1
|
Simpkins LLC, Henriquez LA, Tran M, Adams TNG. Electrical Impedance Spectroscopy as a Tool to Detect the Epithelial to Mesenchymal Transition in Prostate Cancer Cells. BIOSENSORS 2024; 14:503. [PMID: 39451716 PMCID: PMC11506005 DOI: 10.3390/bios14100503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024]
Abstract
Prostate cancer (PCa) remains a significant health threat, with chemoresistance and recurrence posing major challenges despite advances in treatment. The epithelial to mesenchymal transition (EMT), a biochemical process where cells lose epithelial features and gain mesenchymal traits, is linked to chemoresistance and metastasis. Electrical impedance spectroscopy (EIS), a novel label-free electrokinetic technique, offers promise in detecting cell phenotype changes. In this study, we employed EIS to detect EMT in prostate cancer cells (PCCs). PC3, DU145, and LNCaP cells were treated with EMT induction media for five days. EIS characterization revealed unique impedance spectra correlating with metastatic potential, distinguishing DU145 EMT+ and EMT- cells, and LNCaP EMT+ and EMT- cells (in combination with dielectrophoresis), with comparisons made to epithelial and mesenchymal controls. These changes were supported by shifts in electrical signatures, morphologies, and protein expression, including the downregulation of E-cadherin and upregulation of vimentin. No phenotype change was observed in PC3 cells, which maintained a mesenchymal phenotype. EMT+ cells were also distinguishable from mixtures of EMT+ and EMT- cells. This study demonstrates key advancements: the application of EIS and dielectrophoresis for label-free EMT detection in PCCs, characterization of cell electrical signatures after EMT, and EIS sensitivity to EMT transitions. Detecting EMT in PCa is important to the development of more effective treatments and overcoming the challenges of chemoresistance.
Collapse
Affiliation(s)
- Lexi L. C. Simpkins
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA; (L.L.C.S.); (L.A.H.); (M.T.)
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Luis A. Henriquez
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA; (L.L.C.S.); (L.A.H.); (M.T.)
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Mary Tran
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA; (L.L.C.S.); (L.A.H.); (M.T.)
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Tayloria N. G. Adams
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA; (L.L.C.S.); (L.A.H.); (M.T.)
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697, USA
- Department of Materials and Science Engineering, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
2
|
Tsai T, Vyas PD, Crowell LL, Tran M, Ward DW, Qin Y, Castro A, Adams TNG. Electrical signature of heterogeneous human mesenchymal stem cells. Electrophoresis 2024; 45:1562-1573. [PMID: 38738344 DOI: 10.1002/elps.202300202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 03/15/2024] [Accepted: 03/27/2024] [Indexed: 05/14/2024]
Abstract
Human mesenchymal stem cells (hMSCs) have gained traction in transplantation therapy due to their immunomodulatory, paracrine, immune-evasive, and multipotent differentiation potential. The inherent heterogeneity of hMSCs poses a challenge for therapeutic treatments and necessitates the identification of robust biomarkers to ensure reproducibility in both in vivo and in vitro experiments. In this study, we utilized dielectrophoresis (DEP), a label-free electrokinetic phenomenon, to investigate the heterogeneity of hMSCs derived from bone marrow (BM) and adipose tissue (AD). The electrical properties of BM-hMSCs were compared to homogeneous mouse fibroblasts (NIH-3T3), human fibroblasts (WS1), and human embryonic kidney cells (HEK-293). The DEP profile of BM-hMSCs differed most from HEK-293 cells. We compared the DEP profiles of BM-hMSCs and AD-hMSCs and found that they have similar membrane capacitances, differing cytoplasm conductivity, and transient slopes. Inducing both populations to differentiate into adipocyte and osteoblast cells revealed that they behave differently in response to differentiation-inducing cytokines. Histology and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analyses of the differentiation-related genes revealed differences in heterogeneity between BM-hMSCs and AD-hMSCs. The differentiation profiles correlate well with the DEP profiles developed and indicate differences in the heterogeneity of BM-hMSCs and AD-hMSCs. Our results demonstrate that using DEP, membrane capacitance, cytoplasm conductivity, and transient slope can uniquely characterize the inherent heterogeneity of hMSCs to guide robust and reproducible stem cell transplantation therapies.
Collapse
Affiliation(s)
- Tunglin Tsai
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California, USA
| | - Prema D Vyas
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California, USA
| | - Lexi L Crowell
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California, USA
| | - Mary Tran
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California, USA
| | - Destiney W Ward
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California, USA
| | - Yufan Qin
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California, USA
| | - Angie Castro
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California, USA
| | - Tayloria N G Adams
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, USA
| |
Collapse
|
3
|
Acerbo E, Bellotti MI, Bonetto FJ. Determining mammalian cells state by fractal micromotion. Heliyon 2024; 10:e26352. [PMID: 38444473 PMCID: PMC10912215 DOI: 10.1016/j.heliyon.2024.e26352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 03/07/2024] Open
Abstract
We applied four fractal dimension estimation algorithms on the temporal electrical impedance signal of normal MDCK type II cell cultures monitored by ECIS technique and showed that the fractal dimension due to micromotion allows discriminating processes not sensed by the spectral impedance of the culture. In this work we subjected cell cultures to electric current damage and drug exposure to analyze the changes in the fractal structure of the temporal signal. Among the changes presented and detected are the differentiation between a healthy monolayer and one exposed to a drug, as well as the distinction between a seeding process and a wound-healing process performed by electric current. The four algorithms used were validated by applying them on topological functions of known fractal dimension, a study that determined the necessary conditions for a correct estimation.
Collapse
Affiliation(s)
- Esteban Acerbo
- Laboratorio de Cavitación y Tecnología, Centro Atómico Bariloche, San Carlos de Bariloche, R8402AGP, Argentina
- Consejo Nacional de Investigaciones Ciencia y Técnicas (CONICET), Buenos Aires, C1033AAJ, Argentina
- Universidad Nacional de Cuyo Instituto Balseiro, San Carlos de Bariloche, R8402AGP, Argentina
| | - Mariela I. Bellotti
- Laboratorio de Cavitación y Tecnología, Centro Atómico Bariloche, San Carlos de Bariloche, R8402AGP, Argentina
- Comisión Nacional de Energía Atómica (CNEA), Centro Atómico Bariloche, San Carlos de Bariloche, R8402AGP, Argentina
- Universidad Nacional de Cuyo Instituto Balseiro, San Carlos de Bariloche, R8402AGP, Argentina
- Universidad Nacional de Río NegroSede Andina Carrera de Medicina, San Carlos de Bariloche, R8402AGP, Argentina
| | - Fabian J. Bonetto
- Laboratorio de Cavitación y Tecnología, Centro Atómico Bariloche, San Carlos de Bariloche, R8402AGP, Argentina
- Comisión Nacional de Energía Atómica (CNEA), Centro Atómico Bariloche, San Carlos de Bariloche, R8402AGP, Argentina
- Consejo Nacional de Investigaciones Ciencia y Técnicas (CONICET), Buenos Aires, C1033AAJ, Argentina
- Universidad Nacional de Cuyo Instituto Balseiro, San Carlos de Bariloche, R8402AGP, Argentina
| |
Collapse
|
4
|
Ding L, Oh S, Shrestha J, Lam A, Wang Y, Radfar P, Warkiani ME. Scaling up stem cell production: harnessing the potential of microfluidic devices. Biotechnol Adv 2023; 69:108271. [PMID: 37844769 DOI: 10.1016/j.biotechadv.2023.108271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Stem cells are specialised cells characterised by their unique ability to both self-renew and transform into a wide array of specialised cell types. The widespread interest in stem cells for regenerative medicine and cultivated meat has led to a significant demand for these cells in both research and practical applications. Despite the growing need for stem cell manufacturing, the industry faces significant obstacles, including high costs for equipment and maintenance, complicated operation, and low product quality and yield. Microfluidic technology presents a promising solution to the abovementioned challenges. As an innovative approach for manipulating liquids and cells within microchannels, microfluidics offers a plethora of advantages at an industrial scale. These benefits encompass low setup costs, ease of operation and multiplexing, minimal energy consumption, and the added advantage of being labour-free. This review presents a thorough examination of the prominent microfluidic technologies employed in stem cell research and explores their promising applications in the burgeoning stem cell industry. It thoroughly examines how microfluidics can enhance cell harvesting from tissue samples, facilitate mixing and cryopreservation, streamline microcarrier production, and efficiently conduct cell separation, purification, washing, and final cell formulation post-culture.
Collapse
Affiliation(s)
- Lin Ding
- Smart MCs Pty Ltd, Ultimo, Sydney, 2007, Australia.
| | - Steve Oh
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Jesus Shrestha
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Alan Lam
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Yaqing Wang
- School of Biomedical Engineering, University of Science and Technology of China, Hefei 230026, China; Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Payar Radfar
- Smart MCs Pty Ltd, Ultimo, Sydney, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia..
| |
Collapse
|
5
|
Tupper LL, Keese CR, Matteson DS. Classifying contaminated cell cultures using time series features. J Appl Stat 2023; 51:1210-1226. [PMID: 38628445 PMCID: PMC11018005 DOI: 10.1080/02664763.2023.2248413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/30/2023] [Indexed: 04/19/2024]
Abstract
We examine the use of time series data, derived from Electric Cell-substrate Impedance Sensing (ECIS), to differentiate between standard mammalian cell cultures and those infected with a mycoplasma organism. With the goal of easy visualization and interpretation, we perform low-dimensional feature-based classification, extracting application-relevant features from the ECIS time courses. We can achieve very high classification accuracy using only two features, which depend on the cell line under examination. Initial results also show the existence of experimental variation between plates and suggest types of features that may prove more robust to such variation. Our paper is the first to perform a broad examination of ECIS time course features in the context of detecting contamination; to combine different types of features to achieve classification accuracy while preserving interpretability; and to describe and suggest possibilities for ameliorating plate-to-plate variation.
Collapse
|
6
|
Chen X, Liu S, Hu XG, Liu T, Shen M, Peng Y, Hu S, Zhao Y. Enrichment and Selection of Particles through Parallel Induced-Charge Electro-osmotic Streaming for Detection of Low-Abundance Nanoparticles and Targeted Microalgae. Anal Chem 2023; 95:11714-11722. [PMID: 37486806 DOI: 10.1021/acs.analchem.3c01729] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Manipulation of micro- and nanoscale objects is an essential procedure in many detection and sensing applications, including disease diagnosis and environmental monitoring. Induced-charge electro-osmotic (ICEO) vortices present excellent advantages in the enrichment and selection of micro/nanoscale particles for downstream detection due to gentle conditions and contactless operation, but the application of this method is currently constrained by the throughput. Double-layer charging at the ends of bipolar electrodes can maintain a continuous flow of electric current in the fluidically isolated channels, which provides a feasible method to manipulate particles using parallel ICEO vortices, promoting throughput of particle manipulation without compromising efficiency and overcoming the complicated ohmic contact of electrodes. Encouraged by these, we put forward a novel method with parallel ICEO vortices to manipulate micro/nanoscale samples for downstream detection. First, we study the extension regulation of the low-frequency electric field and mediating effect of the open BPEs on the extended electric field and characterize electric equilibrium states of microparticles and their voltage dependence. Afterward, we leverage this method to enrich nanoparticles for detection of low-abundance nanoparticles with about 20- and 40-fold fluorescence intensities by integrating with a simple fiber-optic sensor. Furthermore, this technique is engineered for the selection of targeted microalgae to continuously detect their proliferation behaviors by combining with a homemade electrical impedance spectroscopy device. This method can reinforce the throughput of ICEO vortices and enables it to integrate with simple and economical sensors to accomplish disease diagnosis and environmental monitoring.
Collapse
Affiliation(s)
- Xiaoming Chen
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao 066004, PR China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao 066004, PR China
| | - Shun Liu
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao 066004, PR China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao 066004, PR China
| | - Xu-Guang Hu
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao 066004, PR China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao 066004, PR China
| | - Tengteng Liu
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao 066004, PR China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao 066004, PR China
| | - Mo Shen
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao 066004, PR China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao 066004, PR China
| | - Yun Peng
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao 066004, PR China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao 066004, PR China
| | - Sheng Hu
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao 066004, PR China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao 066004, PR China
| | - Yong Zhao
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao 066004, PR China
- Hebei Key Laboratory of Micro-Nano Precision Optical Sensing and Measurement Technology, Qinhuangdao 066004, PR China
| |
Collapse
|
7
|
Joos-Vandewalle J, Steenkamp V, Prinsloo E. A simplified workflow with end-point validation of real-time electrical cell-substrate impedance sensing of retinoic acid stimulated neurogenesis in human SH-SY5Y cells in vitro. BMC Res Notes 2023; 16:93. [PMID: 37264464 DOI: 10.1186/s13104-023-06369-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 05/25/2023] [Indexed: 06/03/2023] Open
Abstract
OBJECTIVE Retinoic acid (RA) is known to transition proliferating SH-SY5Y neuroblastoma cells towards functional neurons. However, the activity of RA is restricted due to its photolability where any findings from prolonged time course observations using microscopy may alter outcomes. The aim of the study was to establish a real-time, long-term (9-day) protocol for the screening of differentiation events using Electrical cell-substrate impedance sensing (ECIS). RESULTS AND DISCUSSION A differentiation baseline for SH-SY5Y cells was established. Cells were seeded and exposed to repeated spikes of RA using the xCELLigence real-time cell analyser single plate (RTCA-SP) for real-time monitoring and identification of differentiation activity over a 9 day period in order to be more representative of differentiation over a prolonged timeline. Specific features associated with differentiation (growth inhibition, neurite outgrowths) were confirmed by end-point analysis. RA-induced growth inhibition and assumed phenotypic changes (i.e. neurite outgrowth) were identified by the xCELLigence analysis and further confirmed by end-point metabolic and phenotypic assays. Change in cellular morphology and neurite outgrowth length was identified by end-point fluorescence detection followed by computational analysis. Based on this it was possible to identify SH-SY5Y phenotypic differentiation with distinct phases observed over 9 days using Electric cell-substrate impedance sensing (ECIS) cell index traces providing a path to application in larger scale neurotrophic factor screening using this scalable technology.
Collapse
Affiliation(s)
- Julia Joos-Vandewalle
- Biotechnology Innovation Centre, Rhodes University, P.O. Box 94, Makhanda, 6140, South Africa
| | - Vanessa Steenkamp
- Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia, 0007, South Africa
| | - Earl Prinsloo
- Biotechnology Innovation Centre, Rhodes University, P.O. Box 94, Makhanda, 6140, South Africa.
| |
Collapse
|
8
|
Yale AR, Kim E, Gutierrez B, Hanamoto JN, Lav NS, Nourse JL, Salvatus M, Hunt RF, Monuki ES, Flanagan LA. Regulation of neural stem cell differentiation and brain development by MGAT5-mediated N-glycosylation. Stem Cell Reports 2023:S2213-6711(23)00141-8. [PMID: 37172586 DOI: 10.1016/j.stemcr.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023] Open
Abstract
Undifferentiated neural stem and progenitor cells (NSPCs) encounter extracellular signals that bind plasma membrane proteins and influence differentiation. Membrane proteins are regulated by N-linked glycosylation, making it possible that glycosylation plays a critical role in cell differentiation. We assessed enzymes that control N-glycosylation in NSPCs and found that loss of the enzyme responsible for generating β1,6-branched N-glycans, N-acetylglucosaminyltransferase V (MGAT5), led to specific changes in NSPC differentiation in vitro and in vivo. Mgat5 homozygous null NSPCs in culture formed more neurons and fewer astrocytes compared with wild-type controls. In the brain cerebral cortex, loss of MGAT5 caused accelerated neuronal differentiation. Rapid neuronal differentiation led to depletion of cells in the NSPC niche, resulting in a shift in cortical neuron layers in Mgat5 null mice. Glycosylation enzyme MGAT5 plays a critical and previously unrecognized role in cell differentiation and early brain development.
Collapse
Affiliation(s)
- Andrew R Yale
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA 92697, USA; Department of Neurology, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Estelle Kim
- Department of Neurology, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Brenda Gutierrez
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA 92697, USA; Department of Neurology, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - J Nicole Hanamoto
- Department of Neurology, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Nicole S Lav
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Jamison L Nourse
- Department of Neurology, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Marc Salvatus
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Robert F Hunt
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Edwin S Monuki
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA; Department of Pathology & Laboratory Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Lisa A Flanagan
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA 92697, USA; Department of Neurology, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
9
|
Monteduro AG, Rizzato S, Caragnano G, Trapani A, Giannelli G, Maruccio G. Organs-on-chips technologies – A guide from disease models to opportunities for drug development. Biosens Bioelectron 2023; 231:115271. [PMID: 37060819 DOI: 10.1016/j.bios.2023.115271] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 11/24/2022] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
Current in-vitro 2D cultures and animal models present severe limitations in recapitulating human physiopathology with striking discrepancies in estimating drug efficacy and side effects when compared to human trials. For these reasons, microphysiological systems, organ-on-chip and multiorgans microdevices attracted considerable attention as novel tools for high-throughput and high-content research to achieve an improved understanding of diseases and to accelerate the drug development process towards more precise and eventually personalized standards. This review takes the form of a guide on this fast-growing field, providing useful introduction to major themes and indications for further readings. We start analyzing Organs-on-chips (OOC) technologies for testing the major drug administration routes: (1) oral/rectal route by intestine-on-a-chip, (2) inhalation by lung-on-a-chip, (3) transdermal by skin-on-a-chip and (4) intravenous through vascularization models, considering how drugs penetrate in the bloodstream and are conveyed to their targets. Then, we focus on OOC models for (other) specific organs and diseases: (1) neurodegenerative diseases with brain models and blood brain barriers, (2) tumor models including their vascularization, organoids/spheroids, engineering and screening of antitumor drugs, (3) liver/kidney on chips and multiorgan models for gastrointestinal diseases and metabolic assessment of drugs and (4) biomechanical systems recapitulating heart, muscles and bones structures and related diseases. Successively, we discuss technologies and materials for organ on chips, analyzing (1) microfluidic tools for organs-on-chips, (2) sensor integration for real-time monitoring, (3) materials and (4) cell lines for organs on chips. (Nano)delivery approaches for therapeutics and their on chip assessment are also described. Finally, we conclude with a critical discussion on current significance/relevance, trends, limitations, challenges and future prospects in terms of revolutionary impact on biomedical research, preclinical models and drug development.
Collapse
Affiliation(s)
- Anna Grazia Monteduro
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Silvia Rizzato
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Giusi Caragnano
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy
| | - Adriana Trapani
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology IRCCS "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
| | - Giuseppe Maruccio
- Omnics Research Group, Department of Mathematics and Physics "Ennio De Giorgi", University of Salento and Institute of Nanotechnology, CNR-Nanotec and INFN Sezione di Lecce, Via per Monteroni, 73100, Lecce, Italy.
| |
Collapse
|
10
|
Zhang Z, Yuan X, Guo H, Shang P. The Influence of Electrode Design on Detecting the Effects of Ferric Ammonium Citrate (FAC) on Pre-Osteoblast through Electrical Cell-Substrate Impedance Sensing (ECIS). BIOSENSORS 2023; 13:322. [PMID: 36979534 PMCID: PMC10046662 DOI: 10.3390/bios13030322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/15/2023] [Accepted: 02/22/2023] [Indexed: 06/18/2023]
Abstract
Detection sensitivity is a crucial factor in the application of ECIS sensors. For these biosensors, the electrode configuration has a direct impact on sensitivity, yet few studies on monopolar electrodes have been reported. In this study, ECIS sensor arrays, which have a series of working electrode configuration with a wide diameter range and different electrode number, were fabricated to monitor living osteoblast-like MC3T3-E1 cells. The experimental results revealed that when the electrode diameter was larger than 25 μm, electrodes with smaller diameter and number yielded higher impedance values and generated more impedance shift to cell status change. The membrane capacitance obtained by equivalent circuit fitting was at the same level. When the electrode diameter was even smaller, the results in detection of cell monolayer were opposite, and there was no distinct relationship between impedance and membrane capacitance shift to cell status change and electrode geometry. The proposed sensor chip, allowing for a sustained and stable detection of cellular impedance, provides the basis for the selection of the electrode configuration of monopolar electrodes. The test results of electrodes with a diameter of 25 μm and lower indicated the possibility of single cell impedance measurement, which can provide unique insight into the heterogeneous electrical behavior of cells, and, in this case, the electrode size should be close to the cell size.
Collapse
Affiliation(s)
- Zheyuan Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Key Laboratory for Space Biosciences and Biotechnology, Northwestern Polytechnical University, Xi’an 710072, China
| | - Xichen Yuan
- Key Laboratory for Space Biosciences and Biotechnology, Northwestern Polytechnical University, Xi’an 710072, China
- School of Mechanical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| | - Huijie Guo
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Key Laboratory for Space Biosciences and Biotechnology, Northwestern Polytechnical University, Xi’an 710072, China
| | - Peng Shang
- Key Laboratory for Space Biosciences and Biotechnology, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute, Northwestern Polytechnical University in Shenzhen, Shenzhen 518110, China
| |
Collapse
|
11
|
Combining Electrostimulation with Impedance Sensing to Promote and Track Osteogenesis within a Titanium Implant. Biomedicines 2023; 11:biomedicines11030697. [PMID: 36979676 PMCID: PMC10045247 DOI: 10.3390/biomedicines11030697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/03/2023] [Accepted: 02/17/2023] [Indexed: 03/02/2023] Open
Abstract
(1) Background: Electrical stimulation is a promising alternative to promote bone fracture healing but with the limitation of tracking the osteogenesis progress in vivo. To overcome this issue, we present an opportunity to combine the electrical stimulation of a commercial titanium implant, which promotes osteogenesis within the fracture, with a real-time readout of the osteogenic progress by impedance sensing. This makes it possible to adjust the electrical stimulation modalities to the individual patient’s fracture healing process. (2) Methods: In detail, osteogenic differentiation of several cell types was monitored under continuous or pulsatile electrical stimulation at 0.7 V AC/20 Hz for at least seven days on a titanium implant by electric cell-substrate impedance sensing (ECIS). For control, chemical induction of osteogenic differentiation was induced. (3) Results: The most significant challenge was to discriminate impedance changes caused by proliferation events from those initiated by osteogenic differentiation. This discrimination was achieved by remodeling the impedance parameter Alpha (α), which increases over time for pulsatile electrically stimulated stem cells. Boosted α-values were accompanied by an increased formation of actin stress fibers and a reduced expression of the focal adhesion kinase in the cell periphery; morphological alterations known to occur during osteogenesis. (4) Conclusions: This work provided the basis for developing an effective fracture therapy device, which can induce osteogenesis on the one hand, and would allow us to monitor the induction process on the other hand.
Collapse
|
12
|
Chen YS, Huang CH, Pai PC, Seo J, Lei KF. A Review on Microfluidics-Based Impedance Biosensors. BIOSENSORS 2023; 13:bios13010083. [PMID: 36671918 PMCID: PMC9855525 DOI: 10.3390/bios13010083] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 05/30/2023]
Abstract
Electrical impedance biosensors are powerful and continuously being developed for various biological sensing applications. In this line, the sensitivity of impedance biosensors embedded with microfluidic technologies, such as sheath flow focusing, dielectrophoretic focusing, and interdigitated electrode arrays, can still be greatly improved. In particular, reagent consumption reduction and analysis time-shortening features can highly increase the analytical capabilities of such biosensors. Moreover, the reliability and efficiency of analyses are benefited by microfluidics-enabled automation. Through the use of mature microfluidic technology, complicated biological processes can be shrunk and integrated into a single microfluidic system (e.g., lab-on-a-chip or micro-total analysis systems). By incorporating electrical impedance biosensors, hand-held and bench-top microfluidic systems can be easily developed and operated by personnel without professional training. Furthermore, the impedance spectrum provides broad information regarding cell size, membrane capacitance, cytoplasmic conductivity, and cytoplasmic permittivity without the need for fluorescent labeling, magnetic modifications, or other cellular treatments. In this review article, a comprehensive summary of microfluidics-based impedance biosensors is presented. The structure of this article is based on the different substrate material categorizations. Moreover, the development trend of microfluidics-based impedance biosensors is discussed, along with difficulties and challenges that may be encountered in the future.
Collapse
Affiliation(s)
- Yu-Shih Chen
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chun-Hao Huang
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ping-Ching Pai
- Department of Radiation Oncology, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Jungmok Seo
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Electrical & Electronic Engineering, Yonsei University, Seoul 120-749, Republic of Korea
| | - Kin Fong Lei
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Radiation Oncology, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Department of Electrical & Electronic Engineering, Yonsei University, Seoul 120-749, Republic of Korea
| |
Collapse
|
13
|
Mandal M, Shukla J, Datta B, Dutta G. Role of Biosensors in Regenerative Therapeutics: Past, Present, and Future Prospects. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
|
14
|
Marva G, Ünsal S, Benest AV, Bates DO, Ordóñez-Morán P. Novel Approach to Measure Transepithelial Electrical Resistance in Intestinal Cells. Methods Mol Biol 2023; 2650:35-42. [PMID: 37310621 DOI: 10.1007/978-1-0716-3076-1_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The technique electric cell-substrate impedance sensing (ECIS) can be used to detect and monitor the behavior of intestinal cells. The methodology presented was designed to achieve results within a short time frame, and it was tailored to use a colonic cancer cell line. Differentiation of intestinal cancer cells has previously been reported to be regulated by retinoic acid (RA). Here, colonic cancer cells were cultured in the ECIS array before being treated with RA, and any changes in response to RA were monitored after treatment. The ECIS recorded changes in impedance in response to the treatment and vehicle. This methodology poses as a novel way to record the behavior of colonic cells and opens new avenues for in vitro research.
Collapse
Affiliation(s)
- Gurveer Marva
- Translational Medical Sciences Unit, School of Medicine, Centre for Cancer Sciences, Biodiscovery Institute-3, University Park, University of Nottingham, Nottingham, UK
| | - Seyda Ünsal
- Department of Molecular Medicine, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Andrew V Benest
- Translational Medical Sciences Unit, School of Medicine, Centre for Cancer Sciences, Biodiscovery Institute-3, University Park, University of Nottingham, Nottingham, UK
| | - David O Bates
- Translational Medical Sciences Unit, School of Medicine, Centre for Cancer Sciences, Biodiscovery Institute-3, University Park, University of Nottingham, Nottingham, UK
| | - Paloma Ordóñez-Morán
- Translational Medical Sciences Unit, School of Medicine, Centre for Cancer Sciences, Biodiscovery Institute-3, University Park, University of Nottingham, Nottingham, UK.
| |
Collapse
|
15
|
Pérez P, Serrano-Viseas JA, Fernández-Scagliusi S, Martín-Fernández D, Huertas G, Yúfera A. Oscillation-Based Spectroscopy for Cell-Culture Monitorization. FRONTIERS IN ELECTRONICS 2022. [DOI: 10.3389/felec.2022.836669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Biological Impedance is a physical property related to the state and inherent evolution of biological samples. Among the existing impedance measurement methods, Oscillation-Based (OB) tests are a simple and smart solution to indirectly measure impedance correlated with the amplitude and frequency of the generated oscillation which are proportional to the sample under test. An OB test requires tuning of the system blocks to specifications derived from every measurement problem. The OB setup must be done to obtain the optimum measurement sensitivity for the specific constraints imposed by the system under test, electronic interfaces, and electrodes employed for test. This work proposes the extension of OB measurement systems to spectroscopy test, enabling a completely new range of applications for this technology without the restrictions imposed by setting a fixed frequency on the electrical oscillator. Some examples will be presented to the measurement of cell cultures samples, considering the corresponding circuit interfaces and electric models for the electrode-cell system. The proposed analysis method allows the selection of the best oscillator elements for optimum sensitivity range in amplitude and frequency oscillation values, when a specific cell culture is monitored for the OB system.
Collapse
|
16
|
Bounik R, Cardes F, Ulusan H, Modena MM, Hierlemann A. Impedance Imaging of Cells and Tissues: Design and Applications. BME FRONTIERS 2022; 2022:1-21. [PMID: 35761901 PMCID: PMC7612906 DOI: 10.34133/2022/9857485] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 03/28/2022] [Indexed: 11/09/2022] Open
Abstract
Due to their label-free and noninvasive nature, impedance measurements have attracted increasing interest in biological research. Advances in microfabrication and integrated-circuit technology have opened a route to using large-scale microelectrode arrays for real-time, high-spatiotemporal-resolution impedance measurements of biological samples. In this review, we discuss different methods and applications of measuring impedance for cell and tissue analysis with a focus on impedance imaging with microelectrode arrays in in vitro applications. We first introduce how electrode configurations and the frequency range of the impedance analysis determine the information that can be extracted. We then delve into relevant circuit topologies that can be used to implement impedance measurements and their characteristic features, such as resolution and data-acquisition time. Afterwards, we detail design considerations for the implementation of new impedance-imaging devices. We conclude by discussing future fields of application of impedance imaging in biomedical research, in particular applications where optical imaging is not possible, such as monitoring of ex vivo tissue slices or microelectrode-based brain implants.
Collapse
Affiliation(s)
- Raziyeh Bounik
- ETH Zürich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - Fernando Cardes
- ETH Zürich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - Hasan Ulusan
- ETH Zürich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - Mario M. Modena
- ETH Zürich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - Andreas Hierlemann
- ETH Zürich, Department of Biosystems Science and Engineering, Basel, Switzerland
| |
Collapse
|
17
|
Yamaguchi E, Yao J, Aymond A, Chrisey DB, Nieman GF, Bates JHT, Gaver DP. Electric Cell-Substrate Impedance Sensing (ECIS) as a Platform for Evaluating Barrier-Function Susceptibility and Damage from Pulmonary Atelectrauma. BIOSENSORS 2022; 12:390. [PMID: 35735538 PMCID: PMC9221382 DOI: 10.3390/bios12060390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/27/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022]
Abstract
Biophysical insults that either reduce barrier function (COVID-19, smoke inhalation, aspiration, and inflammation) or increase mechanical stress (surfactant dysfunction) make the lung more susceptible to atelectrauma. We investigate the susceptibility and time-dependent disruption of barrier function associated with pulmonary atelectrauma of epithelial cells that occurs in acute respiratory distress syndrome (ARDS) and ventilator-induced lung injury (VILI). This in vitro study was performed using Electric Cell-substrate Impedance Sensing (ECIS) as a noninvasive evaluating technique for repetitive stress stimulus/response on monolayers of the human lung epithelial cell line NCI-H441. Atelectrauma was mimicked through recruitment/derecruitment (RD) of a semi-infinite air bubble to the fluid-occluded micro-channel. We show that a confluent monolayer with a high level of barrier function is nearly impervious to atelectrauma for hundreds of RD events. Nevertheless, barrier function is eventually diminished, and after a critical number of RD insults, the monolayer disintegrates exponentially. Confluent layers with lower initial barrier function are less resilient. These results indicate that the first line of defense from atelectrauma resides with intercellular binding. After disruption, the epithelial layer community protection is diminished and atelectrauma ensues. ECIS may provide a platform for identifying damaging stimuli, ventilation scenarios, or pharmaceuticals that can reduce susceptibility or enhance barrier-function recovery.
Collapse
Affiliation(s)
- Eiichiro Yamaguchi
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA; (J.Y.); (A.A.)
| | - Joshua Yao
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA; (J.Y.); (A.A.)
| | - Allison Aymond
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA; (J.Y.); (A.A.)
| | - Douglas B. Chrisey
- Department of Physics and Engineering Physics, Tulane University, New Orleans, LA 70118, USA;
| | - Gary F. Nieman
- Department of Surgery, Upstate Medical University, Syracuse, NY 13210, USA;
| | - Jason H. T. Bates
- Department of Medicine, University of Vermont, Burlington, VT 05405, USA;
| | - Donald P. Gaver
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA; (J.Y.); (A.A.)
| |
Collapse
|
18
|
Different Effects of Cigarette Smoke, Heated Tobacco Product and E-Cigarette Vapour on Orbital Fibroblasts in Graves' Orbitopathy; a Study by Real Time Cell Electronic Sensing. Molecules 2022; 27:molecules27093001. [PMID: 35566351 PMCID: PMC9102878 DOI: 10.3390/molecules27093001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/28/2022] [Accepted: 05/05/2022] [Indexed: 11/17/2022] Open
Abstract
Thyroid autoimmunity in Graves’ disease (GD) is accompanied by Graves’ orbitopathy (GO) in 40% of the cases. Orbital fibroblasts (OF) play a key role in the pathogenesis and cigarette smoking is a known deteriorating factor. Alongside conventional cigarettes (CC) new alternatives became available for smokers, including heated tobacco products (HTP) and E-cigarettes (ECIG). We aimed to study the cellular effects of smoke extracts (SE) in orbital fibroblasts. Primary OF cultures from GO and NON-GO orbits were exposed to different concentrations of SE (1%, 50%) and the changes were followed using Real Time Cell Electronic Sensing (RT-CES). Untreated GO and NON-GO cells had different maximum cell index (CI) values of 3.3 and 2.79 respectively (p < 0.0001). CC, HTP and ECIG treated NON-GO fibroblasts exhibited peak CIs of 2.62, 3.32 and 3.41 while treated GO cells’ CIs were higher, 5.38, 6.25 and 6.33, respectively (p < 0.0001). The metabolic activity (MTT) decreased (p < 0.001) and hyaluronan production doubled (p < 0.02) after 50% of CC SE treatment in all cell cultures. GO fibroblasts were more sensitive to low concentration SE then NON-GO fibroblasts (p < 0.0001). The studied SEs exerted different effects. RT-CES is a sensitive technique to detect the effects of very low concentration of SE on fibroblasts.
Collapse
|
19
|
Hung YH, Chiu WC, Fuh SR, Lai YT, Tung TH, Huang CC, Lo CM. ECIS Based Electric Fence Method for Measurement of Human Keratinocyte Migration on Different Substrates. BIOSENSORS 2022; 12:bios12050293. [PMID: 35624596 PMCID: PMC9138674 DOI: 10.3390/bios12050293] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/28/2022] [Accepted: 05/01/2022] [Indexed: 05/28/2023]
Abstract
Electric Cell-substrate Impedance Sensing (ECIS) is an impedance-based, real-time, and label-free measuring system for monitoring cellular activities in tissue culture. Previously, ECIS wound healing assay has been used to wound cells with high electric current and monitor the subsequent cell migration. In this study, we applied ECIS electric fence (EF) method, an alternative to electrical wounding, to assess the effects of different surface coatings on human keratinocyte (HaCaT) migration. The EF prevents inoculated cells from attaching or migrating to the fenced electrode surface while maintaining the integrity of the surface coating. After the EF is turned off, cells migrate into the cell-free area, and the increase in measured impedance is monitored. We cultured HaCaT cells on gold electrodes without coating or coated with poly-L-lysin (PLL), poly-D-lysine (PDL), or type-I collagen. We quantified migration rates according to the different slopes in the impedance time series. It was observed that either poly-L-lysine (PLL) or poly-D-lysine (PDL) limits cell adhesion and migration rates. Furthermore, the surface charge of the coated substrate in the culture condition positively correlates with the cell adhesion and migration process. Our results indicate that the EF method is useful for determining cell migration rates on specific surface coatings.
Collapse
Affiliation(s)
- Yu-Han Hung
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| | - Wei-Chih Chiu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| | - Shyh-Rong Fuh
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
- Department of Aesthetic Medicine, Chen Hsin General Hospital, Taipei 112, Taiwan
| | - Yi-Ting Lai
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| | - Tse-Hua Tung
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| | - Chun-Chung Huang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| | - Chun-Min Lo
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| |
Collapse
|
20
|
Hoare D, Tsiamis A, Marland JRK, Czyzewski J, Kirimi MT, Holsgrove M, Russell E, Neale SL, Mirzai N, Mitra S, Mercer JR. Predicting Cardiovascular Stent Complications Using Self-Reporting Biosensors for Noninvasive Detection of Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105285. [PMID: 35322587 PMCID: PMC9130883 DOI: 10.1002/advs.202105285] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/03/2022] [Indexed: 06/14/2023]
Abstract
Self-reporting implantable medical devices are the future of cardiovascular healthcare. Cardiovascular complications such as blocked arteries that lead to the majority of heart attacks and strokes are frequently treated with inert metal stents that reopen affected vessels. Stents frequently re-block after deployment due to a wound response called in-stent restenosis (ISR). Herein, an implantable miniaturized sensor and telemetry system are developed that can detect this process, discern the different cell types associated with ISR, distinguish sub plaque components as demonstrated with ex vivo samples, and differentiate blood from blood clot, all on a silicon substrate making it suitable for integration onto a vascular stent. This work shows that microfabricated sensors can provide clinically relevant information in settings closer to physiological conditions than previous work with cultured cells.
Collapse
Affiliation(s)
- Daniel Hoare
- Institute of Cardiovascular and Medical Sciences/British Heart FoundationUniversity of GlasgowGlasgowUK
| | - Andreas Tsiamis
- School of EngineeringInstitute for Integrated Micro and Nano SystemsUniversity of EdinburghEdinburghUK
| | - Jamie R. K. Marland
- School of EngineeringInstitute for Integrated Micro and Nano SystemsUniversity of EdinburghEdinburghUK
| | - Jakub Czyzewski
- BioElectronics UnitCollege of MedicalVeterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Mahmut T. Kirimi
- Centre for Medical and Industrial UltrasonicsJames Watt School of EngineeringUniversity of GlasgowGlasgowUK
| | - Michael Holsgrove
- BioElectronics UnitCollege of MedicalVeterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Ewan Russell
- Centre for Medical and Industrial UltrasonicsJames Watt School of EngineeringUniversity of GlasgowGlasgowUK
| | - Steven L. Neale
- Centre for Medical and Industrial UltrasonicsJames Watt School of EngineeringUniversity of GlasgowGlasgowUK
| | - Nosrat Mirzai
- BioElectronics UnitCollege of MedicalVeterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Srinjoy Mitra
- School of EngineeringInstitute for Integrated Micro and Nano SystemsUniversity of EdinburghEdinburghUK
| | - John R. Mercer
- Institute of Cardiovascular and Medical Sciences/British Heart FoundationUniversity of GlasgowGlasgowUK
| |
Collapse
|
21
|
Multielectrode biosensor chip for spatial resolution screening of 3D cell models based on microcavity arrays. Biosens Bioelectron 2022; 202:114010. [DOI: 10.1016/j.bios.2022.114010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/24/2021] [Accepted: 01/14/2022] [Indexed: 11/18/2022]
|
22
|
Rebl H, Sawade M, Hein M, Bergemann C, Wende M, Lalk M, Langer P, Emmert S, Nebe B. Synergistic effect of plasma-activated medium and novel indirubin derivatives on human skin cancer cells by activation of the AhR pathway. Sci Rep 2022; 12:2528. [PMID: 35169210 PMCID: PMC8847430 DOI: 10.1038/s41598-022-06523-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 01/27/2022] [Indexed: 01/07/2023] Open
Abstract
Due to the increasing number of human skin cancers and the limited effectiveness of therapies, research into innovative therapeutic approaches is of enormous clinical interest. In recent years, the use of cold atmospheric pressure plasma has become increasingly important as anti-cancer therapy. The combination of plasma with small molecules offers the potential of an effective, tumour-specific, targeted therapy. The synthesised glycosylated and non glycosylated thia-analogous indirubin derivatives KD87 and KD88, respectively, were first to be investigated for their pharmaceutical efficacy in comparison with Indirubin-3'-monoxime (I3M) on human melanoma (A375) and squamous cell carcinoma (A431) cells. In combinatorial studies with plasma-activated medium (PAM) and KD87 we determined significantly decreased cell viability and cell adhesion. Cell cycle analyses revealed a marked G2/M arrest by PAM and a clear apoptotic effect by the glycosylated indirubin derivative KD87 in both cell lines and thus a synergistic anti-cancer effect. I3M had a pro-apoptotic effect only in A431 cells, so we hypothesize a different mode of action of the indirubin derivatives in the two skin cancer cells, possibly due to a different level of the aryl hydrocarbon receptor and an activation of this pathway by nuclear translocation of this receptor and subsequent activation of gene expression.
Collapse
Affiliation(s)
- Henrike Rebl
- grid.413108.f0000 0000 9737 0454Department of Cell Biology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Marie Sawade
- grid.413108.f0000 0000 9737 0454Department of Cell Biology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Martin Hein
- grid.10493.3f0000000121858338Institute for Chemistry, University of Rostock, 18059 Rostock, Germany
| | - Claudia Bergemann
- grid.413108.f0000 0000 9737 0454Department of Cell Biology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Manuela Wende
- grid.5603.0Institute for Biochemistry, University of Greifswald, 17487 Greifswald, Germany
| | - Michael Lalk
- grid.5603.0Institute for Biochemistry, University of Greifswald, 17487 Greifswald, Germany
| | - Peter Langer
- grid.10493.3f0000000121858338Institute for Chemistry, University of Rostock, 18059 Rostock, Germany
| | - Steffen Emmert
- grid.413108.f0000 0000 9737 0454Clinic and Polyclinic for Dermatology and Venerology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Barbara Nebe
- grid.413108.f0000 0000 9737 0454Department of Cell Biology, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
23
|
Zhang Y, Murakami K, Borra VJ, Ozen MO, Demirci U, Nakamura T, Esfandiari L. A Label-Free Electrical Impedance Spectroscopy for Detection of Clusters of Extracellular Vesicles Based on Their Unique Dielectric Properties. BIOSENSORS 2022; 12:bios12020104. [PMID: 35200364 PMCID: PMC8869858 DOI: 10.3390/bios12020104] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 06/01/2023]
Abstract
Extracellular vesicles (EVs) have gained considerable attention as vital circulating biomarkers since their structure and composition resemble the originating cells. The investigation of EVs' biochemical and biophysical properties is of great importance to map them to their parental cells and to better understand their functionalities. In this study, a novel frequency-dependent impedance measurement system has been developed to characterize EVs based on their unique dielectric properties. The system is composed of an insulator-based dielectrophoretic (iDEP) device to entrap and immobilize a cluster of vesicles followed by utilizing electrical impedance spectroscopy (EIS) to measure their impedance at a wide frequency spectrum, aiming to analyze both their membrane and cytosolic charge-dependent contents. The EIS was initially utilized to detect nano-size vesicles with different biochemical compositions, including liposomes synthesized with different lipid compositions, as well as EVs and lipoproteins with similar biophysical properties but dissimilar biochemical properties. Moreover, EVs derived from the same parental cells but treated with different culture conditions were characterized to investigate the correlation of impedance changes with biochemical properties and functionality in terms of pro-inflammatory responses. The system also showed the ability to discriminate between EVs derived from different cellular origins as well as among size-sorted EVs harbored from the same cellular origin. This proof-of-concept approach is the first step towards utilizing EIS as a label-free, non-invasive, and rapid sensor for detection and characterization of pathogenic EVs and other nanovesicles in the future.
Collapse
Affiliation(s)
- Yuqian Zhang
- Department of Surgery, Division of Surgical Research, Mayo Clinic, Rochester, MN 55905, USA;
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Kazutoshi Murakami
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (K.M.); (V.J.B.); (T.N.)
| | - Vishnupriya J. Borra
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (K.M.); (V.J.B.); (T.N.)
| | - Mehmet Ozgun Ozen
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Stanford University, Palo Alto, CA 94305, USA; (M.O.O.); (U.D.)
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Stanford University, Palo Alto, CA 94305, USA; (M.O.O.); (U.D.)
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Takahisa Nakamura
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (K.M.); (V.J.B.); (T.N.)
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
- Department of Metabolic Bioregulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8577, Miyagi, Japan
| | - Leyla Esfandiari
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
- Department of Electrical Engineering and Computer Science, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
- Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
24
|
Choi J, Mathew S, Oerter S, Appelt-Menzel A, Hansmann J, Schmitz T. Online Measurement System for Dynamic Flow Bioreactors to Study Barrier Integrity of hiPSC-Based Blood-Brain Barrier In Vitro Models. Bioengineering (Basel) 2022; 9:bioengineering9010039. [PMID: 35049748 PMCID: PMC8773345 DOI: 10.3390/bioengineering9010039] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 12/31/2022] Open
Abstract
Electrochemical impedance spectroscopy (EIS) is a noninvasive, reliable, and efficient method to analyze the barrier integrity of in vitro tissue models. This well-established tool is used most widely to quantify the transendothelial/epithelial resistance (TEER) of Transwell-based models cultured under static conditions. However, dynamic culture in bioreactors can achieve advanced cell culture conditions that mimic a more tissue-specific environment and stimulation. This requires the development of culture systems that also allow for the assessment of barrier integrity under dynamic conditions. Here, we present a bioreactor system that is capable of the automated, continuous, and non-invasive online monitoring of cellular barrier integrity during dynamic culture. Polydimethylsiloxane (PDMS) casting and 3D printing were used for the fabrication of the bioreactors. Additionally, attachable electrodes based on titanium nitride (TiN)-coated steel tubes were developed to perform EIS measurements. In order to test the monitored bioreactor system, blood–brain barrier (BBB) in vitro models derived from human-induced pluripotent stem cells (hiPSC) were cultured for up to 7 days. We applied equivalent electrical circuit fitting to quantify the electrical parameters of the cell layer and observed that TEER gradually decreased over time from 2513 Ω·cm2 to 285 Ω·cm2, as also specified in the static control culture. Our versatile system offers the possibility to be used for various dynamic tissue cultures that require a non-invasive monitoring system for barrier integrity.
Collapse
Affiliation(s)
- Jihyoung Choi
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (S.M.); (J.H.); (T.S.)
- Correspondence: (J.C.); (A.A.-M.)
| | - Sanjana Mathew
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (S.M.); (J.H.); (T.S.)
| | - Sabrina Oerter
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research, Röntgenring 11, 97070 Würzburg, Germany;
| | - Antje Appelt-Menzel
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (S.M.); (J.H.); (T.S.)
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research, Röntgenring 11, 97070 Würzburg, Germany;
- Correspondence: (J.C.); (A.A.-M.)
| | - Jan Hansmann
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (S.M.); (J.H.); (T.S.)
- Faculty of Electronics, University of Applied Science Würzburg-Schweinfurt, Ignaz-Schön-Straße 11, 97421 Schweinfurt, Germany
| | - Tobias Schmitz
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (S.M.); (J.H.); (T.S.)
| |
Collapse
|
25
|
Pitsalidis C, Pappa AM, Boys AJ, Fu Y, Moysidou CM, van Niekerk D, Saez J, Savva A, Iandolo D, Owens RM. Organic Bioelectronics for In Vitro Systems. Chem Rev 2021; 122:4700-4790. [PMID: 34910876 DOI: 10.1021/acs.chemrev.1c00539] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bioelectronics have made strides in improving clinical diagnostics and precision medicine. The potential of bioelectronics for bidirectional interfacing with biology through continuous, label-free monitoring on one side and precise control of biological activity on the other has extended their application scope to in vitro systems. The advent of microfluidics and the considerable advances in reliability and complexity of in vitro models promise to eventually significantly reduce or replace animal studies, currently the gold standard in drug discovery and toxicology testing. Bioelectronics are anticipated to play a major role in this transition offering a much needed technology to push forward the drug discovery paradigm. Organic electronic materials, notably conjugated polymers, having demonstrated technological maturity in fields such as solar cells and light emitting diodes given their outstanding characteristics and versatility in processing, are the obvious route forward for bioelectronics due to their biomimetic nature, among other merits. This review highlights the advances in conjugated polymers for interfacing with biological tissue in vitro, aiming ultimately to develop next generation in vitro systems. We showcase in vitro interfacing across multiple length scales, involving biological models of varying complexity, from cell components to complex 3D cell cultures. The state of the art, the possibilities, and the challenges of conjugated polymers toward clinical translation of in vitro systems are also discussed throughout.
Collapse
Affiliation(s)
- Charalampos Pitsalidis
- Department of Physics, Khalifa University of Science and Technology, P.O. Box 127788, Abu Dhabi 127788, UAE.,Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Anna-Maria Pappa
- Department of Biomedical Engineering, Khalifa University of Science and Technology, P.O. Box 127788, Abu Dhabi 127788, UAE
| | - Alexander J Boys
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Ying Fu
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.,Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, Glasgow G1 1RD, U.K
| | - Chrysanthi-Maria Moysidou
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Douglas van Niekerk
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Janire Saez
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.,Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Avenida Miguel de Unamuno, 3, 01006 Vitoria-Gasteiz, Spain.,Ikerbasque, Basque Foundation for Science, E-48011 Bilbao, Spain
| | - Achilleas Savva
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Donata Iandolo
- INSERM, U1059 Sainbiose, Université Jean Monnet, Mines Saint-Étienne, Université de Lyon, 42023 Saint-Étienne, France
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| |
Collapse
|
26
|
Validation of a Lab-on-Chip Assay for Measuring Sorafenib Effectiveness on HCC Cell Proliferation. Int J Mol Sci 2021; 22:ijms222313090. [PMID: 34884894 PMCID: PMC8658471 DOI: 10.3390/ijms222313090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly lethal cancer, and although a few drugs are available for treatment, therapeutic effectiveness is still unsatisfactory. New drugs are urgently needed for hepatocellular carcinoma (HCC) patients. In this context, reliable preclinical assays are of paramount importance to screen the effectiveness of new drugs and, in particular, measure their effects on HCC cell proliferation. However, cell proliferation measurement is a time-consuming and operator-dependent procedure. The aim of this study was to validate an engineered miniaturized on-chip platform for real-time, non-destructive cell proliferation assays and drug screening. The effectiveness of Sorafenib, the first-line drug mainly used for patients with advanced HCC, was tested in parallel, comparing the gold standard 96-well-plate assay and our new lab-on-chip platform. Results from the lab-on-chip are consistent in intra-assay replicates and comparable to the output of standard crystal violet proliferation assays for assessing Sorafenib effectiveness on HCC cell proliferation. The miniaturized platform presents several advantages in terms of lesser reagents consumption, operator time, and costs, as well as overcoming a number of technical and operator-dependent pitfalls. Moreover, the number of cells required is lower, a relevant issue when primary cell cultures are used. In conclusion, the availability of inexpensive on-chip assays can speed up drug development, especially by using patient-derived samples to take into account disease heterogeneity and patient-specific characteristics.
Collapse
|
27
|
Mehta N, Shaik S, Prasad A, Chaichi A, Sahu SP, Liu Q, Hasan SMA, Sheikh E, Donnarumma F, Murray KK, Fu X, Devireddy R, Gartia MR. Multimodal Label-Free Monitoring of Adipogenic Stem Cell Differentiation Using Endogenous Optical Biomarkers. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2103955. [PMID: 34924914 PMCID: PMC8680429 DOI: 10.1002/adfm.202103955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Indexed: 05/13/2023]
Abstract
Stem cell-based therapies carry significant promise for treating human diseases. However, clinical translation of stem cell transplants for effective treatment requires precise non-destructive evaluation of the purity of stem cells with high sensitivity (<0.001% of the number of cells). Here, a novel methodology using hyperspectral imaging (HSI) combined with spectral angle mapping-based machine learning analysis is reported to distinguish differentiating human adipose-derived stem cells (hASCs) from control stem cells. The spectral signature of adipogenesis generated by the HSI method enables identifying differentiated cells at single-cell resolution. The label-free HSI method is compared with the standard techniques such as Oil Red O staining, fluorescence microscopy, and qPCR that are routinely used to evaluate adipogenic differentiation of hASCs. HSI is successfully used to assess the abundance of adipocytes derived from transplanted cells in a transgenic mice model. Further, Raman microscopy and multiphoton-based metabolic imaging is performed to provide complementary information for the functional imaging of the hASCs. Finally, the HSI method is validated using matrix-assisted laser desorption/ionization-mass spectrometry imaging of the stem cells. The study presented here demonstrates that multimodal imaging methods enable label-free identification of stem cell differentiation with high spatial and chemical resolution.
Collapse
Affiliation(s)
- Nishir Mehta
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Shahensha Shaik
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Alisha Prasad
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Ardalan Chaichi
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Sushant P Sahu
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Qianglin Liu
- LSU AgCenter, School of Animal Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Syed Mohammad Abid Hasan
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Elnaz Sheikh
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Fabrizio Donnarumma
- Department of Chemistry, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Kermit K Murray
- Department of Chemistry, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Xing Fu
- LSU AgCenter, School of Animal Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Ram Devireddy
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
28
|
Iwakura T, Marschner JA, Zhao ZB, Świderska MK, Anders HJ. Electric cell-substrate impedance sensing in kidney research. Nephrol Dial Transplant 2021; 36:216-223. [PMID: 31598727 DOI: 10.1093/ndt/gfz191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/18/2019] [Indexed: 12/20/2022] Open
Abstract
Electric cell-substrate impedance sensing (ECIS) is a quantitative, label-free, non-invasive analytical method allowing continuous monitoring of the behaviour of adherent cells by online recording of transcellular impedance. ECIS offers a wide range of practical applications to study cell proliferation, migration, differentiation, toxicity and monolayer barrier integrity. All of these applications are relevant for basic kidney research, e.g. on endothelial cells, tubular and glomerular epithelial cells. This review gives an overview on the fundamental principles of the ECIS technology. We name strengths and remaining hurdles for practical applications, present an ECIS array reuse protocol, and review its past, present and potential future contributions to preclinical kidney research.
Collapse
Affiliation(s)
- Takamasa Iwakura
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany.,Internal Medicine I, Division of Nephrology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Julian A Marschner
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Zhi Bo Zhao
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Monika Katarzyna Świderska
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Hans-Joachim Anders
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| |
Collapse
|
29
|
PEDOT:PSS organic electrochemical transistors for electrical cell-substrate impedance sensing down to single cells. Biosens Bioelectron 2021; 180:113101. [DOI: 10.1016/j.bios.2021.113101] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/06/2021] [Accepted: 02/15/2021] [Indexed: 12/26/2022]
|
30
|
Shah FJ, Caviglia C, Zór K, Carminati M, Ferrari G, Sampietro M, Martínez-Serrano A, Emnéus JK, Heiskanen AR. Impedance-based Real-time Monitoring of Neural Stem Cell Differentiation. JOURNAL OF ELECTRICAL BIOIMPEDANCE 2021; 12:34-49. [PMID: 34966467 PMCID: PMC8667812 DOI: 10.2478/joeb-2021-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Indexed: 06/14/2023]
Abstract
We present here the first impedance-based characterization of the differentiation process of two human mesencephalic fetal neural stem lines. The two dopaminergic neural stem cell lines used in this study, Lund human mesencephalic (LUHMES) and human ventral mesencephalic (hVM1 Bcl-XL), have been developed for the study of Parkinsonian pathogenesis and its treatment using cell replacement therapy. We show that if only relying on impedance magnitude analysis, which is by far the most usual approach in, e.g., cytotoxicity evaluation and drug screening applications, one may not be able to distinguish whether the neural stem cells in a population are proliferating or differentiating. However, the presented results highlight that equivalent circuit analysis can provide detailed information on cellular behavior, e.g. simultaneous changes in cell morphology, cell-cell contacts, and cell adhesion during formation of neural projections, which are the fundamental behavioral differences between proliferating and differentiating neural stem cells. Moreover, our work also demonstrates the sensitivity of impedance-based monitoring with capability to provide information on changes in cellular behavior in relation to proliferation and differentiation. For both of the studied cell lines, in already two days (one day after induction of differentiation) equivalent circuit analysis was able to show distinction between proliferation and differentiation conditions, which is significantly earlier than by microscopic imaging. This study demonstrates the potential of impedance-based monitoring as a technique of choice in the study of stem cell behavior, laying the foundation for screening assays to characterize stem cell lines and testing the efficacy epigenetic control.
Collapse
Affiliation(s)
- F. J. Shah
- Department of Micro- and Nanotechnology, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
- Particle Analytical ApS, Agern Allé 3, 2970 Hørsholm, Denmark
| | - C. Caviglia
- Department of Micro- and Nanotechnology, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
- Radiometer Medical ApS, Åkandevej 21, 2700 Brønshøj, Denmark
| | - K. Zór
- Department of Micro- and Nanotechnology, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics, Department of Health Technology, Technical University of Denmark, Ørsteds Plads, Building 345C, 2800Kongens Lyngby, Denmark
| | - M. Carminati
- Dipartimento di Elettronica, Informazione e Bioingegneria - DEIB, Politecnico di Milano, P.za L. da Vinci 32, 20133Milano, Italy
| | - G. Ferrari
- Dipartimento di Elettronica, Informazione e Bioingegneria - DEIB, Politecnico di Milano, P.za L. da Vinci 32, 20133Milano, Italy
| | - M. Sampietro
- Dipartimento di Elettronica, Informazione e Bioingegneria - DEIB, Politecnico di Milano, P.za L. da Vinci 32, 20133Milano, Italy
| | - A. Martínez-Serrano
- Department of Molecular Neuropathology, Center of Molecular Biology Severo Ochoa, Universidad Autónoma de Madrid, Calle Nicolás Cabrera 1, Cantoblanco, 28049Madrid, Spain
| | - J. K. Emnéus
- Department of Micro- and Nanotechnology, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
- Present affiliation: Department of Biotechnology and Biomedicine, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
| | - A. R. Heiskanen
- Department of Micro- and Nanotechnology, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
- Present affiliation: Department of Biotechnology and Biomedicine, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
| |
Collapse
|
31
|
Giduthuri AT, Theodossiou SK, Schiele NR, Srivastava SK. Dielectrophoresis as a tool for electrophysiological characterization of stem cells. BIOPHYSICS REVIEWS 2020; 1:011304. [PMID: 38505626 PMCID: PMC10903368 DOI: 10.1063/5.0025056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/20/2020] [Indexed: 03/21/2024]
Abstract
Dielectrophoresis (DEP), a nonlinear electrokinetic technique caused by Maxwell-Wagner interfacial polarization of neutral particles in an electrolyte solution, is a powerful cell manipulation method used widely for various applications such as enrichment, trapping, and sorting of heterogeneous cell populations. While conventional cell characterization and sorting methods require tagging or labeling of cells, DEP has the potential to manipulate cells in a label-free way. Due to its unique ability to characterize and sort cells without the need of labeling, there is renewed interest in using DEP for stem cell research and regenerative medicine. Stem cells have the potential to differentiate into various lineages, but achieving homogeneous cell phenotypes from an initially heterogeneous cell population is a challenge. Using DEP to efficiently and affordably identify, sort, and enrich either undifferentiated or differentiated stem cell populations in a label-free way would advance their potential uses for applications in tissue engineering and regenerative medicine. This review summarizes recent, significant research findings regarding the electrophysiological characterization of stem cells, with a focus on cellular dielectric properties, i.e., permittivity and conductivity, and on studies that have obtained these measurements using techniques that preserve cell viability, such as crossover frequency. Potential applications for DEP in regenerative medicine are also discussed. Overall, DEP is a promising technique and, when used to characterize, sort, and enrich stem cells, will advance stem cell-based regenerative therapies.
Collapse
Affiliation(s)
- Anthony T. Giduthuri
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| | - Sophia K. Theodossiou
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| | - Nathan R. Schiele
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| | - Soumya K. Srivastava
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho 83844, USA
| |
Collapse
|
32
|
Wei X, Zhuang L, Li H, He C, Wan H, Hu N, Wang P. Advances in Multidimensional Cardiac Biosensing Technologies: From Electrophysiology to Mechanical Motion and Contractile Force. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2005828. [PMID: 33230867 DOI: 10.1002/smll.202005828] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 06/11/2023]
Abstract
Cardiovascular disease is currently a leading killer to human, while drug-induced cardiotoxicity remains the main cause of the withdrawal and attrition of drugs. Taking clinical correlation and throughput into account, cardiomyocyte is perfect as in vitro cardiac model for heart disease modeling, drug discovery, and cardiotoxicity assessment by accurately measuring the physiological multiparameters of cardiomyocytes. Remarkably, cardiomyocytes present both electrophysiological and biomechanical characteristics due to the unique excitation-contraction coupling, which plays a significant role in studying the cardiomyocytes. This review mainly focuses on the recent advances of biosensing technologies for the 2D and 3D cardiac models with three special properties: electrophysiology, mechanical motion, and contractile force. These high-performance multidimensional cardiac models are popular and effective to rebuild and mimic the heart in vitro. To help understand the high-quality and accurate physiologies, related detection techniques are highly demanded, from microtechnology to nanotechnology, from extracellular to intracellular recording, from multiple cells to single cell, and from planar to 3D models. Furthermore, the characteristics, advantages, limitations, and applications of these cardiac biosensing technologies, as well as the future development prospects should contribute to the systematization and expansion of knowledge.
Collapse
Affiliation(s)
- Xinwei Wei
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Liujing Zhuang
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Hongbo Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chuanjiang He
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
| | - Hao Wan
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ping Wang
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| |
Collapse
|
33
|
Zhang Z, Zheng T, Zhu R. Long-term and label-free monitoring for osteogenic differentiation of mesenchymal stem cells using force sensor and impedance measurement. J Mater Chem B 2020; 8:9913-9920. [PMID: 33034334 DOI: 10.1039/d0tb01968b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Stem cells have attracted increasing research interest in the field of regenerative medicine due to their unique abilities to differentiate into multiple cell lineages. Label-free, real-time, and long-term monitoring for stem cell differentiation is requisite in studying directional differentiation and development mechanisms for tissue engineering applications, but a great challenge because of the rigorous demands for sensitivity, stability and biocompatibility of devices. In this article, a label-free and real-time monitoring approach using a zinc oxide (ZnO) nanorod field effect transistor (FET) is proposed to detect cell traction forces (CTFs) exerted by cells on underlying substrates. The ZnO nanorod FET with the approach of difference-frequency lock-in detection achieves high sensitivity, good stability, and excellent biocompatibility, by which real-time and long-term (over 20 days) monitoring of cellular mechanical changes in osteogenic differentiation of mesenchymal stem cells (MSCs) is successfully achieved. We also employ electrical impedance monitoring using microelectrode array chips and microscopic observation to investigate cell migration and nodular aggregation behaviors of MSCs in osteogenic differentiation. Various biochemical assays including alkaline phosphatase (ALP), osteopontin expression and alizarin red staining are utilized to verify osteogenic differentiation of MSCs. We propose a combination of cell traction force measurement, impedance measurement and microscopic observation to provide multimodal profiling of cell morphology, and cellular biomechanical and electrophysiological phenotypes, which can track cellular dynamics in stem cell development and help to deeply understand the mechanism of osteogenic differentiation.
Collapse
Affiliation(s)
- Zhizhong Zhang
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China.
| | - Tianyang Zheng
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China.
| | - Rong Zhu
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
34
|
Abayzeed SA. Plasmonic-based impedance microspectroscopy of optically heterogeneous samples. BIOMEDICAL OPTICS EXPRESS 2020; 11:6168-6180. [PMID: 33282482 PMCID: PMC7687972 DOI: 10.1364/boe.395474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 05/20/2023]
Abstract
A robust impedance microscopy technique is presented. This optical tool enables high resolution imaging of electrical properties with promising biophysical applications. The underlying principle is that surface plasmon resonance (SPR) sensors are able to measure perturbations of surface charge density and therefore can be used to compute the impedance of surface-adhered cells. However, the ability to perform reliable quantitative impedance imaging is affected by the optical heterogeneity of the cell-sensor interface. To address this issue, a novel method for quantitative time-resolved resonance angle tracking is developed and applied to correct for the effect of the optical properties. To demonstrate the capability of this technique, impedance microspectroscopy of bovine serum albumin (BSA) patterns was performed enabling measurements of capacitance with submicroscopic resolution. The work presented offers an impedance microspectroscopy method that will create new avenues in studying the electrical properties of single cells and biomolecules as well as bio-electrical currents.
Collapse
|
35
|
Bussooa A. Characterising Vascular Cell Monolayers Using Electrochemical Impedance Spectroscopy and a Novel Electroanalytical Plot. Nanotechnol Sci Appl 2020; 13:89-101. [PMID: 33061321 PMCID: PMC7520662 DOI: 10.2147/nsa.s266663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/27/2020] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Biological research relies on the culture of mammalian cells, which are prone to changes in phenotype during experiments involving several passages of cells. In regenerative medicine, specifically, there is an increasing need to expand the characterisation landscape for stem cells by identifying novel stable markers. This paper reports on a novel electric cell-substrate impedance sensing-based electroanalytical diagram which can be used for the "electrical characterisation" of cell monolayers consisting of smooth muscle cells, endothelial cells or co-culture. MATERIALS AND METHODS Interdigitated electrodes were microfabricated using standard cleanroom procedures and integrated into cell chambers. Electrochemical impedance spectroscopy data were acquired for 2 vascular cell types after they formed monolayers on the electrodes. RESULTS AND DISCUSSION A Mean impedance per unit area vs Mean phase plots provided a reproducible, visually obvious and statistically significant method of characterising cell monolayers. This electroanalytic diagram has never been used in previous papers, but it confirms findings by other research groups using similar approaches that the complex impedance spectra of different cell type are different. Further work is required to determine whether this method could be extended to other cell types, and if this is the case, a library of "signature spectra" could be generated for "electrical characterisation" of cells.
Collapse
Affiliation(s)
- Anubhav Bussooa
- BHF Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
| |
Collapse
|
36
|
Zhang Z, Zheng T, Zhu R. Microchip with Single-Cell Impedance Measurements for Monitoring Osteogenic Differentiation of Mesenchymal Stem Cells under Electrical Stimulation. Anal Chem 2020; 92:12579-12587. [PMID: 32859132 DOI: 10.1021/acs.analchem.0c02556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Effective induction methods and in situ monitoring are essential for studying the mechanism of biological responses in stem cell differentiation. This article proposes an induction method incorporating electrical stimulation under an inhomogeneous field with single-cell impedance monitoring for studying osteogenic differentiation of mesenchymal stem cells (MSCs) using a microchip. The microchip contains an array of sextupole-electrode units for implementing a combination of controllable electrical stimulation and single-cell impedance measurements. MSCs are inducted to osteogenic differentiation under electrical stimulation using quadrupole electrodes and single-cell impedances are monitored in situ using a pair of microelectrodes at each unit center. The proposed microchip adopts an array design to monitor a number of MSCs in parallel, which improves measurement throughput and facilitates to carry out statistic tests. We perform osteogenic differentiation of MSCs on the microchip with and without electrical stimulation meanwhile monitoring single-cell impedance in real time for 21 days. The recorded impedance results show the detailed characteristic change of MSCs at the single-cell level during osteogenic differentiation, which demonstrates a significant difference between the conditions with and without electrical stimulation. The cell morphology and various staining analyses are also used to validate osteogenesis and correlate with the impedance expression. Correlation analysis of the impedance measurement, cell morphology, and various staining assays proves the great acceleration effect of the proposed electrical stimulation on osteogenic differentiation of MSCs. The proposed impedance method can monitor the dynamic process of cell development and study heterogeneity of stem cell differentiation at the single-cell level.
Collapse
Affiliation(s)
- Zhizhong Zhang
- State Key Laboratory of Precision Measurements Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing 100084, China
| | - Tianyang Zheng
- State Key Laboratory of Precision Measurements Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing 100084, China
| | - Rong Zhu
- State Key Laboratory of Precision Measurements Technology and Instruments, Department of Precision Instrument, Tsinghua University, Beijing 100084, China
| |
Collapse
|
37
|
Kuo MH, Lee AC, Hsiao SH, Lin SE, Chiu YF, Yang LH, Yu CC, Chiou SH, Huang HN, Ko JC, Chou YT. Cross-talk between SOX2 and TGFβ Signaling Regulates EGFR-TKI Tolerance and Lung Cancer Dissemination. Cancer Res 2020; 80:4426-4438. [PMID: 32816907 DOI: 10.1158/0008-5472.can-19-3228] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 07/18/2020] [Accepted: 08/13/2020] [Indexed: 11/16/2022]
Abstract
Regulation of the stemness factor, SOX2, by cytokine stimuli controls self-renewal and differentiation in cells. Activating mutations in EGFR are proven therapeutic targets for tyrosine kinase inhibitors (TKI) in lung adenocarcinoma, but acquired resistance to TKIs inevitably occurs. The mechanism by which stemness and differentiation signaling emerge in lung cancers to affect TKI tolerance and lung cancer dissemination has yet to be elucidated. Here, we report that cross-talk between SOX2 and TGFβ signaling affects lung cancer cell plasticity and TKI tolerance. TKI treatment favored selection of lung cancer cells displaying mesenchymal morphology with deficient SOX2 expression, whereas SOX2 expression promoted TKI sensitivity and inhibited the mesenchymal phenotype. Preselection of EGFR-mutant lung cancer cells with the mesenchymal phenotype diminished SOX2 expression and TKI sensitivity, whereas SOX2 silencing induced vimentin, but suppressed BCL2L11, expression and promoted TKI tolerance. TGFβ stimulation downregulated SOX2 and induced epithelial-to-mesenchymal transdifferentiation accompanied by increased TKI tolerance, which can interfere with ectopic SOX2 expression. SOX2-positive lung cancer cells exhibited a lower dissemination capacity than their SOX2-negative counterparts. Tumors expressing low SOX2 and high vimentin signature were associated with worse survival outcomes in patients with EGFR mutations. These findings provide insights into how cancer cell plasticity regulated by SOX2 and TGFβ signaling affects EGFR-TKI tolerance and lung cancer dissemination. SIGNIFICANCE: These findings suggest the potential of SOX2 as a prognostic marker in EGFR-mutant lung cancer, as SOX2-mediated cell plasticity regulated by TGFβ stimulation and epigenetic control affects EGFR-TKI tolerance and cancer dissemination.
Collapse
Affiliation(s)
- Ming-Han Kuo
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - An-Chun Lee
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Shih-Hsin Hsiao
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Xinyi, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Xinyi, Taipei, Taiwan
| | - Sey-En Lin
- Department of Pathology, New Taipei City Municipal Tucheng Hospital (Chang Gung Memorial Hospital, Tucheng Branch, Taiwan), New Taipei City, Taiwan
| | - Yu-Fan Chiu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Li-Hao Yang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Cherng Yu
- Department of Medical Research, National Taiwan University Hospital, Zhongzheng, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Institute of Pharmacology, National Yang-Ming University, Beitou, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Beitou, Taipei, Taiwan
| | - Hsien-Neng Huang
- Department of Pathology, National Taiwan University Hospital, Hsin-Chu Branch, Hsinchu, Taiwan
| | - Jen-Chung Ko
- Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsinchu, Taiwan.
| | - Yu-Ting Chou
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
38
|
Li YCE, Lee IC. The Current Trends of Biosensors in Tissue Engineering. BIOSENSORS 2020; 10:E88. [PMID: 32756393 PMCID: PMC7459738 DOI: 10.3390/bios10080088] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 12/30/2022]
Abstract
Biosensors constitute selective, sensitive, and rapid tools for disease diagnosis in tissue engineering applications. Compared to standard enzyme-linked immunosorbent assay (ELISA) analytical technology, biosensors provide a strategy to real-time and on-site monitor micro biophysiological signals via a combination of biological, chemical, and physical technologies. This review summarizes the recent and significant advances made in various biosensor technologies for different applications of biological and biomedical interest, especially on tissue engineering applications. Different fabrication techniques utilized for tissue engineering purposes, such as computer numeric control (CNC), photolithographic, casting, and 3D printing technologies are also discussed. Key developments in the cell/tissue-based biosensors, biomolecular sensing strategies, and the expansion of several biochip approaches such as organs-on-chips, paper based-biochips, and flexible biosensors are available. Cell polarity and cell behaviors such as proliferation, differentiation, stimulation response, and metabolism detection are included. Biosensors for diagnosing tissue disease modes such as brain, heart, lung, and liver systems and for bioimaging are discussed. Finally, we discuss the challenges faced by current biosensing techniques and highlight future prospects of biosensors for tissue engineering applications.
Collapse
Affiliation(s)
- Yi-Chen Ethan Li
- Department of Chemical Engineering, Feng Chia University, Taichung 40724, Taiwan
| | - I-Chi Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| |
Collapse
|
39
|
Kasiviswanathan U, Poddar S, Kumar C, Jit S, Mahto SK, Sharma N. A portable standalone wireless electric cell-substrate impedance sensing (ECIS) system for assessing dynamic behavior of mammalian cells. J Anal Sci Technol 2020. [DOI: 10.1186/s40543-020-00223-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
40
|
Olmo A, Yuste Y, Serrano JA, Maldonado-Jacobi A, Pérez P, Huertas G, Pereira S, Yufera A, de la Portilla F. Electrical Modeling of the Growth and Differentiation of Skeletal Myoblasts Cell Cultures for Tissue Engineering. SENSORS 2020; 20:s20113152. [PMID: 32498394 PMCID: PMC7309147 DOI: 10.3390/s20113152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 11/16/2022]
Abstract
In tissue engineering, of utmost importance is the control of tissue formation, in order to form tissue constructs of clinical relevance. In this work, we present the use of an impedance spectroscopy technique for the real-time measurement of the dielectric properties of skeletal myoblast cell cultures. The processes involved in the growth and differentiation of these cell cultures in skeletal muscle are studied. A circuit based on the oscillation-based test technique was used, avoiding the use of high-performance circuitry or external input signals. The effect of electrical pulse stimulation applied to cell cultures was also studied. The technique proved useful for monitoring in real-time the processes of cell growth and estimating the fill factor of muscular stem cells. Impedance spectroscopy was also useful to study the real-time monitoring of cell differentiation, obtaining different oscillation amplitude levels for differentiated and undifferentiated cell cultures. Finally, an electrical model was implemented to better understand the physical properties of the cell culture and control the tissue formation process.
Collapse
Affiliation(s)
- Alberto Olmo
- Instituto de Microelectrónica de Sevilla, IMSE, CNM (CSIC, Universidad de Sevilla), Av. Américo Vespucio, sn 41092 Sevilla, Spain; (J.A.S.); (A.M.-J.); (P.P.); (G.H.); (A.Y.)
- Escuela Técnica Superior de Ingeniería Informática, Departamento de Tecnología Electrónica, Universidad de Sevilla, Av. Reina Mercedes, sn 41012 Sevilla, Spain
- Correspondence: ; Tel.: +34-954-55-43-25
| | - Yaiza Yuste
- Instituto de Biomedicina de Sevilla (IBIS), Campus Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot, s/n 41013, Sevilla, Spain; (Y.Y.); (S.P.); (F.d.l.P.)
| | - Juan Alfonso Serrano
- Instituto de Microelectrónica de Sevilla, IMSE, CNM (CSIC, Universidad de Sevilla), Av. Américo Vespucio, sn 41092 Sevilla, Spain; (J.A.S.); (A.M.-J.); (P.P.); (G.H.); (A.Y.)
| | - Andres Maldonado-Jacobi
- Instituto de Microelectrónica de Sevilla, IMSE, CNM (CSIC, Universidad de Sevilla), Av. Américo Vespucio, sn 41092 Sevilla, Spain; (J.A.S.); (A.M.-J.); (P.P.); (G.H.); (A.Y.)
| | - Pablo Pérez
- Instituto de Microelectrónica de Sevilla, IMSE, CNM (CSIC, Universidad de Sevilla), Av. Américo Vespucio, sn 41092 Sevilla, Spain; (J.A.S.); (A.M.-J.); (P.P.); (G.H.); (A.Y.)
- Escuela Técnica Superior de Ingeniería Informática, Departamento de Tecnología Electrónica, Universidad de Sevilla, Av. Reina Mercedes, sn 41012 Sevilla, Spain
| | - Gloria Huertas
- Instituto de Microelectrónica de Sevilla, IMSE, CNM (CSIC, Universidad de Sevilla), Av. Américo Vespucio, sn 41092 Sevilla, Spain; (J.A.S.); (A.M.-J.); (P.P.); (G.H.); (A.Y.)
- Facultad de Física, Departamento de Electrónica y Electromagnetismo, Universidad de Sevilla, Av. Reina Mercedes, sn 41012 Sevilla, Spain
| | - Sheila Pereira
- Instituto de Biomedicina de Sevilla (IBIS), Campus Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot, s/n 41013, Sevilla, Spain; (Y.Y.); (S.P.); (F.d.l.P.)
| | - Alberto Yufera
- Instituto de Microelectrónica de Sevilla, IMSE, CNM (CSIC, Universidad de Sevilla), Av. Américo Vespucio, sn 41092 Sevilla, Spain; (J.A.S.); (A.M.-J.); (P.P.); (G.H.); (A.Y.)
- Escuela Técnica Superior de Ingeniería Informática, Departamento de Tecnología Electrónica, Universidad de Sevilla, Av. Reina Mercedes, sn 41012 Sevilla, Spain
| | - Fernando de la Portilla
- Instituto de Biomedicina de Sevilla (IBIS), Campus Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot, s/n 41013, Sevilla, Spain; (Y.Y.); (S.P.); (F.d.l.P.)
| |
Collapse
|
41
|
Liang W, Yang X, Wang J, Wang Y, Yang W, Liu L. Determination of Dielectric Properties of Cells using AC Electrokinetic-based Microfluidic Platform: A Review of Recent Advances. MICROMACHINES 2020; 11:E513. [PMID: 32438680 PMCID: PMC7281274 DOI: 10.3390/mi11050513] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/18/2022]
Abstract
Cell dielectric properties, a type of intrinsic property of cells, can be used as electrophysiological biomarkers that offer a label-free way to characterize cell phenotypes and states, purify clinical samples, and identify target cancer cells. Here, we present a review of the determination of cell dielectric properties using alternating current (AC) electrokinetic-based microfluidic mechanisms, including electro-rotation (ROT) and dielectrophoresis (DEP). The review covers theoretically how ROT and DEP work to extract cell dielectric properties. We also dive into the details of differently structured ROT chips, followed by a discussion on the determination of cell dielectric properties and the use of these properties in bio-related applications. Additionally, the review offers a look at the future challenges facing the AC electrokinetic-based microfluidic platform in terms of acquiring cell dielectric parameters. Our conclusion is that this platform will bring biomedical and bioengineering sciences to the next level and ultimately achieve the shift from lab-oriented research to real-world applications.
Collapse
Affiliation(s)
- Wenfeng Liang
- School of Mechanical Engineering, Shenyang Jianzhu University, Shenyang 110168, China; (X.Y.); (J.W.)
| | - Xieliu Yang
- School of Mechanical Engineering, Shenyang Jianzhu University, Shenyang 110168, China; (X.Y.); (J.W.)
| | - Junhai Wang
- School of Mechanical Engineering, Shenyang Jianzhu University, Shenyang 110168, China; (X.Y.); (J.W.)
| | - Yuechao Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China;
| | - Wenguang Yang
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China;
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China;
| |
Collapse
|
42
|
Khorolsuren Z, Lang O, Pallinger E, Foldes A, Szabolcs GG, Varga G, Mezo G, Vag J, Kohidai L. Functional and cell surface characteristics of periodontal ligament cells (PDLCs) on RGD-synthetic polypeptide conjugate coatings. J Periodontal Res 2020; 55:713-723. [PMID: 32406091 DOI: 10.1111/jre.12760] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 03/30/2020] [Accepted: 04/17/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVE Periodontal ligament cells (PDLCs) are an important source for periodontal tissue healing and regeneration. Proper cell adhesion is a key for survival of anchorage-dependent cells and also initiates further intracellular signals for essential cellular functions. We aimed to test 3 different synthetic conjugates with integrin-binding RGD sequence (SAK-c[RGDfC], AK-c[RGDfC], and SAK-opn on the adhesion of human PDLCs and subsequent events including proliferation, migration, behavior of cell surface molecules, and osteogenic differentiation. MATERIALS AND METHODS Synthetic peptides were synthesized by solid-phase technique and attached to branched chain polymeric polypeptides via thioether linkage. Simple adsorption method was used to coat tissue culture plastic or electric arrays. PDLCs were isolated from 24 surgically extracted human third molars. Cell adhesion and proliferation were measured with real-time impedimetric xCELLigence SP system. Cell migration assay was performed with Ibidi® Culture inserts. Cell surface antigens were detected using flow cytometry analysis. Osteogenic differentiation was assessed with alkaline phosphatase (ALP) assay and Alizarin Red S staining, and real-time qPCR was performed to analyze the osteoblast-related gene expression. Osteogenic differentiation and adipogenic differentiation of PDLCs were monitored by real-time Electrical Cell-Substrate Impedance Spectroscopy (ECIS). RESULTS Primary outcome of this study relies on that all three synthetic RGD peptides improved PDLC adhesion (P < .05). When animal serum is absent in culture medium, SAK-c[RGDfC] and AK-c[RGDfC] elevated cell adhesion (P < .05). Cell migration was enhanced by SAK-c[RGDfC] and AK-c[RGDfC] (P < .05). After 1-week treatment, all synthetic peptides elevated CD105 (1.7- to 2.2-fold) and CD146 (1.3- to 1.5-fold) markers and caused different integrin patterns. ALP activity (1.4-fold) and ARS (1.8- and 2.0-fold) were increased by SAK-c[RGDfC] and AK-c[RGDfC] in absence of osteogenic supplements, and all the peptides supported the mineralization under osteogenic condition (P < .05). RT-qPCR revealed the upregulation of bone sialoprotein (5.0- to 7.8-fold), osteocalcin (2.3- to 2.7-fold), and ALP (1.9- to 2.3-fold) gene expression in osteogenesis-induced PDLCs. ECIS monitoring showed that higher impedance was generated by the osteogenic induction compared with the adipogenic or the non-induced (P < .05). CONCLUSIONS Our study demonstrates that SAK-c[RGDfC] and AK-c[RGDfC] improved adhesion and migration of PDLCs and supported osteogenic differentiation of PDLCs. These cyclic RGD peptides proved to be applicable biocompatible material in regenerative medicine.
Collapse
Affiliation(s)
- Zambaga Khorolsuren
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary.,Department of Conservative Dentistry, Semmelweis University, Budapest, Hungary
| | - Orsolya Lang
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Eva Pallinger
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Anna Foldes
- Department of Oral Biology, Semmelweis University, Budapest, Hungary
| | - Gyulai-Gaál Szabolcs
- Department of Oral Diagnostics, Faculty of Dentistry, Semmelweis University, Budapest, Hungary
| | - Gabor Varga
- Department of Oral Biology, Semmelweis University, Budapest, Hungary
| | - Gabor Mezo
- MTA-ELTE Research Group of Peptide Chemistry, Budapest, Hungary
| | - Janos Vag
- Department of Conservative Dentistry, Semmelweis University, Budapest, Hungary
| | - Laszlo Kohidai
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
43
|
Interdigitated aluminium and titanium sensors for assessing epithelial barrier functionality by electric cell-substrate impedance spectroscopy (ECIS). Biomed Microdevices 2020; 22:30. [PMID: 32328801 PMCID: PMC7181462 DOI: 10.1007/s10544-020-00486-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Electric cell-substrate impedance spectroscopy (ECIS) enables non-invasive and continuous read-out of electrical parameters of living tissue. The aim of the current study was to investigate the performance of interdigitated sensors with 50 μm electrode width and 50 μm inter-electrode distance made of gold, aluminium, and titanium for monitoring the barrier properties of epithelial cells in tissue culture. At first, the measurement performance of the photolithographic fabricated sensors was characterized by defined reference electrolytes. The sensors were used to monitor the electrical properties of two adherent epithelial barrier tissue models: renal proximal tubular LLC-PK1 cells, representing a normal functional transporting epithelium, and human cervical cancer-derived HeLa cells, forming non-transporting cancerous epithelial tissue. Then, the impedance spectra obtained were analysed by numerically fitting the parameters of the two different models to the measured impedance spectrum. Aluminium sensors proved to be as sensitive and consistent in repeated online-recordings for continuous cell growth and differentiation monitoring as sensors made of gold, the standard electrode material. Titanium electrodes exhibited an elevated intrinsic ohmic resistance in comparison to gold reflecting its lower electric conductivity. Analysis of impedance spectra through applying models and numerical data fitting enabled the detailed investigation of the development and properties of a functional transporting epithelial tissue using either gold or aluminium sensors. The result of the data obtained, supports the consideration of aluminium and titanium sensor materials as potential alternatives to gold sensors for advanced application of ECIS spectroscopy.
Collapse
|
44
|
Adams TNG, Jiang AYL, Mendoza NS, Ro CC, Lee DH, Lee AP, Flanagan LA. Label-free enrichment of fate-biased human neural stem and progenitor cells. Biosens Bioelectron 2020; 152:111982. [PMID: 32056730 PMCID: PMC8860404 DOI: 10.1016/j.bios.2019.111982] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/21/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Abstract
Human neural stem and progenitor cells (hNSPCs) have therapeutic potential to treat neural diseases and injuries since they provide neuroprotection and differentiate into astrocytes, neurons, and oligodendrocytes. However, cultures of hNSPCs are heterogeneous, containing cells linked to distinct differentiated cell fates. HNSPCs that differentiate into astrocytes are of interest for specific neurological diseases, creating a need for approaches that can detect and isolate these cells. Astrocyte-biased hNSPCs differ from other cell types in electrophysiological properties, namely membrane capacitance, and we hypothesized that this could be used to enrich these cells using dielectrophoresis (DEP). We implemented a two-step DEP sorting scheme, consisting of analysis to define the optimal sorting frequency followed by separation of cells at that frequency, to test whether astrocyte-biased cells could be separated from the other cell types present in hNSPC cultures. We developed a novel device that increased sorting reproducibility and provided both enriched and depleted cell populations in a single sort. Astrocyte-biased cells were successfully enriched from hNSPC cultures by DEP sorting, making this the first study to use electrophysiological properties for label-free enrichment of human astrocyte-biased cells. Enriched astrocyte-biased human cells enable future experiments to determine the specific properties of these important cells and test their therapeutic efficacy in animal models of neurological diseases.
Collapse
Affiliation(s)
- Tayloria N G Adams
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, 92697-2580, USA; Department of Neurology, University of California, Irvine, Irvine, CA, 92697-6750, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, CA, 92697-1705, USA.
| | - Alan Y L Jiang
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697-2627, USA; Department of Neurology, University of California, Irvine, Irvine, CA, 92697-6750, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, CA, 92697-1705, USA
| | - Nicolo S Mendoza
- Department of Neurology, University of California, Irvine, Irvine, CA, 92697-6750, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, CA, 92697-1705, USA
| | - Clarissa C Ro
- Department of Neurology, University of California, Irvine, Irvine, CA, 92697-6750, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, CA, 92697-1705, USA
| | - Do-Hyun Lee
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697-2627, USA
| | - Abraham P Lee
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697-2627, USA
| | - Lisa A Flanagan
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697-2627, USA; Department of Neurology, University of California, Irvine, Irvine, CA, 92697-6750, USA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, CA, 92697-1705, USA; Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA, 92697-4291, USA.
| |
Collapse
|
45
|
Mesenchymal MACF1 Facilitates SMAD7 Nuclear Translocation to Drive Bone Formation. Cells 2020; 9:cells9030616. [PMID: 32143362 PMCID: PMC7140458 DOI: 10.3390/cells9030616] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/27/2020] [Accepted: 02/29/2020] [Indexed: 02/07/2023] Open
Abstract
Microtubule actin crosslinking factor 1 (MACF1) is a large crosslinker that contributes to cell integrity and cell differentiation. Recent studies show that MACF1 is involved in multiple cellular functions such as neuron development and epidermal migration, and is the molecular basis for many degenerative diseases. MACF1 is highly abundant in bones, especially in mesenchymal stem cells; however, its regulatory role is still less understood in bone formation and degenerative bone diseases. In this study, we found MACF1 expression in mesenchymal stem cells (MSCs) of osteoporotic bone specimens was significantly lower. By conditional gene targeting to delete the mesenchymal Macf1 gene in mice, we observed in MSCs decreased osteogenic differentiation capability. During early stage bone development, the MACF1 conditional knockout (cKO) mice exhibit significant ossification retardation in skull and hindlimb, and by adulthood, mesenchymal loss of MACF1 attenuated bone mass, bone microarchitecture, and bone formation capability significantly. Further, we showed that MACF1 interacts directly with SMAD family member 7 (SMAD7) and facilitates SMAD7 nuclear translocation to initiate downstream osteogenic pathways. Hopefully these findings will expand the biological scope of the MACF1 gene, and provide an experimental basis for targeting MACF1 in degenerative bone diseases such as osteoporosis.
Collapse
|
46
|
Morgan K, Gamal W, Samuel K, Morley SD, Hayes PC, Bagnaninchi P, Plevris JN. Application of Impedance-Based Techniques in Hepatology Research. J Clin Med 2019; 9:jcm9010050. [PMID: 31878354 PMCID: PMC7019217 DOI: 10.3390/jcm9010050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/22/2022] Open
Abstract
There are a variety of end-point assays and techniques available to monitor hepatic cell cultures and study toxicity within in vitro models. These commonly focus on one aspect of cell metabolism and are often destructive to cells. Impedance-based cellular assays (IBCAs) assess biological functions of cell populations in real-time by measuring electrical impedance, which is the resistance to alternating current caused by the dielectric properties of proliferating of cells. While the uses of IBCA have been widely reported for a number of tissues, specific uses in the study of hepatic cell cultures have not been reported to date. IBCA monitors cellular behaviour throughout experimentation non-invasively without labelling or damage to cell cultures. The data extrapolated from IBCA can be correlated to biological events happening within the cell and therefore may inform drug toxicity studies or other applications within hepatic research. Because tight junctions comprise the blood/biliary barrier in hepatocytes, there are major consequences when these junctions are disrupted, as many pathologies centre around the bile canaliculi and flow of bile out of the liver. The application of IBCA in hepatology provides a unique opportunity to assess cellular polarity and patency of tight junctions, vital to maintaining normal hepatic function. Here, we describe how IBCAs have been applied to measuring the effect of viral infection, drug toxicity /IC50, cholangiopathies, cancer metastasis and monitoring of the gut-liver axis. We also highlight key areas of research where IBCAs could be used in future applications within the field of hepatology.
Collapse
Affiliation(s)
- Katie Morgan
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
- Correspondence:
| | - Wesam Gamal
- James Nasmyth Building, Institute of Mechanical, Process and Energy Engineering, Heriot-Watt University School of Engineering and Physical Sciences, Edinburgh EH14 4AS, UK;
| | - Kay Samuel
- The Jack Copland Centre, Advanced Therapeutics, Scottish National Blood Transfusion Service, 52 Research Avenue North, Edinburgh EH14 4BE, UK;
| | - Steven D. Morley
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| | - Peter C. Hayes
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| | - Pierre Bagnaninchi
- MRC Centre for Regenerative Medicine 5 Little France Drive, Edinburgh EH16 4UU, UK;
| | - John N. Plevris
- The University of Edinburgh Hepatology Laboratory, Division of Heath Sciences, University of Edinburgh Medical School, Chancellor’s Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh EH16 4SB, UK; (S.D.M.); (P.C.H.); (J.N.P.)
| |
Collapse
|
47
|
High-throughput label-free characterization of viable, necrotic and apoptotic human lymphoma cells in a coplanar-electrode microfluidic impedance chip. Biosens Bioelectron 2019; 150:111887. [PMID: 31780405 DOI: 10.1016/j.bios.2019.111887] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 01/08/2023]
Abstract
The study and the characterization of cell death mechanisms are fundamental in cell biology research. Traditional death/viability assays usually involve laborious sample preparation and expensive equipment or reagents. In this work, we use electrical impedance spectroscopy as a label-free methodology to characterize viable, necrotic and apoptotic human lymphoma U937 cells. A simple three-electrode coplanar layout is used in a differential measurement scheme and thousands of cells are measured at high-throughput (≈200 cell/s). Tailored signal processing enables accurate and robust cell characterization without the need for cell focusing systems. The results suggest that, at low frequency (0.5 MHz), signal magnitude enables the discrimination between viable/necrotic cells and cell fragments, whereas phase information allows discriminating between viable cells and necrotic cells. At higher frequency (10 MHz) two subpopulations of cell fragments are distinguished. This work substantiates the prominent role of electrical impedance spectroscopy for the development of next-generation cell viability assays.
Collapse
|
48
|
On-chip label-free determination of cell survival rate. Biosens Bioelectron 2019; 148:111820. [PMID: 31706174 DOI: 10.1016/j.bios.2019.111820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 10/01/2019] [Accepted: 10/23/2019] [Indexed: 12/25/2022]
Abstract
Cell survival rate (CSR) is a very important parameter in biological and medical fields. Today, the routine method to determine this parameter is time-consuming; it also makes the labeled cells no longer useable for subsequent experiments. Here, we developed an on-chip label-free method for determining the CSR. For the method, a hypertonic stimulus was designed to create volume differences between living and dead cells, and then, the differences were characterized with measurements of impedance as the cells flowed through two electrodes. Based on the method, a microfluidic hypertonic stimulus-based impedance flow cytometry chip (HSIFC) was designed, and the localized function of the HSIFC was verified. Finally, the performance of the HSIFC was confirmed by measuring the different CSRs for the different types of cells. The results show that the HSIFC can accurately determine the CSR, and the accuracy is comparable to that of flow cytometry. This work paves the way for the label-free evaluation of CSR after various cell manipulations and treatments on the chip and promotes the versatility of lab-on-a-chip devices.
Collapse
|
49
|
Large scale and integrated platform for digital mass culture of anchorage dependent cells. Nat Commun 2019; 10:4824. [PMID: 31645567 PMCID: PMC6811641 DOI: 10.1038/s41467-019-12777-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/26/2019] [Indexed: 01/17/2023] Open
Abstract
Industrial applications of anchorage-dependent cells require large-scale cell culture with multifunctional monitoring of culture conditions and control of cell behaviour. Here, we introduce a large-scale, integrated, and smart cell-culture platform (LISCCP) that facilitates digital mass culture of anchorage-dependent cells. LISCCP is devised through large-scale integration of ultrathin sensors and stimulator arrays in multiple layers. LISCCP provides real-time, 3D, and multimodal monitoring and localized control of the cultured cells, which thereby allows minimizing operation labour and maximizing cell culture performance. Wireless integration of multiple LISCCPs across multiple incubators further amplifies the culture scale and enables digital monitoring and local control of numerous culture layers, making the large-scale culture more efficient. Thus, LISCCP can transform conventional labour-intensive and high-cost cell cultures into efficient digital mass cell cultures. This platform could be useful for industrial applications of cell cultures such as in vitro toxicity testing of drugs and cosmetics and clinical scale production of cells for cell therapy. Large scale culture of adherent cells would benefit from a platform for continuous monitoring and control of cell growth and culture conditions. Here the authors develop an integrated, smart cell culture platform where cells are grown on multiple layers of thin sensors that can be wirelessly integrated across several incubators.
Collapse
|
50
|
Hedayatipour A, Aslanzadeh S, McFarlane N. CMOS based whole cell impedance sensing: Challenges and future outlook. Biosens Bioelectron 2019; 143:111600. [PMID: 31479988 DOI: 10.1016/j.bios.2019.111600] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/05/2019] [Accepted: 08/13/2019] [Indexed: 01/14/2023]
Abstract
With the increasing need for multi-analyte point-of-care diagnosis devices, cell impedance measurement is a promising technique for integration with other sensing modalities. In this comprehensive review, the theory underlying cell impedance sensing, including the history, complementary metal-oxide-semiconductor (CMOS) based implementations, and applications are critically assessed. Whole cell impedance sensing, also known as electric cell-substrate impedance sensing (ECIS) or electrical impedance spectroscopy (EIS), is an approach for studying and diagnosing living cells in in-vitro and in-vivo environments. The technique is popular since it is label-free, non-invasive, and low cost when compared to standard biochemical assays. CMOS cell impedance measurement systems have been focused on expanding their applications to numerous aspects of biological, environmental, and food safety applications. This paper presents and evaluates circuit topologies for whole cell impedance measurement. The presented review compares several existing CMOS designs, including the classification, measurement speed, and sensitivity of varying topologies.
Collapse
Affiliation(s)
- Ava Hedayatipour
- Department of Electrical Engineering and Computer Science, University of Tennessee, Knoxville, TN, USA.
| | - Shaghayegh Aslanzadeh
- Department of Electrical Engineering and Computer Science, University of Tennessee, Knoxville, TN, USA
| | - Nicole McFarlane
- Department of Electrical Engineering and Computer Science, University of Tennessee, Knoxville, TN, USA
| |
Collapse
|