rate of medium exchange for 0.3 μL samples containing micro-volume cells. For implementing an automatic continuous cell medium exchange, the proposed method can be integrated into the automatic cell processing system conveniently. Furthermore, the proposed method is a great candidate in micro-volume cell analysis and processing, cell electroporation, single cell sequencing, and other scenarios.
Collapse
Affiliation(s)
- Zhouyang Ma
- College of Information Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Hongwang Zhao
- School of Automobile and Traffic Engineering, Guilin University of Aerospace Technology, Guilin, Guangxi, P. R. China
| | - Liujia Shi
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, P. R. China
| | - Duli Yu
- College of Information Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China.,Beijing Advance Innovation Center for Soft Matter Science and Engineering, Beijing, P. R. China
| | - Xiaoliang Guo
- College of Information Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| |
Collapse
|
14
|
Pereiro I, Aubert J, Kaigala GV. Micro-scale technologies propel biology and medicine. BIOMICROFLUIDICS 2021; 15:021302. [PMID: 33948133 PMCID: PMC8081554 DOI: 10.1063/5.0047196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/05/2021] [Indexed: 05/05/2023]
Abstract
Historically, technology has been central to new discoveries in biology and progress in medicine. Among various technologies, microtechnologies, in particular, have had a prominent role in the revolution experienced by the life sciences in the last few decades, which will surely continue in the years to come. In this Perspective, we illustrate how microtechnologies, with a focus on microfluidics, have evolved in trends/waves to tackle the boundary of knowledge in the life sciences. We provide illustrative examples of technology-enabled biological breakthroughs and their current and future use in clinics. Finally, we take a closer look at the translational process to understand why the incorporation of new micro-scale technologies in medicine has been comparatively slow so far.
Collapse
|
15
|
Choi S, Lee H, Lee S, Park I, Kim YS, Key J, Lee SY, Yang S, Lee SW. A novel automatic segmentation and tracking method to measure cellular dielectrophoretic mobility from individual cell trajectories for high throughput assay. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 195:105662. [PMID: 32712504 DOI: 10.1016/j.cmpb.2020.105662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/09/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND AND OBJECTIVE The dielectrophoresis (DEP) technique is increasingly being recognised as a potentially valuable tool for non-contact manipulation of numerous cells as well as for biological single cell analysis with non-invasive characterisation of a cell's electrical properties. Several studies have attempted to track multiple cells to characterise their cellular DEP mobility. However, they encountered difficulties in simultaneously tracking the movement of a large number of individual cells in a bright-field image sequence because of interference from the background electrode pattern. Consequently, this present study aims to develop an automatic system for imaging-based characterisation of cellular DEP mobility, which enables the simultaneous tracking of several hundred of cells inside a microfluidic device. METHODS The proposed method for segmentation and tracking of cells consists of two main stages: pre-processing and particle centre localisation. In the pre-processing stage, background subtraction and contrast enhancement were performed to distinguish the cell region from the background image. In the particle centre localisation stage, the unmarked cell was automatically detected via graph-cut algorithm-based K-means clustering. RESULTS Our algorithm enabled segmentation and tracking of numerous Michigan Cancer Foundation-7 (MCF-7) cell trajectories while the DEP force was oscillated between positive and negative. The cell tracking accuracy and cell count capability was at least 90% of the total number of cells with the newly developed algorithm. In addition, the cross-over frequency was measured by analysing the segmented and tracked trajectory data of the cellular movements caused by the positive and negative DEP force. The measured cross-over frequency was compared with previous results. The multi-cellular movements investigation based on the measured cross-over frequency was repeated until the viability of cells was unchanged in the same environment as in a microfluidic device. The results were statistically consistent, indicating that the developed algorithm was reliable for the investigation of DEP cellular mobility. CONCLUSION This study developed a powerful platform to simultaneously measure the DEP-induced trajectories of numerous cells, and to investigate in a robust, efficient, and accurate manner the DEP properties at both the single cell and cell ensemble level.
Collapse
Affiliation(s)
- Seungyeop Choi
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Hyunwoo Lee
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Sena Lee
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Insu Park
- Holonyak Micro and Nanotechnology Laboratory, University of Illinois, Urbana, IL, USA
| | - Yoon Suk Kim
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26493, Republic of Korea
| | - Jaehong Key
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Sei Young Lee
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Sejung Yang
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea.
| | - Sang Woo Lee
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea.
| |
Collapse
|
16
|
Chen P, Li S, Guo Y, Zeng X, Liu BF. A review on microfluidics manipulation of the extracellular chemical microenvironment and its emerging application to cell analysis. Anal Chim Acta 2020; 1125:94-113. [PMID: 32674786 DOI: 10.1016/j.aca.2020.05.065] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022]
Abstract
Spatiotemporal manipulation of extracellular chemical environments with simultaneous monitoring of cellular responses plays an essential role in exploring fundamental biological processes and expands our understanding of underlying mechanisms. Despite the rapid progress and promising successes in manipulation strategies, many challenges remain due to the small size of cells and the rapid diffusion of chemical molecules. Fortunately, emerging microfluidic technology has become a powerful approach for precisely controlling the extracellular chemical microenvironment, which benefits from its integration capacity, automation, and high-throughput capability, as well as its high resolution down to submicron. Here, we summarize recent advances in microfluidics manipulation of the extracellular chemical microenvironment, including the following aspects: i) Spatial manipulation of chemical microenvironments realized by convection flow-, diffusion-, and droplet-based microfluidics, and surface chemical modification; ii) Temporal manipulation of chemical microenvironments enabled by flow switching/shifting, moving/flowing cells across laminar flows, integrated microvalves/pumps, and droplet manipulation; iii) Spatiotemporal manipulation of chemical microenvironments implemented by a coupling strategy and open-space microfluidics; and iv) High-throughput manipulation of chemical microenvironments. Finally, we briefly present typical applications of the above-mentioned technical advances in cell-based analyses including cell migration, cell signaling, cell differentiation, multicellular analysis, and drug screening. We further discuss the future improvement of microfluidics manipulation of extracellular chemical microenvironments to fulfill the needs of biological and biomedical research and applications.
Collapse
Affiliation(s)
- Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Shunji Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yiran Guo
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xuemei Zeng
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
17
|
Zhu S, Jiang F, Han Y, Xiang N, Ni Z. Microfluidics for label-free sorting of rare circulating tumor cells. Analyst 2020; 145:7103-7124. [DOI: 10.1039/d0an01148g] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A review discussing the working principles and performances of label-free CTC sorting methods.
Collapse
Affiliation(s)
- Shu Zhu
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Fengtao Jiang
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Yu Han
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Nan Xiang
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| | - Zhonghua Ni
- School of Mechanical Engineering
- and Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments
- Southeast University
- Nanjing
- China
| |
Collapse
|
18
|
Tavakoli H, Zhou W, Ma L, Perez S, Ibarra A, Xu F, Zhan S, Li X. Recent advances in microfluidic platforms for single-cell analysis in cancer biology, diagnosis and therapy. Trends Analyt Chem 2019; 117:13-26. [PMID: 32831435 PMCID: PMC7434086 DOI: 10.1016/j.trac.2019.05.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Understanding molecular, cellular, genetic and functional heterogeneity of tumors at the single-cell level has become a major challenge for cancer research. The microfluidic technique has emerged as an important tool that offers advantages in analyzing single-cells with the capability to integrate time-consuming and labour-intensive experimental procedures such as single-cell capture into a single microdevice at ease and in a high-throughput fashion. Single-cell manipulation and analysis can be implemented within a multi-functional microfluidic device for various applications in cancer research. Here, we present recent advances of microfluidic devices for single-cell analysis pertaining to cancer biology, diagnostics, and therapeutics. We first concisely introduce various microfluidic platforms used for single-cell analysis, followed with different microfluidic techniques for single-cell manipulation. Then, we highlight their various applications in cancer research, with an emphasis on cancer biology, diagnosis, and therapy. Current limitations and prospective trends of microfluidic single-cell analysis are discussed at the end.
Collapse
Affiliation(s)
- Hamed Tavakoli
- College of Environmental Science and Engineering, Nankai
University, Tianjin 300071, People’s Republic of China
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
| | - Wan Zhou
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
| | - Lei Ma
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
| | - Stefani Perez
- Biomedical Engineering, Border Biomedical Research Center,
Environmental Science & Engineering, University of Texas at El Paso, 500 West
University Ave, El Paso, TX 79968, USA
| | - Andrea Ibarra
- Biomedical Engineering, Border Biomedical Research Center,
Environmental Science & Engineering, University of Texas at El Paso, 500 West
University Ave, El Paso, TX 79968, USA
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center,
Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of
China
| | - Sihui Zhan
- College of Environmental Science and Engineering, Nankai
University, Tianjin 300071, People’s Republic of China
| | - XiuJun Li
- College of Environmental Science and Engineering, Nankai
University, Tianjin 300071, People’s Republic of China
- Department of Chemistry and Biochemistry, University of
Texas at El Paso, 500 West University Ave, El Paso, TX 79968, USA
- Biomedical Engineering, Border Biomedical Research Center,
Environmental Science & Engineering, University of Texas at El Paso, 500 West
University Ave, El Paso, TX 79968, USA
| |
Collapse
|
19
|
Sridhar K, Singh A, Butzmann A, Jangam D, Ohgami RS. Molecular genetic testing methodologies in hematopoietic diseases: current and future methods. Int J Lab Hematol 2019; 41 Suppl 1:102-116. [PMID: 31069972 DOI: 10.1111/ijlh.13024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 03/08/2019] [Accepted: 03/12/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Rapid technological advancements in clinical molecular genetics have increased our diagnostic and prognostic capabilities in health care. Understanding these assays, as well as how they may change over time, is critical for pathologists, clinicians, and translational researchers alike. METHODS This review provides a practical summary and basic reference for current molecular genetic technologies, as well as new testing methodologies that are in use, gaining momentum, or anticipated to contribute more broadly in the future. RESULTS Here, we discuss DNA and RNA based methodologies including classic assays such as the polymerase chain reaction (PCR), Sanger sequencing, and microarrays, to more cutting-edge next-generation sequencing (NGS) based assays and emerging molecular technologies such as cell-free DNA (cfDNA) or circulating tumor DNA (ctDNA), and NGS-based detection of infectious disease organisms. CONCLUSION This review serves as a basic foundation for knowledge in current and emerging clinical molecular genetic technologies.
Collapse
Affiliation(s)
- Kaushik Sridhar
- Department of Pathology, Stanford University, Stanford, California
| | - Amol Singh
- Department of Pathology, Stanford University, Stanford, California
| | | | - Diwash Jangam
- Department of Pathology, Stanford University, Stanford, California
| | - Robert S Ohgami
- Department of Pathology, Stanford University, Stanford, California.,Department of Pathology, University of California, San Francisco, CA
| |
Collapse
|
20
|
Chowdury MA, Heileman KL, Moore TA, Young EWK. Biomicrofluidic Systems for Hematologic Cancer Research and Clinical Applications. SLAS Technol 2019; 24:457-476. [PMID: 31173533 DOI: 10.1177/2472630319846878] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A persistent challenge in developing personalized treatments for hematologic cancers is the lack of patient specific, physiologically relevant disease models to test investigational drugs in clinical trials and to select therapies in a clinical setting. Biomicrofluidic systems and organ-on-a-chip technologies have the potential to change how researchers approach the fundamental study of hematologic cancers and select clinical treatment for individual patient. Here, we review microfluidics cell-based technology with application toward studying hematologic tumor microenvironments (TMEs) for the purpose of drug discovery and clinical treatment selection. We provide an overview of state-of-the-art microfluidic systems designed to address questions related to hematologic TMEs and drug development. Given the need to develop personalized treatment platforms involving this technology, we review pharmaceutical drugs and different modes of immunotherapy for hematologic cancers, followed by key considerations for developing a physiologically relevant microfluidic companion diagnostic tool for mimicking different hematologic TMEs for testing with different drugs in clinical trials. Opportunities lie ahead for engineers to revolutionize conventional drug discovery strategies of hematologic cancers, including integrating cell-based microfluidics technology with machine learning and automation techniques, which may stimulate pharma and regulatory bodies to promote research and applications of microfluidics technology for drug development.
Collapse
Affiliation(s)
- Mosfera A Chowdury
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Khalil L Heileman
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Thomas A Moore
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Edmond W K Young
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, Canada.,Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Sharifi Noghabi H, Soo M, Khamenehfar A, Li PC. Dielectrophoretic trapping of single leukemic cells using the conventional and compact optical measurement systems. Electrophoresis 2019; 40:1478-1485. [DOI: 10.1002/elps.201800451] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/25/2019] [Accepted: 01/26/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Hamideh Sharifi Noghabi
- Department of chemistrySimon Fraser University Burnaby British Columbia Canada
- Department of chemistryFaculty of SciencesFerdowsi University of Mashhad Mashhad Iran
| | - Mandy Soo
- Department of chemistrySimon Fraser University Burnaby British Columbia Canada
| | - Avid Khamenehfar
- Department of chemistrySimon Fraser University Burnaby British Columbia Canada
| | - Paul C.H. Li
- Department of chemistrySimon Fraser University Burnaby British Columbia Canada
| |
Collapse
|
22
|
Armbrecht L, Müller RS, Nikoloff J, Dittrich PS. Single-cell protein profiling in microchambers with barcoded beads. MICROSYSTEMS & NANOENGINEERING 2019; 5:55. [PMID: 31700673 PMCID: PMC6826046 DOI: 10.1038/s41378-019-0099-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 05/21/2023]
Abstract
Single-cell profiling provides insights into cellular behaviour that macroscale cell cultures and bulk measurements cannot reveal. In the context of personalized cancer treatment, the profiling of individual tumour cells may lead to higher success rates for therapies by rapidly selecting the most efficacious drugs. Currently, genomic analysis at the single-cell level is available through highly sensitive sequencing approaches. However, the identification and quantification of intracellular or secreted proteins or metabolites remains challenging. Here, we introduce a microfluidic method that facilitates capture, automated data acquisition and the multiplexed quantification of proteins from individual cells. The microfluidic platform comprises 1026 chambers with a volume of 152 pL each, in which single cells and barcoded beads are co-immobilized. We demonstrated multiplexed single-cell protein quantification with three different mammalian cell lines, including two model breast cancer cell lines. We established on-chip immunoassays for glyceraldehyde-3-phosphate dehydrogenase (GAPDH), galectin-3 (Gal-3) and galectin-3 binding protein (Gal-3bp) with detection limits as low as 7.0 × 104, 2.3 × 105 and 1.8 × 103 molecules per cell, respectively. The three investigated cell types had high cytosolic levels of GAPDH and could be clearly differentiated by their expression levels of Gal-3 and Gal-3bp, which are important factors that contribute to cancer metastasis. Because it employed commercially available barcoded beads for this study, our platform could be easily used for the single-cell protein profiling of several hundred different targets. Moreover, this versatile method is applicable to the analysis of bacteria, yeast and mammalian cells and nanometre-sized lipid vesicles.
Collapse
Affiliation(s)
- Lucas Armbrecht
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Rafael Sebastian Müller
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Jonas Nikoloff
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Petra Stephanie Dittrich
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| |
Collapse
|
23
|
Shrirao AB, Fritz Z, Novik EM, Yarmush GM, Schloss RS, Zahn JD, Yarmush ML. Microfluidic flow cytometry: The role of microfabrication methodologies, performance and functional specification. TECHNOLOGY 2018; 6:1-23. [PMID: 29682599 PMCID: PMC5907470 DOI: 10.1142/s2339547818300019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Flow cytometry is an invaluable tool utilized in modern biomedical research and clinical applications requiring high throughput, high resolution particle analysis for cytometric characterization and/or sorting of cells and particles as well as for analyzing results from immunocytometric assays. In recent years, research has focused on developing microfluidic flow cytometers with the motivation of creating smaller, less expensive, simpler, and more autonomous alternatives to conventional flow cytometers. These devices could ideally be highly portable, easy to operate without extensive user training, and utilized for research purposes and/or point-of-care diagnostics especially in limited resource facilities or locations requiring on-site analyses. However, designing a device that fulfills the criteria of high throughput analysis, automation and portability, while not sacrificing performance is not a trivial matter. This review intends to present the current state of the field and provide considerations for further improvement by focusing on the key design components of microfluidic flow cytometers. The recent innovations in particle focusing and detection strategies are detailed and compared. This review outlines performance matrix parameters of flow cytometers that are interdependent with each other, suggesting trade offs in selection based on the requirements of the applications. The ongoing contribution of microfluidics demonstrates that it is a viable technology to advance the current state of flow cytometry and develop automated, easy to operate and cost-effective flow cytometers.
Collapse
Affiliation(s)
- Anil B Shrirao
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| | - Zachary Fritz
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| | - Eric M Novik
- Hurel Corporation, 671, Suite B, U.S. Highway 1, North Brunswick, NJ 08902
| | - Gabriel M Yarmush
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| | - Rene S Schloss
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| | - Jeffrey D Zahn
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| | - Martin L Yarmush
- Department of Biomedical Engineering, Rutgers University, 599, Taylor Road, Piscataway, NJ 08854
| |
Collapse
|
24
|
Cellular dielectrophoresis coupled with single-cell analysis. Anal Bioanal Chem 2018; 410:2499-2515. [DOI: 10.1007/s00216-018-0896-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/11/2018] [Accepted: 01/17/2018] [Indexed: 01/09/2023]
|
25
|
Mansoorifar A, Koklu A, Ma S, Raj GV, Beskok A. Electrical Impedance Measurements of Biological Cells in Response to External Stimuli. Anal Chem 2018; 90:4320-4327. [PMID: 29402081 DOI: 10.1021/acs.analchem.7b05392] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Dielectric spectroscopy (DS) is a noninvasive technique for real-time measurements of the impedance spectra of biological cells. DS enables characterization of cellular dielectric properties such as membrane capacitance and cytoplasmic conductivity. We have developed a lab-on-a-chip device that uses an electro-activated microwells array for capturing, DS measurements, and unloading of biological cells. Impedance measurements were conducted at 0.2 V in the 10 kHz to 40 MHz range with 6 s time resolution. An equivalent circuit model was developed to extract the cell membrane capacitance and cell cytoplasmic conductivity from the impedance spectra. A human prostate cancer cell line, PC-3, was used to evaluate the device performance. Suspension of PC-3 cells in low conductivity buffers (LCB) enhanced their dielectrophoretic trapping and impedance response. We report the time course of the variations in dielectric properties of PC-3 cells suspended in LCB and their response to sudden pH change from a pH of 7.3 to a pH of 5.8. Importantly, we demonstrated that our device enabled real-time measurements of dielectric properties of live cancer cells and allowed the assessment of the cellular response to variations in buffer conductivity and pH. These data support further development of this device toward single cell measurements.
Collapse
Affiliation(s)
- Amin Mansoorifar
- Department of Mechanical Engineering , Southern Methodist University , Dallas , Texas 75205 , United States
| | - Anil Koklu
- Department of Mechanical Engineering , Southern Methodist University , Dallas , Texas 75205 , United States
| | - Shihong Ma
- Departments of Urology and Pharmacology , University of Texas Southwestern Medical Center , Dallas , Texas 75390 , United States
| | - Ganesh V Raj
- Departments of Urology and Pharmacology , University of Texas Southwestern Medical Center , Dallas , Texas 75390 , United States
| | - Ali Beskok
- Department of Mechanical Engineering , Southern Methodist University , Dallas , Texas 75205 , United States
| |
Collapse
|
26
|
Gupta SD, Sachs Z. Novel single-cell technologies in acute myeloid leukemia research. Transl Res 2017; 189:123-135. [PMID: 28802867 PMCID: PMC6584944 DOI: 10.1016/j.trsl.2017.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/18/2017] [Accepted: 07/20/2017] [Indexed: 12/29/2022]
Abstract
Acute myeloid leukemia (AML) is a lethal malignancy because patients who initially respond to chemotherapy eventually relapse with treatment refractory disease. Relapse is caused by leukemia stem cells (LSCs) that reestablish the disease through self-renewal. Self-renewal is the ability of a stem cell to produce copies of itself and give rise to progeny cells. Therefore, therapeutic strategies eradicating LSCs are essential to prevent relapse and achieve long-term remission in AML. AML is a heterogeneous disease both at phenotypic and genotypic levels, and this heterogeneity extends to LSCs. Classical studies in AML have aimed at characterization of the bulk tumor population, thereby masking cellular heterogeneity. Single-cell approaches provide a novel opportunity to elucidate molecular mechanisms in heterogeneous diseases such as AML. In recent years, major advancements in single-cell measurement systems have revolutionized our understanding of the pathophysiology of AML and enabled the characterization of LSCs. Identifying the molecular mechanisms critical to AML LSCs will aid in the development of targeted therapeutic strategies to combat this deadly disease.
Collapse
Affiliation(s)
- Soumyasri Das Gupta
- Division of Hematology, Oncology, and Transplantation, Department Medicine, University of Minnesota, Minneapolis, Minn
| | - Zohar Sachs
- Division of Hematology, Oncology, and Transplantation, Department Medicine, University of Minnesota, Minneapolis, Minn; Masonic Cancer Center, University of Minnesota, Minneapolis, Minn.
| |
Collapse
|
27
|
Hansen CE, Lam WA. Clinical Implications of Single-Cell Microfluidic Devices for Hematological Disorders. Anal Chem 2017; 89:11881-11892. [PMID: 28942646 DOI: 10.1021/acs.analchem.7b01013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Single-cell microfluidic devices are poised to substantially impact the hematology field by providing a high-throughput and rapid device to analyze disease-mediated biophysical cellular changes in the clinical setting in order to diagnose patients and monitor disease prognosis. In this Feature, we cover recent advances of single-cell microfluidic devices for studying and diagnosing hematological dysfunctions and the clinical impact made possible by these advances.
Collapse
Affiliation(s)
- Caroline E Hansen
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta/Emory University School of Medicine , Atlanta, Georgia 30322, United States.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University , Atlanta, Georgia 30332, United States.,School of Chemistry and Biochemistry, Georgia Institute of Technology , Atlanta, Georgia 30332, United States
| | - Wilbur A Lam
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta/Emory University School of Medicine , Atlanta, Georgia 30322, United States.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University , Atlanta, Georgia 30332, United States.,School of Chemistry and Biochemistry, Georgia Institute of Technology , Atlanta, Georgia 30332, United States
| |
Collapse
|
28
|
|
29
|
Affiliation(s)
- Lucas Armbrecht
- Department of Biosystems Science and Engineering, ETH Zurich, CH-8093 Zurich, Switzerland
| | | |
Collapse
|