1
|
Lee US, Sim DB, Lee JH, Kim BH. Fabrication of Micro Carbon Mold for Glass-Based Micro Hole Array. MICROMACHINES 2024; 15:194. [PMID: 38398923 PMCID: PMC10890415 DOI: 10.3390/mi15020194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024]
Abstract
In glass molding to produce biochips with micro holes, cavities, and channels, it is important to machine micro molds. This study presents a novel process for fabricating micro pin arrays on carbon graphite, one of the glass molding materials. The micro pin array was used as a mold to fabricate a glass-based micro hole array. Using conventional micro endmill tools, machining micro-cylindrical pins requires complex toolpaths and is time-consuming. In order to machine micro pin arrays with high efficiency, a micro eccentric tool was introduced. Micro pin arrays with a diameter of 200 µm and a height of 200 µm were easily fabricated on graphite using the micro eccentric tool. In the machining of micro pin arrays using eccentric tools, the machining characteristics such as cutting force and tool wear were investigated.
Collapse
Affiliation(s)
- Ui Seok Lee
- Department of Mechanical Engineering, Graduate School, Soongsil University, 369 Sangdo-ro, Dongjak-gu, Seoul 06978, Republic of Korea; (U.S.L.); (D.B.S.); (J.H.L.)
| | - Dae Bo Sim
- Department of Mechanical Engineering, Graduate School, Soongsil University, 369 Sangdo-ro, Dongjak-gu, Seoul 06978, Republic of Korea; (U.S.L.); (D.B.S.); (J.H.L.)
| | - Ji Hyo Lee
- Department of Mechanical Engineering, Graduate School, Soongsil University, 369 Sangdo-ro, Dongjak-gu, Seoul 06978, Republic of Korea; (U.S.L.); (D.B.S.); (J.H.L.)
| | - Bo Hyun Kim
- School of Mechanical Engineering, Soongsil University, 369 Sangdo-ro, Dongjak-gu, Seoul 06978, Republic of Korea
| |
Collapse
|
2
|
Alpsoy L, Sedeky AS, Rehbein U, Thedieck K, Brandstetter T, Rühe J. Particle ID: A Multiplexed Hydrogel Bead Platform for Biomedical Applications. ACS APPLIED MATERIALS & INTERFACES 2023; 15:55346-55357. [PMID: 37982803 PMCID: PMC10712431 DOI: 10.1021/acsami.3c12122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/11/2023] [Accepted: 10/18/2023] [Indexed: 11/21/2023]
Abstract
We present a new platform based on hydrogel beads for multiplex analysis that can be fabricated, barcoded, and functionalized in a single step using a simple microfluidic assembly and a photo-cross-linking process. The beads are generated in a two-phase flow fluidic system and photo-cross-linking of the polymer in the aqueous phase by C,H insertion cross-linking (CHic). The size and shape of the hydrogel particles can be controlled over a wide range by fluidic parameters. During the fabrication of the beads, they are barcoded by using physical and optical barcoding strategies. Magnetic beads and fluorescent particles, which allow identification of the production batch number, are added simultaneously as desired, resulting in complex, multifunctional beads in a one-step reaction. As an example of biofunctionalization, Borrelia antigens were immobilized on the beads. Serum samples that originated from infected and non-infected patients could be clearly distinguished, and the sensitivity was as good as or even better than ELISA, the state of the art in clinical diagnostics. The ease of the one-step production process and the wide range of barcoding parameters offer strong advantages for multiplexed analytics in the life sciences and medical diagnostics.
Collapse
Affiliation(s)
- Lokman Alpsoy
- Department of Microsystems Engineering (IMTEK), Chemistry & Physics of Interfaces, University of Freiburg, Freiburg im Breisgau 79110, Germany
- livMatS@FIT (Freiburg Center for Interactive Materials and Bioinspired Technologies), University of Freiburg, Freiburg 79110, Germany
| | - Abanoub Selim Sedeky
- Department of Microsystems Engineering (IMTEK), Chemistry & Physics of Interfaces, University of Freiburg, Freiburg im Breisgau 79110, Germany
| | - Ulrike Rehbein
- Institute of Biochemistry, Center of Chemistry and Biomedicine, University of Innsbruck, 6020 Innsbruck, Austria
| | - Kathrin Thedieck
- Institute of Biochemistry, Center of Chemistry and Biomedicine, University of Innsbruck, 6020 Innsbruck, Austria
- Freiburg Materials Research Center FMF, Albert-Ludwigs-University of Freiburg, 79104 Freiburg, Germany
- Department of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| | - Thomas Brandstetter
- Department of Microsystems Engineering (IMTEK), Chemistry & Physics of Interfaces, University of Freiburg, Freiburg im Breisgau 79110, Germany
| | - Jürgen Rühe
- Department of Microsystems Engineering (IMTEK), Chemistry & Physics of Interfaces, University of Freiburg, Freiburg im Breisgau 79110, Germany
- livMatS@FIT (Freiburg Center for Interactive Materials and Bioinspired Technologies), University of Freiburg, Freiburg 79110, Germany
| |
Collapse
|
3
|
Yi K, Wang X, Filippov SK, Zhang H. Emerging ctDNA detection strategies in clinical cancer theranostics. SMART MEDICINE 2023; 2:e20230031. [PMID: 39188296 PMCID: PMC11235813 DOI: 10.1002/smmd.20230031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/13/2023] [Indexed: 08/28/2024]
Abstract
Circulating tumor DNA (ctDNA) is naked DNA molecules shed from the tumor cells into the peripheral blood circulation. They contain tumor-specific gene mutations and other valuable information. ctDNA is considered to be one of the most significant analytes in liquid biopsies. Over the past decades, numerous researchers have developed various detection strategies to perform quantitative or qualitative ctDNA analysis, including PCR-based detection and sequencing-based detection. More and more studies have illustrated the great value of ctDNA as a biomarker in the diagnosis, prognosis and heterogeneity of tumor. In this review, we first outlined the development of digital PCR (dPCR)-based and next generation sequencing (NGS)-based ctDNA detection systems. Besides, we presented the introduction of the emerging ctDNA analysis strategies based on various biosensors, such as electrochemical biosensors, fluorescent biosensors, surface plasmon resonance and Raman spectroscopy, as well as their applications in the field of biomedicine. Finally, we summarized the essentials of the preceding discussions, and the existing challenges and prospects for the future are also involved.
Collapse
Affiliation(s)
- Kexin Yi
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
| | - Xiaoju Wang
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
| | - Sergey K. Filippov
- DWI‐Leibniz Institute for Interactive Materials e. V.AachenGermany
- School of PharmacyUniversity of ReadingReadingUK
| | - Hongbo Zhang
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| |
Collapse
|
4
|
Van den Eeckhoudt R, Christiaens AS, Ceyssens F, Vangalis V, Verstrepen KJ, Boon N, Tavernier F, Kraft M, Taurino I. Full-electric microfluidic platform to capture, analyze and selectively release single cells. LAB ON A CHIP 2023; 23:4276-4286. [PMID: 37668159 DOI: 10.1039/d3lc00645j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Current single-cell technologies require large and expensive equipment, limiting their use to specialized labs. In this paper, we present for the first time a microfluidic device which demonstrates a combined method for full-electric cell capturing, analyzing, and selectively releasing with single-cell resolution. All functionalities are experimentally demonstrated on Saccharomyces cerevisiae. Our microfluidic platform consists of traps centered around a pair of individually accessible coplanar electrodes, positioned under a microfluidic channel. Using this device, we validate our novel Two-Voltage method for trapping single cells by positive dielectrophoresis (pDEP). Cells are attracted to the trap when a high voltage (VH) is applied. A low voltage (VL) holds the already trapped cell in place without attracting additional cells, allowing full control over the number of trapped cells. After trapping, the cells are analyzed by broadband electrochemical impedance spectroscopy. These measurements allow the detection of single cells and the extraction of cell parameters. Additionally, these measurements show a strong correlation between average phase change and cell size, enabling the use of our system for size measurements in biological applications. Finally, our device allows selectively releasing trapped cells by turning off the pDEP signal in their trap. The experimental results show the techniques potential as a full-electric single-cell analysis tool with potential for miniaturization and automation which opens new avenues towards small-scale, high throughput single-cell analysis and sorting lab-on-CMOS devices.
Collapse
Affiliation(s)
- Ruben Van den Eeckhoudt
- Micro- and Nanosystems (MNS), Department of Electrical Engineering (ESAT), KU Leuven, Leuven, Belgium.
| | - An-Sofie Christiaens
- Chemical and Biochemical Reactor Engineering and Safety (CREaS), Department of Chemical Engineering, KU Leuven, Leuven, Belgium
| | - Frederik Ceyssens
- Micro- and Nanosystems (MNS), Department of Electrical Engineering (ESAT), KU Leuven, Leuven, Belgium.
- Leuven Institute for Micro- and Nanoscale Integration (LIMNI), KU Leuven, Leuven, Belgium
| | - Vasileios Vangalis
- VIB - KU Leuven Center for Microbiology, Leuven, Belgium
- CMPG Laboratory for Genetics and Genomics, KU Leuven, Leuven, Belgium
| | - Kevin J Verstrepen
- VIB - KU Leuven Center for Microbiology, Leuven, Belgium
- CMPG Laboratory for Genetics and Genomics, KU Leuven, Leuven, Belgium
| | - Nico Boon
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Filip Tavernier
- MICAS, Department of Electrical Engineering (ESAT), KU Leuven, Leuven, Belgium
| | - Michael Kraft
- Micro- and Nanosystems (MNS), Department of Electrical Engineering (ESAT), KU Leuven, Leuven, Belgium.
- Leuven Institute for Micro- and Nanoscale Integration (LIMNI), KU Leuven, Leuven, Belgium
| | - Irene Taurino
- Micro- and Nanosystems (MNS), Department of Electrical Engineering (ESAT), KU Leuven, Leuven, Belgium.
- Semiconductor Physics, Department of Physics and Astronomy, KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Sandström N, Brandt L, Sandoz PA, Zambarda C, Guldevall K, Schulz-Ruhtenberg M, Rösener B, Krüger RA, Önfelt B. Live single cell imaging assays in glass microwells produced by laser-induced deep etching. LAB ON A CHIP 2022; 22:2107-2121. [PMID: 35470832 DOI: 10.1039/d2lc00090c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Miniaturization of cell culture substrates enables controlled analysis of living cells in confined micro-scale environments. This is particularly suitable for imaging individual cells over time, as they can be monitored without escaping the imaging field-of-view (FoV). Glass materials are ideal for most microscopy applications. However, with current methods used in life sciences, glass microfabrication is limited in terms of either freedom of design, quality, or throughput. In this work, we introduce laser-induced deep etching (LIDE) as a method for producing glass microwell arrays for live single cell imaging assays. We demonstrate novel microwell arrays with deep, high-aspect ratio wells that have rounded, dimpled or flat bottom profiles in either single-layer or double-layer glass chips. The microwells are evaluated for microscopy-based analysis of long-term cell culture, clonal expansion, laterally organized cell seeding, subcellular mechanics during migration and immune cell cytotoxicity assays of both adherent and suspension cells. It is shown that all types of microwells can support viable cell cultures and imaging with single cell resolution, and we highlight specific benefits of each microwell design for different applications. We believe that high-quality glass microwell arrays enabled by LIDE provide a great option for high-content and high-resolution imaging-based live cell assays with a broad range of potential applications within life sciences.
Collapse
Affiliation(s)
- Niklas Sandström
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Ludwig Brandt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Patrick A Sandoz
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Chiara Zambarda
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Karolin Guldevall
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| | | | | | | | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
6
|
Abstract
Microfluidics has enabled a new era of cellular and molecular assays due to the small length scales, parallelization, and the modularity of various analysis and actuation functions. Droplet microfluidics, in particular, has been instrumental in providing new tools for biology with its ability to quickly and reproducibly generate drops that act as individual reactors. A notable beneficiary of this technology has been single-cell RNA sequencing, which has revealed new heterogeneities and interactions for the fundamental unit of life. However, viruses far surpass the diversity of cellular life, affect the dynamics of all ecosystems, and are a chronic source of global health crises. Despite their impact on the world, high-throughput and high-resolution viral profiling has been difficult, with conventional methods being limited to population-level averaging, large sample volumes, and few cultivable hosts. Consequently, most viruses have not been identified and studied. Droplet microfluidics holds the potential to address many of these limitations and offers new levels of sensitivity and throughput for virology. This Feature highlights recent efforts that have applied droplet microfluidics to the detection and study of viruses, including for diagnostics, virus-host interactions, and cell-independent virus assays. In combination with traditional virology methods, droplet microfluidics should prove a potent tool toward achieving a better understanding of the most abundant biological species on Earth.
Collapse
Affiliation(s)
- Wenyang Jing
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hee-Sun Han
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States.,Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, Illinois 61801, United States
| |
Collapse
|
7
|
Struyfs C, Breukers J, Spasic D, Lammertyn J, Cammue BPA, Thevissen K. Multiplex Analysis to Unravel the Mode of Antifungal Activity of the Plant Defensin HsAFP1 in Single Yeast Cells. Int J Mol Sci 2022; 23:ijms23031515. [PMID: 35163438 PMCID: PMC8836000 DOI: 10.3390/ijms23031515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023] Open
Abstract
Single cell analyses have gained increasing interest over bulk approaches because of considerable cell-to-cell variability within isogenic populations. Herein, flow cytometry remains golden standard due to its high-throughput efficiency and versatility, although it does not allow to investigate the interdependency of cellular events over time. Starting from our microfluidic platform that enables to trap and retain individual cells on a fixed location over time, here, we focused on unraveling kinetic responses of single Saccharomyces cerevisiae yeast cells upon treatment with the antifungal plant defensin HsAFP1. We monitored the time between production of reactive oxygen species (ROS) and membrane permeabilization (MP) in single yeast cells for different HsAFP1 doses using two fluorescent dyes with non-overlapping spectra. Within a time frame of 2 min, only <0.3% cells displayed time between the induction of ROS and MP. Reducing the time frame to 30 s did not result in increased numbers of cells with time between these events, pointing to ROS and MP induction as highly dynamic and correlated processes. In conclusion, using an in-house developed continuous microfluidic platform, we investigated the mode of action of HsAFP1 at single cell level, thereby uncovering the close interdependency between ROS induction and MP in yeast.
Collapse
Affiliation(s)
- Caroline Struyfs
- Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (MS), KU Leuven, Kasteelpark Arenberg 20, 3001 Leuven, Belgium; (C.S.); (B.P.A.C.)
| | - Jolien Breukers
- Biosensors Group, Department of Biosystems (BIOSYST), KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium; (J.B.); (D.S.); (J.L.)
| | - Dragana Spasic
- Biosensors Group, Department of Biosystems (BIOSYST), KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium; (J.B.); (D.S.); (J.L.)
| | - Jeroen Lammertyn
- Biosensors Group, Department of Biosystems (BIOSYST), KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium; (J.B.); (D.S.); (J.L.)
| | - Bruno P. A. Cammue
- Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (MS), KU Leuven, Kasteelpark Arenberg 20, 3001 Leuven, Belgium; (C.S.); (B.P.A.C.)
| | - Karin Thevissen
- Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (MS), KU Leuven, Kasteelpark Arenberg 20, 3001 Leuven, Belgium; (C.S.); (B.P.A.C.)
- Correspondence: ; Tel.: +32-16-32-96-88
| |
Collapse
|
8
|
Van Lent J, Breukers J, Ven K, Ampofo L, Horta S, Pollet F, Imbrechts M, Geukens N, Vanhoorelbeke K, Declerck P, Lammertyn J. Miniaturized single-cell technologies for monoclonal antibody discovery. LAB ON A CHIP 2021; 21:3627-3654. [PMID: 34505611 DOI: 10.1039/d1lc00243k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Antibodies (Abs) are among the most important class of biologicals, showcasing a high therapeutic and diagnostic value. In the global therapeutic Ab market, fully-human monoclonal Abs (FH-mAbs) are flourishing thanks to their low immunogenicity and high specificity. The rapidly emerging field of single-cell technologies has paved the way to efficiently discover mAbs by facilitating a fast screening of the antigen (Ag)-specificity and functionality of Abs expressed by B cells. This review summarizes the principles and challenges of the four key concepts to discover mAbs using these technologies, being confinement of single cells using either droplet microfluidics or microstructure arrays, identification of the cells of interest, retrieval of those cells and single-cell sequence determination required for mAb production. This review reveals the enormous potential for mix-and-matching of the above-mentioned strategies, which is illustrated by the plethora of established, highly integrated devices. Lastly, an outlook is given on the many opportunities and challenges that still lie ahead to fully exploit miniaturized single-cell technologies for mAb discovery.
Collapse
Affiliation(s)
- Julie Van Lent
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
| | - Jolien Breukers
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
| | - Karen Ven
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
| | - Louanne Ampofo
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, Leuven 3000, Belgium
| | - Sara Horta
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk 8500, Belgium
| | - Francesca Pollet
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
| | - Maya Imbrechts
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, Leuven 3000, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Leuven 3000, Belgium
| | - Nick Geukens
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Leuven 3000, Belgium
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk 8500, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Leuven 3000, Belgium
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven, Leuven 3000, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Leuven 3000, Belgium
| | - Jeroen Lammertyn
- Department of Biosystems, Biosensors Group, KU Leuven, Leuven 3001, Belgium.
| |
Collapse
|
9
|
Grubb ML, Caliari SR. Fabrication approaches for high-throughput and biomimetic disease modeling. Acta Biomater 2021; 132:52-82. [PMID: 33716174 PMCID: PMC8433272 DOI: 10.1016/j.actbio.2021.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/15/2021] [Accepted: 03/02/2021] [Indexed: 12/24/2022]
Abstract
There is often a tradeoff between in vitro disease modeling platforms that capture pathophysiologic complexity and those that are amenable to high-throughput fabrication and analysis. However, this divide is closing through the application of a handful of fabrication approaches-parallel fabrication, automation, and flow-driven assembly-to design sophisticated cellular and biomaterial systems. The purpose of this review is to highlight methods for the fabrication of high-throughput biomaterial-based platforms and showcase examples that demonstrate their utility over a range of throughput and complexity. We conclude with a discussion of future considerations for the continued development of higher-throughput in vitro platforms that capture the appropriate level of biological complexity for the desired application. STATEMENT OF SIGNIFICANCE: There is a pressing need for new biomedical tools to study and understand disease. These platforms should mimic the complex properties of the body while also permitting investigation of many combinations of cells, extracellular cues, and/or therapeutics in high-throughput. This review summarizes emerging strategies to fabricate biomimetic disease models that bridge the gap between complex tissue-mimicking microenvironments and high-throughput screens for personalized medicine.
Collapse
Affiliation(s)
- Mackenzie L Grubb
- Department of Biomedical Engineering, University of Virginia, Unites States
| | - Steven R Caliari
- Department of Biomedical Engineering, University of Virginia, Unites States; Department of Chemical Engineering, University of Virginia, Unites States.
| |
Collapse
|
10
|
Monitoring Contractile Cardiomyocytes via Impedance Using Multipurpose Thin Film Ruthenium Oxide Electrodes. SENSORS 2021; 21:s21041433. [PMID: 33670743 PMCID: PMC7923073 DOI: 10.3390/s21041433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/11/2021] [Accepted: 02/16/2021] [Indexed: 11/16/2022]
Abstract
A ruthenium oxide (RuOx) electrode was used to monitor contractile events of human pluripotent stem cells-derived cardiomyocytes (hPSC-CMs) through electrical impedance spectroscopy (EIS). Using RuOx electrodes presents an advantage over standard thin film Pt electrodes because the RuOx electrodes can also be used as electrochemical sensor for pH, O2, and nitric oxide, providing multisensory functionality with the same electrode. First, the EIS signal was validated in an optically transparent well-plate setup using Pt wire electrodes. This way, visual data could be recorded simultaneously. Frequency analyses of both EIS and the visual data revealed almost identical frequency components. This suggests both the EIS and visual data captured the similar events of the beating of (an area of) hPSC-CMs. Similar EIS measurement was then performed using the RuOx electrode, which yielded comparable signal and periodicity. This mode of operation adds to the versatility of the RuOx electrode's use in in vitro studies.
Collapse
|
11
|
Breukers J, Horta S, Struyfs C, Spasic D, Feys HB, Geukens N, Thevissen K, Cammue BPA, Vanhoorelbeke K, Lammertyn J. Tuning the Surface Interactions between Single Cells and an OSTE+ Microwell Array for Enhanced Single Cell Manipulation. ACS APPLIED MATERIALS & INTERFACES 2021; 13:2316-2326. [PMID: 33411502 DOI: 10.1021/acsami.0c19657] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Retrieving single cells of interest from an array of microwells for further off-chip analysis is crucial in numerous biological applications. To this end, several single cell manipulation strategies have been developed, including optical tweezers (OT). OT represent a unique approach for contactless cell retrieval, but their performance is often suboptimal due to nonspecific cell adhesion to the microwell surface. In this study, we focused on improving the surface chemistry of microwell arrays to ensure efficient single cell manipulation using OT. For this purpose, the surface of an off-stoichiometry thiol-ene-epoxy (OSTE+) microwell array was grafted with polyethylene glycol (PEG) molecules with different molecular weights: PEG 360, PEG 500, PEG 2000, and a PEG Mix (an equimolar ratio of PEG 500 and PEG 2000). Contact angle measurements showed that the PEG grafting process resulted in an increased surface energy, which was stable for at least 16 weeks. Next, cell adhesion of two cell types, baker's yeast (Saccharomyces cerevisiae) and human B cells, to surfaces treated with different PEGs was evaluated by registering the presence of cellular motion inside microwells and the efficiency of optical lifting of cells that display motion. Optimal results were obtained for surfaces grafted with PEG 2000 and PEG Mix, reaching an average fraction of cells with motion of over 93% and an average lifting efficiency of over 96% for both cell types. Upon the integration of this microwell array with a polydimethylsiloxane (PDMS) microfluidic channel, PEG Mix resulted in proper washing of non-seeded cells. We further demonstrated the wide applicability of the platform by manipulating non-responding yeast cells to antifungal treatment and B cells expressing surface IgG antibodies. The combination of the optimized microwell surface with continuous microfluidics results in a powerful and versatile platform, allowing high-throughput single cell studies and retrieval of target cells for off-chip analysis.
Collapse
Affiliation(s)
- Jolien Breukers
- Department of Biosystems, Biosensors Group, KU Leuven, Willem De Croylaan 42, Leuven 3001, Belgium
| | - Sara Horta
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Etienne Sabbelaan 53, Kortrijk 8500, Belgium
| | - Caroline Struyfs
- Centre of Microbial and Plant Genetics, KU Leuven, Kasteelpark Arenberg 20, Leuven 3001, Belgium
| | - Dragana Spasic
- Department of Biosystems, Biosensors Group, KU Leuven, Willem De Croylaan 42, Leuven 3001, Belgium
| | - Hendrik B Feys
- Transfusion Research Center, Belgian Red Cross-Flanders Ottergemsesteenweg 413, Gent 9000, Belgium
- Diagnostic Sciences, Ghent University, C. Heymanslaan 10, Gent 9000, Belgium
| | - Nick Geukens
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Herestraat 49, Leuven 3000, Belgium
| | - Karin Thevissen
- Centre of Microbial and Plant Genetics, KU Leuven, Kasteelpark Arenberg 20, Leuven 3001, Belgium
| | - Bruno P A Cammue
- Centre of Microbial and Plant Genetics, KU Leuven, Kasteelpark Arenberg 20, Leuven 3001, Belgium
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Etienne Sabbelaan 53, Kortrijk 8500, Belgium
| | - Jeroen Lammertyn
- Department of Biosystems, Biosensors Group, KU Leuven, Willem De Croylaan 42, Leuven 3001, Belgium
| |
Collapse
|
12
|
Ye S, Feng S, Huang L, Bian S. Recent Progress in Wearable Biosensors: From Healthcare Monitoring to Sports Analytics. BIOSENSORS 2020; 10:E205. [PMID: 33333888 PMCID: PMC7765261 DOI: 10.3390/bios10120205] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 02/07/2023]
Abstract
Recent advances in lab-on-a-chip technology establish solid foundations for wearable biosensors. These newly emerging wearable biosensors are capable of non-invasive, continuous monitoring by miniaturization of electronics and integration with microfluidics. The advent of flexible electronics, biochemical sensors, soft microfluidics, and pain-free microneedles have created new generations of wearable biosensors that explore brand-new avenues to interface with the human epidermis for monitoring physiological status. However, these devices are relatively underexplored for sports monitoring and analytics, which may be largely facilitated by the recent emergence of wearable biosensors characterized by real-time, non-invasive, and non-irritating sensing capacities. Here, we present a systematic review of wearable biosensing technologies with a focus on materials and fabrication strategies, sampling modalities, sensing modalities, as well as key analytes and wearable biosensing platforms for healthcare and sports monitoring with an emphasis on sweat and interstitial fluid biosensing. This review concludes with a summary of unresolved challenges and opportunities for future researchers interested in these technologies. With an in-depth understanding of the state-of-the-art wearable biosensing technologies, wearable biosensors for sports analytics would have a significant impact on the rapidly growing field-microfluidics for biosensing.
Collapse
Affiliation(s)
- Shun Ye
- Microfluidics Research & Innovation Laboratory, School of Sport Science, Beijing Sport University, Beijing 100084, China;
- Biomedical Engineering Department, College of Engineering, Pennsylvania State University, University Park, PA 16802, USA
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shilun Feng
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China;
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Liang Huang
- School of Instrument Science and Opto–Electronics Engineering, Hefei University of Technology, Hefei 230009, China;
| | - Shengtai Bian
- Microfluidics Research & Innovation Laboratory, School of Sport Science, Beijing Sport University, Beijing 100084, China;
| |
Collapse
|
13
|
Manzoor AA, Romita L, Hwang DK. A review on microwell and microfluidic geometric array fabrication techniques and its potential applications in cellular studies. CAN J CHEM ENG 2020. [DOI: 10.1002/cjce.23875] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Ahmad Ali Manzoor
- Department of Chemical Engineering Ryerson University Toronto Ontario Canada
- Keenan Research Centre for Biomedical Science St. Michael's Hospital Toronto Ontario Canada
- Institute for Biomedical Engineering Science and Technology (iBEST) A partnership between Ryerson University and St. Michael's Hospital Toronto Ontario Canada
| | - Lauren Romita
- Department of Chemical Engineering Ryerson University Toronto Ontario Canada
- Keenan Research Centre for Biomedical Science St. Michael's Hospital Toronto Ontario Canada
- Institute for Biomedical Engineering Science and Technology (iBEST) A partnership between Ryerson University and St. Michael's Hospital Toronto Ontario Canada
| | - Dae Kun Hwang
- Department of Chemical Engineering Ryerson University Toronto Ontario Canada
- Keenan Research Centre for Biomedical Science St. Michael's Hospital Toronto Ontario Canada
- Institute for Biomedical Engineering Science and Technology (iBEST) A partnership between Ryerson University and St. Michael's Hospital Toronto Ontario Canada
| |
Collapse
|
14
|
Romita L, Thompson S, Hwang DK. Rapid fabrication of sieved microwells and cross-flow microparticle trapping. Sci Rep 2020; 10:15687. [PMID: 32973304 PMCID: PMC7518267 DOI: 10.1038/s41598-020-72700-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/31/2020] [Indexed: 11/09/2022] Open
Abstract
The use of microwells is popular for a wide range of applications due to its' simplicity. However, the seeding of conventional microwells, which are closed at the bottom, is restricted to gravitational sedimentation for cell or particle deposition and therefore require lengthy settling times to maximize well occupancy. The addition of microfluidics to the capture process has accelerated cell or particle dispersion and improved capture ability but is mostly limited to gravitationally-driven settling for capture into the wells. An alternative approach to conventional closed-microwells, sieved microwells supersedes reliance on gravity by using hydrodynamic forces through the open pores at the bottom of the microwells to draw targets into the wells. We have developed a rapid fabrication method, based on flow lithography techniques, which allows us to easily customize the mesh pore sizes in a simple two-step process. Finally, by combining this microwell design with cross-flow trapping in a microfluidic two-layered channel, we achieve an 88 ± 6% well occupancy in under 10 s.
Collapse
Affiliation(s)
- Lauren Romita
- Department of Chemical Engineering, Ryerson University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada
| | - Shyan Thompson
- Department of Chemical Engineering, Ryerson University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada
| | - Dae Kun Hwang
- Department of Chemical Engineering, Ryerson University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada.
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada.
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada.
| |
Collapse
|
15
|
Di Girolamo S, Puorger C, Lipps G. Stable and selective permeable hydrogel microcapsules for high-throughput cell cultivation and enzymatic analysis. Microb Cell Fact 2020; 19:170. [PMID: 32854709 PMCID: PMC7451113 DOI: 10.1186/s12934-020-01427-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Miniaturization of biochemical reaction volumes within artificial microcompartments has been the key driver for directed evolution of several catalysts in the past two decades. Typically, single cells are co-compartmentalized within water-in-oil emulsion droplets with a fluorogenic substrate whose conversion allows identification of catalysts with improved performance. However, emulsion droplet-based technologies prevent cell proliferation to high density and preclude the feasibility of biochemical reactions that require the exchange of small molecule substrates. Here, we report on the development of a high-throughput screening method that addresses these shortcomings and that relies on a novel selective permeable polymer hydrogel microcapsule. RESULTS Hollow-core polyelectrolyte-coated chitosan alginate microcapsules (HC-PCAMs) with selective permeability were successfully constructed by jet break-up and layer-by-layer (LBL) technology. We showed that HC-PCAMs serve as miniaturized vessels for single cell encapsulation, enabling cell growth to high density and cell lysis to generate monoclonal cell lysate compartments suitable for high-throughput analysis using a large particle sorter (COPAS). The feasibility of using HC-PCAMs as reaction compartments which exchange small molecule substrates was demonstrated using the transpeptidation reaction catalyzed by the bond-forming enzyme sortase F from P. acnes. The polyelectrolyte shell surrounding microcapsules allowed a fluorescently labelled peptide substrate to enter the microcapsule and take part in the transpeptidation reaction catalyzed by the intracellularly expressed sortase enzyme retained within the capsule upon cell lysis. The specific retention of fluorescent transpeptidation products inside microcapsules enabled the sortase activity to be linked with a fluorescent readout and allowed clear separation of microcapsules expressing the wild type SrtF from those expressing the inactive variant. CONCLUSION A novel polymer hydrogel microcapsule-based method, which allows for high-throughput analysis based on encapsulation of single cells has been developed. The method has been validated for the transpeptidation activity of sortase enzymes and represents a powerful tool for screening of libraries of sortases, other bond-forming enzymes, as well as of binding affinities in directed evolution experiments. Moreover, selective permeable microcapsules encapsulating microcolonies provide a new and efficient means for preparing novel caged biocatalyst and biosensor agents.
Collapse
Affiliation(s)
- Salvatore Di Girolamo
- University of Applied Sciences and Arts Northwestern Switzerland, Institute for Chemistry and Bioanalytics, Hofackerstrasse 30, 4132, Muttenz, Switzerland
| | - Chasper Puorger
- University of Applied Sciences and Arts Northwestern Switzerland, Institute for Chemistry and Bioanalytics, Hofackerstrasse 30, 4132, Muttenz, Switzerland
| | - Georg Lipps
- University of Applied Sciences and Arts Northwestern Switzerland, Institute for Chemistry and Bioanalytics, Hofackerstrasse 30, 4132, Muttenz, Switzerland.
| |
Collapse
|
16
|
Liu W, He H, Zheng SY. Microfluidics in Single-Cell Virology: Technologies and Applications. Trends Biotechnol 2020; 38:1360-1372. [PMID: 32430227 DOI: 10.1016/j.tibtech.2020.04.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/17/2022]
Abstract
Microfluidics has proven to be a powerful tool for probing biology at the single-cell level. However, it is only in the past 5 years that single-cell microfluidics has been used in the field of virology. An array of strategies based on microwells, microvalves, and droplets is now available for tracking viral infection dynamics, identifying cell subpopulations with particular phenotypes, as well as high-throughput screening. The insights into the virus-host interactions gained at the single-cell level are unprecedented and usually inaccessible by population-based experiments. Therefore, single-cell microfluidics, which opens new avenues for mechanism elucidation and development of antiviral therapeutics, would be a valuable tool for the study of viral pathogenesis.
Collapse
Affiliation(s)
- Wu Liu
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| | - Hongzhang He
- Captis Diagnostics Inc., Pittsburgh, PA 15213, USA
| | - Si-Yang Zheng
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA; Department of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| |
Collapse
|
17
|
Sun G, Teng Y, Zhao Z, Cheow LF, Yu H, Chen CH. Functional Stem Cell Sorting via Integrative Droplet Synchronization. Anal Chem 2020; 92:7915-7923. [DOI: 10.1021/acs.analchem.0c01312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Guoyun Sun
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, 04-08, Singapore
| | - Yao Teng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, MD9, Singapore
| | - Zixuan Zhao
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, 04-08 Singapore
| | - Lih Feng Cheow
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, 04-08, Singapore
| | - Hanry Yu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, MD9, Singapore
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, 04-08 Singapore
- Institute of Bioengineering and Nanotechnology, A*STAR, 31 Biopolis Way, The Nanos 07-01, Singapore
- CAMP, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, 04-01, Singapore
| | - Chia-Hung Chen
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong SAR China
| |
Collapse
|
18
|
Gao X, Li J, Li C, Zhang Z, Zhang W, Yao J, Guan M, Guo Z, Li C, Zhou L. High filling rate digital PCR through-hole array chip with double independent S-shaped flow channels. BIOMICROFLUIDICS 2020; 14:034109. [PMID: 32509051 PMCID: PMC7266645 DOI: 10.1063/5.0006374] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/16/2020] [Indexed: 05/13/2023]
Abstract
Sample digital technology is a powerful method for absolute quantification of target molecules such as nucleic acids and proteins. The excellent sample stability and mass production capability has enabled the development of microwell array-based sample digitizing methods. However, in current microwell array chips, samples are loaded by the liquid scraping method, which requires complex manual operation and results in a low filling rate and limited hole filling uniformity. Here, we perform sample loading of a through-hole array chip by a microfluidics-driven method and design a double independent S-shaped flow channels sandwiched through-hole array chip. Because of the capillary force and capillary burst pressure, the sample flowing in the channel can be trapped into through-holes, but cannot flow through the other side. Via air flow and displacement of the remaining sample in the channel, the sample can be partitioned consistently, with zero surplus sample residue in the channel. We evaluated the actual performance of the sample-loading process: the chip enables 99.10% filling rate of 18 500 through-holes, with a grayscale coefficient of variation value of 6.03% determined from fluorescence images. In performing digital polymerase chain reaction on chip, the chip demonstrates good performance for the absolute quantification of target DNA. The simple and robust design of our chip, with excellent filling rate and microsample uniformity, indicates potential for use in a variety of sample digitization applications.
Collapse
Affiliation(s)
| | - Jinze Li
- CAS Key Laboratory of Bio-medical Diagnostics,
Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of
Sciences, Suzhou, Jiangsu 215163, People's Republic of
China
| | | | | | | | | | - Ming Guan
- Huashan Hospital, Fudan University,
Shanghai 200040, People's Republic of China
| | | | - Chao Li
- CAS Key Laboratory of Bio-medical Diagnostics,
Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of
Sciences, Suzhou, Jiangsu 215163, People's Republic of
China
| | - Lianqun Zhou
- Author to whom correspondence should be addressed:
| |
Collapse
|
19
|
Fung CW, Chan SN, Wu AR. Microfluidic single-cell analysis-Toward integration and total on-chip analysis. BIOMICROFLUIDICS 2020; 14:021502. [PMID: 32161631 PMCID: PMC7060088 DOI: 10.1063/1.5131795] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/25/2020] [Indexed: 06/10/2023]
Abstract
Various types of single-cell analyses are now extensively used to answer many biological questions, and with this growth in popularity, potential drawbacks to these methods are also becoming apparent. Depending on the specific application, workflows can be laborious, low throughput, and run the risk of contamination. Microfluidic designs, with their advantages of being high throughput, low in reaction volume, and compatible with bio-inert materials, have been widely used to improve single-cell workflows in all major stages of single-cell applications, from cell sorting to lysis, to sample processing and readout. Yet, designing an integrated microfluidic chip that encompasses the entire single-cell workflow from start to finish remains challenging. In this article, we review the current microfluidic approaches that cover different stages of processing in single-cell analysis and discuss the prospects and challenges of achieving a full integrated workflow to achieve total single-cell analysis in one device.
Collapse
Affiliation(s)
- Cheuk Wang Fung
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Shek Nga Chan
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Angela Ruohao Wu
- Author to whom correspondence should be addressed:. Tel.: +852 3469-2577
| |
Collapse
|
20
|
Opalski AS, Ruszczak A, Promovych Y, Horka M, Derzsi L, Garstecki P. Combinatorial Antimicrobial Susceptibility Testing Enabled by Non-Contact Printing. MICROMACHINES 2020; 11:mi11020142. [PMID: 32012854 PMCID: PMC7074582 DOI: 10.3390/mi11020142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/21/2022]
Abstract
We demonstrate the utility of non-contact printing to fabricate the mAST—an easy-to-operate, microwell-based microfluidic device for combinatorial antibiotic susceptibility testing (AST) in a point-of-care format. The wells are prefilled with antibiotics in any desired concentration and combination by non-contact printing (spotting). For the execution of the AST, the only requirements are the mAST device, the sample, and the incubation chamber. Bacteria proliferation can be continuously monitored by using an absorbance reader. We investigate the profile of resistance of two reference Escherichia coli strains, report the minimum inhibitory concentration (MIC) for single antibiotics, and assess drug–drug interactions in cocktails by using the Bliss independence model.
Collapse
Affiliation(s)
| | | | | | | | - Ladislav Derzsi
- Correspondence: (L.D.); (P.G.); Tel.: +48-(22)-343-22-33 (L.D. & P.G.)
| | - Piotr Garstecki
- Correspondence: (L.D.); (P.G.); Tel.: +48-(22)-343-22-33 (L.D. & P.G.)
| |
Collapse
|
21
|
Padmanabhan S, Han JY, Nanayankkara I, Tran K, Ho P, Mesfin N, White I, DeVoe DL. Enhanced sample filling and discretization in thermoplastic 2D microwell arrays using asymmetric contact angles. BIOMICROFLUIDICS 2020; 14:014113. [PMID: 32095199 PMCID: PMC7028432 DOI: 10.1063/1.5126938] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 02/09/2020] [Indexed: 05/04/2023]
Abstract
Sample filling and discretization within thermoplastic 2D microwell arrays is investigated toward the development of low cost disposable microfluidics for passive sample discretization. By using a high level of contact angle asymmetry between the filling channel and microwell surfaces, a significant increase in the range of well geometries that can be successfully filled is revealed. The performance of various array designs is characterized numerically and experimentally to assess the impact of contact angle asymmetry and device geometry on sample filling and discretization, resulting in guidelines to ensure robust microwell filling and sample isolation over a wide range of well dimensions. Using the developed design rules, reliable and bubble-free sample filling and discretization is achieved in designs with critical dimensions ranging from 20 μm to 800 μm. The resulting devices are demonstrated for discretized nucleic acid amplification by performing loop-mediated isothermal amplification for the detection of the mecA gene associated with methicillin-resistant Staphylococcus aureus.
Collapse
Affiliation(s)
- S. Padmanabhan
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, USA
| | - J. Y. Han
- Department of Mechanical Engineering, University of Maryland, College Park, Maryland 20742, USA
| | - I. Nanayankkara
- Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA
| | - K. Tran
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, USA
| | - P. Ho
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, USA
| | - N. Mesfin
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland 20742, USA
| | - I. White
- Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA
| | - D. L. DeVoe
- Author to whom correspondence should be addressed:. Tel.: +1-301-405-8125
| |
Collapse
|
22
|
Abraham DH, Anttila MM, Gallion LA, Petersen BV, Proctor A, Allbritton NL. Design of an automated capillary electrophoresis platform for single-cell analysis. Methods Enzymol 2019; 628:191-221. [PMID: 31668230 DOI: 10.1016/bs.mie.2019.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Single-cell analysis of cellular contents by highly sensitive analytical instruments is known as chemical cytometry. A chemical cytometer typically samples one cell at a time, quantifies the cellular contents of interest, and then processes and reports that data. Automation adds the potential to perform this entire sequence of events with minimal intervention, increasing throughput and repeatability. In this chapter, we discuss the design considerations for an automated capillary electrophoresis-based instrument for assay of enzymatic activity within single cells. We describe the key requirements of the microscope base and capillary electrophoresis platforms. We also provide detailed protocols and schematic designs of our cell isolation, lysis, sampling, and detection strategies. Additionally, we describe our signal processing and instrument automation workflows. The described automated system has demonstrated single-cell throughput at rates above 100cells/h and analyte limits of detection as low as 10-20mol.
Collapse
Affiliation(s)
- David H Abraham
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States
| | - Matthew M Anttila
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States
| | - Luke A Gallion
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States
| | - Brae V Petersen
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States
| | - Angela Proctor
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States
| | - Nancy L Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, United States; Joint Department of Biomedical Engineering, University of North Carolina, Chapel and North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
23
|
Abstract
Advances in microfluidic techniques have prompted researchers to study the inherent heterogeneity of single cells in cell populations.
Collapse
Affiliation(s)
- Qiushi Huang
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Sifeng Mao
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Mashooq Khan
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Jin-Ming Lin
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| |
Collapse
|
24
|
Sreejith KR, Ooi CH, Jin J, Dao DV, Nguyen NT. Digital polymerase chain reaction technology - recent advances and future perspectives. LAB ON A CHIP 2018; 18:3717-3732. [PMID: 30402632 DOI: 10.1039/c8lc00990b] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Digital polymerase chain reaction (dPCR) technology has remained a "hot topic" in the last two decades due to its potential applications in cell biology, genetic engineering, and medical diagnostics. Various advanced techniques have been reported on sample dispersion, thermal cycling and output monitoring of digital PCR. However, a fully automated, low-cost and handheld digital PCR platform has not been reported in the literature. This paper attempts to critically evaluate the recent developments in techniques for sample dispersion, thermal cycling and output evaluation for dPCR. The techniques are discussed in terms of hardware simplicity, portability, cost-effectiveness and suitability for automation. The present paper also discusses the research gaps observed in each step of dPCR and concludes with possible improvements toward portable, low-cost and automatic digital PCR systems.
Collapse
Affiliation(s)
- Kamalalayam Rajan Sreejith
- Queensland Micro- and Nanotechnology Centre, Griffith University, 170 Kessels Road, 4111 Queensland, Australia.
| | | | | | | | | |
Collapse
|
25
|
Liu Y, Ren D, Ling X, Liang W, Li J, You Z, Yalikun Y, Tanaka Y. Time Sequential Single-Cell Patterning with High Efficiency and High Density. SENSORS 2018; 18:s18113672. [PMID: 30380644 PMCID: PMC6264106 DOI: 10.3390/s18113672] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/24/2022]
Abstract
Single-cell capture plays an important role in single-cell manipulation and analysis. This paper presents a microfluidic device for deterministic single-cell trapping based on the hydrodynamic trapping mechanism. The device is composed of an S-shaped loop channel and thousands of aligned trap units. This arrayed structure enables each row of the device to be treated equally and independently, as it has row periodicity. A theoretical model was established and a simulation was conducted to optimize the key geometric parameters, and the performance was evaluated by conducting experiments on MCF-7 and Jurkat cells. The results showed improvements in single-cell trapping ability, including loading efficiency, capture speed, and the density of the patterned cells. The optimized device can achieve a capture efficiency of up to 100% and single-cell capture efficiency of up to 95%. This device offers 200 trap units in an area of 1 mm2, which enables 100 single cells to be observed simultaneously using a microscope with a 20× objective lens. One thousand cells can be trapped sequentially within 2 min; this is faster than the values obtained with previously reported devices. Furthermore, the cells can also be recovered by reversely infusing solutions. The structure can be easily extended to a large scale, and a patterned array with 32,000 trap sites was accomplished on a single chip. This device can be a powerful tool for high-throughput single-cell analysis, cell heterogeneity investigation, and drug screening.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Precision Measurement Technology and Instruments, Tsinghua University, Beijing 100084, China.
| | - Dahai Ren
- State Key Laboratory of Precision Measurement Technology and Instruments, Tsinghua University, Beijing 100084, China.
| | - Xixin Ling
- State Key Laboratory of Precision Measurement Technology and Instruments, Tsinghua University, Beijing 100084, China.
| | - Weibin Liang
- State Key Laboratory of Precision Measurement Technology and Instruments, Tsinghua University, Beijing 100084, China.
| | - Jing Li
- State Key Laboratory of Precision Measurement Technology and Instruments, Tsinghua University, Beijing 100084, China.
| | - Zheng You
- State Key Laboratory of Precision Measurement Technology and Instruments, Tsinghua University, Beijing 100084, China.
| | - Yaxiaer Yalikun
- Center for Biosystems Dynamics Research (BDR), RIKEN, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan.
- Division of Materials Science, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara 630-0192, Japan.
| | - Yo Tanaka
- Center for Biosystems Dynamics Research (BDR), RIKEN, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
26
|
Antypas H, Veses-Garcia M, Weibull E, Andersson-Svahn H, Richter-Dahlfors A. A universal platform for selection and high-resolution phenotypic screening of bacterial mutants using the nanowell slide. LAB ON A CHIP 2018; 18:1767-1777. [PMID: 29781496 PMCID: PMC5996734 DOI: 10.1039/c8lc00190a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/07/2018] [Indexed: 06/08/2023]
Abstract
The Petri dish and microtiter plate are the golden standard for selection and screening of bacteria in microbiological research. To improve on the limited resolution and throughput of these methods, we developed a universal, user-friendly platform for selection and high-resolution phenotypic screening based on the nanowell slide. This miniaturized platform has an optimal ratio between throughput and assay complexity, holding 672 nanowells of 500 nl each. As monoclonality is essential in bacterial genetics, we used FACS to inoculate each nanowell with a single bacterium in 15 min. We further extended the protocol to select and sort only bacteria of interest from a mixed culture. We demonstrated this by isolating single transposon mutants generated by a custom-made transposon with dual selection for GFP fluorescence and kanamycin resistance. Optical compatibility of the nanowell slide enabled phenotypic screening of sorted mutants by spectrophotometric recording during incubation. By processing the absorbance data with our custom algorithm, a phenotypic screen for growth-associated mutations was performed. Alternatively, by processing fluorescence data, we detected metabolism-associated mutations, exemplified by a screen for β-galactosidase activity. Besides spectrophotometry, optical compatibility enabled us to perform microscopic analysis directly in the nanowells to screen for mutants with altered morphologies. Despite the miniaturized format, easy transition from nano- to macroscale cultures allowed retrieval of bacterial mutants for downstream genetic analysis, demonstrated here by a cloning-free single-primer PCR protocol. Taken together, our FACS-linked nanowell slide replaces manual selection of mutants on agar plates, and enables combined selection and phenotypic screening in a one-step process. The versatility of the nanowell slide, and the modular workflow built on mainstream technologies, makes our universal platform widely applicable in microbiological research.
Collapse
Affiliation(s)
- H. Antypas
- Swedish Medical Nanoscience Center
, Department of Neuroscience
, Karolinska Institutet
,
Stockholm
, Sweden
.
| | - M. Veses-Garcia
- Swedish Medical Nanoscience Center
, Department of Neuroscience
, Karolinska Institutet
,
Stockholm
, Sweden
.
| | - E. Weibull
- Division of Proteomics and Nanobiotechnology
, Science for Life Laboratory
, KTH-Royal Institute of Technology
,
Stockholm
, Sweden
| | - H. Andersson-Svahn
- Division of Proteomics and Nanobiotechnology
, Science for Life Laboratory
, KTH-Royal Institute of Technology
,
Stockholm
, Sweden
| | - A. Richter-Dahlfors
- Swedish Medical Nanoscience Center
, Department of Neuroscience
, Karolinska Institutet
,
Stockholm
, Sweden
.
| |
Collapse
|
27
|
Wang C, Liu W, Wei Q, Ren L, Tan M, Yu Y. A novel dual-well array chip for efficiently trapping single-cell in large isolated micro-well without complicated accessory equipment. BIOMICROFLUIDICS 2018; 12:034103. [PMID: 29774084 PMCID: PMC5938174 DOI: 10.1063/1.5030203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/20/2018] [Indexed: 06/08/2023]
Abstract
Conventional cell-sized well arrays have advantages of high occupancy, simple operation, and low cost for capturing single-cells. However, they have insufficient space for including reagents required for cell treatment or analysis, which restricts the wide application of cell-sized well arrays as a single-cell research tool alone. Here, we present a novel dual-well array chip, which integrates capture-wells (20 μm in diameter) with reaction-wells (100 μm in diameter) and describe a flow method for convenient single-cell analysis requiring neither complicated infra-structure nor high expenditure, while enabling highly efficient single cell trapping (75.8%) with only 11.3% multi-cells. Briefly, the cells are first loaded into the dual-wells by gravity and then multi-cells in the reaction-wells are washed out by phosphate buffer saline. Next, biochemical reagents are loaded into reaction-wells using the scraping method and the chip is packed as a sandwich structure. We thereby successfully measured intracellular β-galactosidase activity of K562 cells at the single-cell level. We also used computational simulations to illustrate the working principle of dual-well structure and found out a relationship between the wall shear stress distribution and the aspect ratio of the dual-well array chip which provides theoretical guidance for designing multi-wells chip for convenient single-cell analysis. Our work produced the first dual-well chip that can simultaneously provide a high occupancy rate for single cells and sufficient space for reagents, as well as being low in cost and simple to operate. We believe that the feasibility and convenience of our method will enhance its use as a practical single-cell research tool.
Collapse
Affiliation(s)
| | | | | | | | | | - Yude Yu
- Author to whom correspondence should be addressed: . Tel.: 86-10-82304979
| |
Collapse
|
28
|
Steinke M, Zunhammer F, Chatzopoulou EI, Teller H, Schütze K, Walles H, Rädler JO, Grüttner C. Rapid Analysis of Cell-Nanoparticle Interactions using Single-Cell Raman Trapping Microscopy. Angew Chem Int Ed Engl 2018; 57:4946-4950. [DOI: 10.1002/anie.201713151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/06/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Maria Steinke
- Fraunhofer Institute for Silicate Research ISC; c/o University Hospital Würzburg; Chair of Tissue Engineering and Regenerative Medicine; Röntgenring 11 97070 Würzburg Germany
| | | | - Elisavet I. Chatzopoulou
- Ludwig-Maximilians-University Munich; Faculty of Physics; Geschwister-Scholl-Platz 1 80539 München Germany
| | - Henrik Teller
- Micromod Partikeltechnologie GmbH; Friedrich-Barnewitz-Straße 4 18119 Rostock Germany
| | | | - Heike Walles
- Fraunhofer Institute for Silicate Research ISC; c/o University Hospital Würzburg; Chair of Tissue Engineering and Regenerative Medicine; Röntgenring 11 97070 Würzburg Germany
| | - Joachim O. Rädler
- Ludwig-Maximilians-University Munich; Faculty of Physics; Geschwister-Scholl-Platz 1 80539 München Germany
| | - Cordula Grüttner
- Micromod Partikeltechnologie GmbH; Friedrich-Barnewitz-Straße 4 18119 Rostock Germany
| |
Collapse
|
29
|
Steinke M, Zunhammer F, Chatzopoulou EI, Teller H, Schütze K, Walles H, Rädler JO, Grüttner C. Rapid Analysis of Cell-Nanoparticle Interactions using Single-Cell Raman Trapping Microscopy. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201713151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Maria Steinke
- Fraunhofer Institute for Silicate Research ISC; c/o University Hospital Würzburg; Chair of Tissue Engineering and Regenerative Medicine; Röntgenring 11 97070 Würzburg Germany
| | | | - Elisavet I. Chatzopoulou
- Ludwig-Maximilians-University Munich; Faculty of Physics; Geschwister-Scholl-Platz 1 80539 München Germany
| | - Henrik Teller
- Micromod Partikeltechnologie GmbH; Friedrich-Barnewitz-Straße 4 18119 Rostock Germany
| | | | - Heike Walles
- Fraunhofer Institute for Silicate Research ISC; c/o University Hospital Würzburg; Chair of Tissue Engineering and Regenerative Medicine; Röntgenring 11 97070 Würzburg Germany
| | - Joachim O. Rädler
- Ludwig-Maximilians-University Munich; Faculty of Physics; Geschwister-Scholl-Platz 1 80539 München Germany
| | - Cordula Grüttner
- Micromod Partikeltechnologie GmbH; Friedrich-Barnewitz-Straße 4 18119 Rostock Germany
| |
Collapse
|
30
|
Ven K, Vanspauwen B, Pérez-Ruiz E, Leirs K, Decrop D, Gerstmans H, Spasic D, Lammertyn J. Target Confinement in Small Reaction Volumes Using Microfluidic Technologies: A Smart Approach for Single-Entity Detection and Analysis. ACS Sens 2018; 3:264-284. [PMID: 29363316 DOI: 10.1021/acssensors.7b00873] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Over the last decades, the study of cells, nucleic acid molecules, and proteins has evolved from ensemble measurements to so-called single-entity studies. The latter offers huge benefits, not only as biological research tools to examine heterogeneities among individual entities within a population, but also as biosensing tools for medical diagnostics, which can reach the ultimate sensitivity by detecting single targets. Whereas various techniques for single-entity detection have been reported, this review focuses on microfluidic systems that physically confine single targets in small reaction volumes. We categorize these techniques as droplet-, microchamber-, and nanostructure-based and provide an overview of their implementation for studying single cells, nucleic acids, and proteins. We furthermore reflect on the advantages and limitations of these techniques and highlight future opportunities in the field.
Collapse
Affiliation(s)
- Karen Ven
- Department
of Biosystems, KU Leuven - University of Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Bram Vanspauwen
- Department
of Biosystems, KU Leuven - University of Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Elena Pérez-Ruiz
- Department
of Biosystems, KU Leuven - University of Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Karen Leirs
- Department
of Biosystems, KU Leuven - University of Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Deborah Decrop
- Department
of Biosystems, KU Leuven - University of Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Hans Gerstmans
- Department
of Biosystems, KU Leuven - University of Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
- Department
of Applied biosciences, Ghent University, Valentyn Vaerwyckweg 1 - building
C, 9000 Gent, Belgium
- Department
of Biosystems, KU Leuven - University of Leuven, Kasteelpark Arenberg
21, 3001 Leuven, Belgium
| | - Dragana Spasic
- Department
of Biosystems, KU Leuven - University of Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Jeroen Lammertyn
- Department
of Biosystems, KU Leuven - University of Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| |
Collapse
|
31
|
Duan BK, Cavanagh PE, Li X, Walt DR. Ultrasensitive Single-Molecule Enzyme Detection and Analysis Using a Polymer Microarray. Anal Chem 2018; 90:3091-3098. [PMID: 29425025 DOI: 10.1021/acs.analchem.7b03980] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
This report describes a novel method for isolating and detecting individual enzyme molecules using polymer arrays of picoliter microwells. A fluidic flow-cell device containing an array of microwells is fabricated in cyclic olefin polymer (COP). The use of COP microwell arrays simplifies experiments by eliminating extensive device preparation and surface functionalization that are common in other microwell array formats. Using a simple and robust loading method to introduce the reaction solution, individual enzyme molecules are trapped in picoliter microwells and subsequently isolated and sealed by fluorinated oil. The sealing is stable for hours in the COP device. The picoliter microwell device can measure enzyme concentrations in the low-femtomolar range by counting the number of active wells using a digital read-out. These picoliter microwell arrays can also easily be regenerated and reused.
Collapse
Affiliation(s)
- Barrett K Duan
- Department of Pathology , Harvard Medical School , Brigham and Women's Hospital, Wyss Institute for Biologically Inspired Engineering, Building for Transformative Medicine, 60 Fenwood Road , Boston , Massachusetts 02115 , United States
| | - Peter E Cavanagh
- Department of Pathology , Harvard Medical School , Brigham and Women's Hospital, Wyss Institute for Biologically Inspired Engineering, Building for Transformative Medicine, 60 Fenwood Road , Boston , Massachusetts 02115 , United States
| | - Xiang Li
- Department of Pathology , Harvard Medical School , Brigham and Women's Hospital, Wyss Institute for Biologically Inspired Engineering, Building for Transformative Medicine, 60 Fenwood Road , Boston , Massachusetts 02115 , United States
| | - David R Walt
- Department of Pathology , Harvard Medical School , Brigham and Women's Hospital, Wyss Institute for Biologically Inspired Engineering, Building for Transformative Medicine, 60 Fenwood Road , Boston , Massachusetts 02115 , United States
| |
Collapse
|
32
|
Mansoorifar A, Koklu A, Ma S, Raj GV, Beskok A. Electrical Impedance Measurements of Biological Cells in Response to External Stimuli. Anal Chem 2018; 90:4320-4327. [PMID: 29402081 DOI: 10.1021/acs.analchem.7b05392] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Dielectric spectroscopy (DS) is a noninvasive technique for real-time measurements of the impedance spectra of biological cells. DS enables characterization of cellular dielectric properties such as membrane capacitance and cytoplasmic conductivity. We have developed a lab-on-a-chip device that uses an electro-activated microwells array for capturing, DS measurements, and unloading of biological cells. Impedance measurements were conducted at 0.2 V in the 10 kHz to 40 MHz range with 6 s time resolution. An equivalent circuit model was developed to extract the cell membrane capacitance and cell cytoplasmic conductivity from the impedance spectra. A human prostate cancer cell line, PC-3, was used to evaluate the device performance. Suspension of PC-3 cells in low conductivity buffers (LCB) enhanced their dielectrophoretic trapping and impedance response. We report the time course of the variations in dielectric properties of PC-3 cells suspended in LCB and their response to sudden pH change from a pH of 7.3 to a pH of 5.8. Importantly, we demonstrated that our device enabled real-time measurements of dielectric properties of live cancer cells and allowed the assessment of the cellular response to variations in buffer conductivity and pH. These data support further development of this device toward single cell measurements.
Collapse
Affiliation(s)
- Amin Mansoorifar
- Department of Mechanical Engineering , Southern Methodist University , Dallas , Texas 75205 , United States
| | - Anil Koklu
- Department of Mechanical Engineering , Southern Methodist University , Dallas , Texas 75205 , United States
| | - Shihong Ma
- Departments of Urology and Pharmacology , University of Texas Southwestern Medical Center , Dallas , Texas 75390 , United States
| | - Ganesh V Raj
- Departments of Urology and Pharmacology , University of Texas Southwestern Medical Center , Dallas , Texas 75390 , United States
| | - Ali Beskok
- Department of Mechanical Engineering , Southern Methodist University , Dallas , Texas 75205 , United States
| |
Collapse
|
33
|
Giupponi E, Visone R, Occhetta P, Colombo F, Rasponi M, Candiani G. Development of a microfluidic platform for high-throughput screening of non-viral gene delivery vectors. Biotechnol Bioeng 2017; 115:775-784. [DOI: 10.1002/bit.26506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/27/2017] [Accepted: 11/28/2017] [Indexed: 01/14/2023]
Affiliation(s)
- Elisa Giupponi
- Department of Chemistry; Materials, and Chemical Engineering “Giulio Natta,”; Politecnico di Milano; Milan Italy
| | - Roberta Visone
- Department of Electronics; Information and Bioengineering; Politecnico di Milano; Milan Italy
| | - Paola Occhetta
- Department of Electronics; Information and Bioengineering; Politecnico di Milano; Milan Italy
- Department of Biomedicine; University Hospital Basel; University of Basel; Basel Switzerland
| | - Federica Colombo
- Department of Chemistry; Materials, and Chemical Engineering “Giulio Natta,”; Politecnico di Milano; Milan Italy
| | - Marco Rasponi
- Department of Electronics; Information and Bioengineering; Politecnico di Milano; Milan Italy
| | - Gabriele Candiani
- Department of Chemistry; Materials, and Chemical Engineering “Giulio Natta,”; Politecnico di Milano; Milan Italy
- “The Protein Factory” Research Centre; Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta,”; Politecnico di Milano, and Department of Biotechnology and Life Science - University of Insubria; 20131, Milan Italy
| |
Collapse
|
34
|
Kamalakshakurup G, Lee AP. High-efficiency single cell encapsulation and size selective capture of cells in picoliter droplets based on hydrodynamic micro-vortices. LAB ON A CHIP 2017; 17:4324-4333. [PMID: 29138790 DOI: 10.1039/c7lc00972k] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Single cell analysis has emerged as a paradigm shift in cell biology to understand the heterogeneity of individual cells in a clone for pathological interrogation. Microfluidic droplet technology is a compelling platform to perform single cell analysis by encapsulating single cells inside picoliter-nanoliter (pL-nL) volume droplets. However, one of the primary challenges for droplet based single cell assays is single cell encapsulation in droplets, currently achieved either randomly, dictated by Poisson statistics, or by hydrodynamic techniques. In this paper, we present an interfacial hydrodynamic technique which initially traps the cells in micro-vortices, and later releases them one-to-one into the droplets, controlled by the width of the outer streamline that separates the vortex from the flow through the streaming passage adjacent to the aqueous-oil interface (dgap). One-to-one encapsulation is achieved at a dgap equal to the radius of the cell, whereas complete trapping of the cells is realized at a dgap smaller than the radius of the cell. The unique feature of this technique is that it can perform 1. high efficiency single cell encapsulations and 2. size-selective capturing of cells, at low cell loading densities. Here we demonstrate these two capabilities with a 50% single cell encapsulation efficiency and size selective separation of platelets, RBCs and WBCs from a 10× diluted blood sample (WBC capture efficiency at 70%). The results suggest a passive, hydrodynamic micro-vortex based technique capable of performing high-efficiency single cell encapsulation for cell based assays.
Collapse
|
35
|
Silva NFD, Magalhães JMCS, Freire C, Delerue-Matos C. Electrochemical biosensors for Salmonella: State of the art and challenges in food safety assessment. Biosens Bioelectron 2017; 99:667-682. [PMID: 28858763 DOI: 10.1016/j.bios.2017.08.019] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/03/2017] [Accepted: 08/07/2017] [Indexed: 10/19/2022]
Abstract
According to the recent statistics, Salmonella is still an important public health issue in the whole world. Legislated reference methods, based on counting plate methods, are sensitive enough but are inadequate as an effective emergency response tool, and are far from a rapid device, simple to use out of lab. An overview of the commercially available rapid methods for Salmonella detection is provided along with a critical discussion of their limitations, benefits and potential use in a real context. The distinguished potentialities of electrochemical biosensors for the development of rapid devices are highlighted. The state-of-art and the newest technologic approaches in electrochemical biosensors for Salmonella detection are presented and a critical analysis of the literature is made in an attempt to identify the current challenges towards a complete solution for Salmonella detection in microbial food control based on electrochemical biosensors.
Collapse
Affiliation(s)
- Nádia F D Silva
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, 4200-072 Porto, Portugal
| | - Júlia M C S Magalhães
- REQUIMTE/LAQV, Departamento de Engenharia Química, Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal.
| | - Cristina Freire
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| | - Cristina Delerue-Matos
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, 4200-072 Porto, Portugal
| |
Collapse
|
36
|
Wang C, Liu W, Tan M, Sun H, Yu Y. An open-pattern droplet-in-oil planar array for single cell analysis based on sequential inkjet printing technology. BIOMICROFLUIDICS 2017; 11:044106. [PMID: 28794816 PMCID: PMC5519398 DOI: 10.1063/1.4995294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/10/2017] [Indexed: 05/08/2023]
Abstract
Cellular heterogeneity represents a fundamental principle of cell biology for which a readily available single-cell research tool is urgently required. Here, we present a novel method combining cell-sized well arrays with sequential inkjet printing. Briefly, K562 cells with phosphate buffer saline buffer were captured at high efficiency (74.5%) in a cell-sized well as a "primary droplet" and sealed using fluorinated oil. Then, piezoelectric inkjet printing technology was adapted to precisely inject the cell lysis buffer and the fluorogenic substrate, fluorescein-di-β-D-galactopyranoside, as a "secondary droplet" to penetrate the sealing oil and fuse with the "primary droplet." We thereby successfully measured the intracellular β-galactosidase activity of K562 cells at the single-cell level. Our method allows, for the first time, the ability to simultaneously accommodate the high occupancy rate of single cells and sequential addition of reagents while retaining an open structure. We believe that the feasibility and flexibility of our method will enhance its use as a universal single-cell research tool as well as accelerate the adoption of inkjet printing in the study of cellular heterogeneity.
Collapse
Affiliation(s)
| | | | | | - Hongbo Sun
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, People's Republic of China
| | | |
Collapse
|
37
|
Béné MC. Microfluidics in flow cytometry and related techniques. Int J Lab Hematol 2017; 39 Suppl 1:93-97. [PMID: 28447416 DOI: 10.1111/ijlh.12669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/03/2017] [Indexed: 12/17/2022]
Abstract
Technological advances in laboratory automation are now well understood and applied as they considerably improved the speed and robustness of haematological laboratory data, in the companion fields of blood analyzers and flow cytometry. Still rather confidential is the field of microfluidics, mostly confined so far to academic settings and research laboratories. The literature in the field of microfluidics is growing and applications in hematology range from cell counting to flow cytometry, cell sorting, or ex vivo testing. A literature search allows to identify many innovative solutions developed to master the specific physics of fluid movements in microchips. Miniaturization also dwells on findings that have emerged from different areas such as electronics and nanoengineering. This review proposes an overview of the major principles guiding developments in microfluidics and describes a necessarily limited and nonexhaustive series of specific applications. Readers are strongly encouraged to consult the documents referred to in the references section to learn more about this world knocking at our door and possibly liable to revolutionize our profession of hematology biologists in a not so far future.
Collapse
Affiliation(s)
- M C Béné
- Hematology Biology, University Hospital, Nantes, France
| |
Collapse
|
38
|
Cao L, Cui X, Hu J, Li Z, Choi JR, Yang Q, Lin M, Ying Hui L, Xu F. Advances in digital polymerase chain reaction (dPCR) and its emerging biomedical applications. Biosens Bioelectron 2017; 90:459-474. [PMID: 27818047 DOI: 10.1016/j.bios.2016.09.082] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 09/23/2016] [Accepted: 09/24/2016] [Indexed: 12/18/2022]
Abstract
Since the invention of polymerase chain reaction (PCR) in 1985, PCR has played a significant role in molecular diagnostics for genetic diseases, pathogens, oncogenes and forensic identification. In the past three decades, PCR has evolved from end-point PCR, through real-time PCR, to its current version, which is the absolute quantitive digital PCR (dPCR). In this review, we first discuss the principles of all key steps of dPCR, i.e., sample dispersion, amplification, and quantification, covering commercialized apparatuses and other devices still under lab development. We highlight the advantages and disadvantages of different technologies based on these steps, and discuss the emerging biomedical applications of dPCR. Finally, we provide a glimpse of the existing challenges and future perspectives for dPCR.
Collapse
Affiliation(s)
- Lei Cao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Xingye Cui
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Jie Hu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Zedong Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Jane Ru Choi
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Qingzhen Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Li Ying Hui
- Foundation of State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, PR China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
39
|
Brown CL, Fleischauer V, Heo J. High-throughput Screening of Erratic Cell Volume Regulation Using a Hydrogel-based Single-cell Microwell Array. ANAL SCI 2017; 33:525-530. [PMID: 28392532 DOI: 10.2116/analsci.33.525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Here, we report that a single-cell microwell array based on photocrosslinked hydrogel can be used to screen cells exhibiting a defective regulatory volume decrease (RVD) in high-throughput. The RVD is a regulatory function of cells that maintains cell volume homeostasis in a hypotonic medium. Single Madin-Darby canine kidney (MDCK) cells grown in the microwells were loaded with a volume-sensitive fluorescence dye. Changes in the volume of discrete single cells were traced for 20 min in a hypotonic solution using a wide-field fluorescence microscopy. The volume changes of more than 100 single cells were analyzed simultaneously using time-lapse fluorescence micrographs. Cells showing erratic RVD could be easily screened from the image analysis. Nearly 40% of the MDCK single cells exhibited weak, or no, RVD. Since other previously reported methods could not detect as many changes in the volume of discrete singles cells as the method used in this report, we anticipate that our reported method will provide an efficient way of elucidating the RVD mechanisms of cells that have not yet been completely understood.
Collapse
|
40
|
Berthuy OI, Muldur SK, Rossi F, Colpo P, Blum LJ, Marquette CA. Multiplex cell microarrays for high-throughput screening. LAB ON A CHIP 2016; 16:4248-4262. [PMID: 27731880 DOI: 10.1039/c6lc00831c] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Microarray technology was developed in the early 1990s to measure the transcription levels of thousands of genes in parallel. The basic premise of high-density arraying has since been expanded to create cell microarrays. Cells on chip are powerful experimental tools for high-throughput and multiplex screening of samples or cellular functions. Miniaturization increases assay throughput while reducing both reagent consumption and cell population heterogeneity effect, making these systems attractive for a wide range of assays, from drug discovery to toxicology, stem cell research and therapy. It is usual to functionalize the surface of a substrate to design cell microarrays. One form of cell microarrays, the transfected cell microarray, wherein plasmid DNA or siRNA spotted on the surface of a substrate is reverse-transfected locally into adherent cells, has become a standard tool for parallel cell-based analysis. With the advent of technology, cells can also be directly spotted onto functionalized surfaces using robotic fluid-dispensing devices or printed directly on bio-ink material. We are providing herein an overview of the latest developments in optical cell microarrays allowing high-throughput and high-content analysis.
Collapse
Affiliation(s)
- Ophélie I Berthuy
- Univ Lyon, Université Lyon1, CNRS, INSA, CPE-Lyon, ICBMS, UMR 5246, 43, Bd du 11 novembre 1918, 69622 Villeurbanne cedex, France.
| | - Sinan K Muldur
- Européen Commission, Joint Research Centre, Institute for Heath and Consumer Protection, Ispra, VA, Italy
| | - François Rossi
- Européen Commission, Joint Research Centre, Institute for Heath and Consumer Protection, Ispra, VA, Italy
| | - Pascal Colpo
- Européen Commission, Joint Research Centre, Institute for Heath and Consumer Protection, Ispra, VA, Italy
| | - Loïc J Blum
- Univ Lyon, Université Lyon1, CNRS, INSA, CPE-Lyon, ICBMS, UMR 5246, 43, Bd du 11 novembre 1918, 69622 Villeurbanne cedex, France.
| | - Christophe A Marquette
- Univ Lyon, Université Lyon1, CNRS, INSA, CPE-Lyon, ICBMS, UMR 5246, 43, Bd du 11 novembre 1918, 69622 Villeurbanne cedex, France.
| |
Collapse
|
41
|
Vajrala VS, Suraniti E, Rigoulet M, Devin A, Sojic N, Arbault S. PDMS microwells for multi-parametric monitoring of single mitochondria on a large scale: a study of their individual membrane potential and endogenous NADH. Integr Biol (Camb) 2016; 8:836-43. [PMID: 27384613 DOI: 10.1039/c6ib00064a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Microwell arrays have been developed to monitor simultaneously, and on a large scale, multiple metabolic responses of single mitochondria. Wells of 50 to 1000 μm-diameter were prepared based on easy structuration of thin polydimethylsiloxane layers (PDMS; 100 μm thickness). Their surface treatment with oxygen plasma allowed the immobilization in situ and observation with time of populations of single isolated mitochondria. Their metabolic activities could be monitored individually by fluorescence microscopy under several activation/inhibition conditions. We measured the concomitant variations of two main metabolic parameters - the endogenous NADH level and the internal membrane potential difference Δψ owing to a cationic fluorescent probe (TMRM) - at energized, uncoupled and inhibited stages of the mitochondrial respiratory chain. Microwell arrays allowed analyses on large populations, and consequently statistical studies with a single organelle resolution. Thus, we observed rapid individual polarizations and depolarizations of mitochondria following their supply with the energetic substrate, while an averaged global polarization (increase of TMRM fluorescence within mitochondria) and NADH increase were detected for the whole population. In addition, statistical correlation studies show that the NADH content of all mitochondria tends toward a metabolic limit and that their polarization-depolarization ability is ubiquitous. These results demonstrate that PDMS microwell platforms provide an innovative approach to better characterize the individual metabolic status of isolated mitochondria, possibly as a function of their cell or organ origin or in different physio-pathological situations.
Collapse
|
42
|
Lin CH, Chang HC, Hsu CH. A Microfluidic Platform for High-throughput Single-cell Isolation and Culture. J Vis Exp 2016. [PMID: 27341146 DOI: 10.3791/54105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Studying the heterogeneity of single cells is crucial for many biological questions, but is technically difficult. Thus, there is a need for a simple, yet high-throughput, method to perform single-cell culture experiments. Here, we report a microfluidic chip-based strategy for high-efficiency single-cell isolation (~77%) and demonstrate its capability of performing long-term single-cell culture (up to 7 d) and cellular heterogeneity analysis using clonogenic assay. These applications were demonstrated with KT98 mouse neural stem cells, and A549 and MDA-MB-435 human cancer cells. High single-cell isolation efficiency and long-term culture capability are achieved by using different sizes of microwells on the top and bottom of the microfluidic channel. The small microwell array is designed for precisely isolating single-cells, and the large microwell array is used for single-cell clonal culture in the microfluidic chip. This microfluidic platform constitutes an attractive approach for single-cell culture applications, due to its flexibility of adjustable cell culture spaces for different culture strategies, without decreasing isolation efficiency.
Collapse
Affiliation(s)
- Ching-Hui Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Taiwan; Tissue Engineering and Regenerative Medicine, National Chung Hsing University
| | - Hao-Chen Chang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Taiwan; Tissue Engineering and Regenerative Medicine, National Chung Hsing University
| | - Chia-Hsien Hsu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Taiwan; Tissue Engineering and Regenerative Medicine, National Chung Hsing University; Institute of NanoEngineering and MicroSystems, National Tsing Hua University;
| |
Collapse
|
43
|
Micro- and Nanoscale Technologies for Delivery into Adherent Cells. Trends Biotechnol 2016; 34:665-678. [PMID: 27287927 DOI: 10.1016/j.tibtech.2016.05.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 12/28/2022]
Abstract
Several recent micro- and nanotechnologies have provided novel methods for biological studies of adherent cells because the small features of these new biotools provide unique capabilities for accessing cells without the need for suspension or lysis. These novel approaches have enabled gentle but effective delivery of molecules into specific adhered target cells, with unprecedented spatial resolution. We review here recent progress in the development of these technologies with an emphasis on in vitro delivery into adherent cells utilizing mechanical penetration or electroporation. We discuss the major advantages and limitations of these approaches and propose possible strategies for improvements. Finally, we discuss the impact of these technologies on biological research concerning cell-specific temporal studies, for example non-destructive sampling and analysis of intracellular molecules.
Collapse
|
44
|
Hahm JI. Fundamental Properties of One-Dimensional Zinc Oxide Nanomaterials and Implementations in Various Detection Modes of Enhanced Biosensing. Annu Rev Phys Chem 2016. [PMID: 27215822 DOI: 10.1146/annurev‐physchem‐031215‐010949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Recent bioapplications of one-dimensional (1D) zinc oxide (ZnO) nanomaterials, despite the short development period, have shown promising signs as new sensors and assay platforms offering exquisite biomolecular sensitivity and selectivity. The incorporation of 1D ZnO nanomaterials has proven beneficial to various modes of biodetection owing to their inherent properties. The more widely explored electrochemical and electrical approaches tend to capitalize on the reduced physical dimensionality, yielding a high surface-to-volume ratio, as well as on the electrical properties of ZnO. The newer development of the use of 1D ZnO nanomaterials in fluorescence-based biodetection exploits the innate optical property of their high anisotropy. This review considers stimulating research advances made to identify and understand fundamental properties of 1D ZnO nanomaterials, and examines various biosensing modes utilizing them, while focusing on the unique optical properties of individual and ensembles of 1D ZnO nanomaterials specifically pertaining to their bio-optical applications in simple and complex fluorescence assays.
Collapse
Affiliation(s)
- Jong-In Hahm
- Department of Chemistry, Georgetown University, Washington, DC 20057;
| |
Collapse
|
45
|
Hahm JI. Fundamental Properties of One-Dimensional Zinc Oxide Nanomaterials and Implementations in Various Detection Modes of Enhanced Biosensing. Annu Rev Phys Chem 2016; 67:691-717. [PMID: 27215822 PMCID: PMC4894344 DOI: 10.1146/annurev-physchem-031215-010949] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent bioapplications of one-dimensional (1D) zinc oxide (ZnO) nanomaterials, despite the short development period, have shown promising signs as new sensors and assay platforms offering exquisite biomolecular sensitivity and selectivity. The incorporation of 1D ZnO nanomaterials has proven beneficial to various modes of biodetection owing to their inherent properties. The more widely explored electrochemical and electrical approaches tend to capitalize on the reduced physical dimensionality, yielding a high surface-to-volume ratio, as well as on the electrical properties of ZnO. The newer development of the use of 1D ZnO nanomaterials in fluorescence-based biodetection exploits the innate optical property of their high anisotropy. This review considers stimulating research advances made to identify and understand fundamental properties of 1D ZnO nanomaterials, and examines various biosensing modes utilizing them, while focusing on the unique optical properties of individual and ensembles of 1D ZnO nanomaterials specifically pertaining to their bio-optical applications in simple and complex fluorescence assays.
Collapse
Affiliation(s)
- Jong-In Hahm
- Department of Chemistry, Georgetown University, Washington, DC 20057;
| |
Collapse
|
46
|
Van Malderen SJM, Vergucht E, De Rijcke M, Janssen C, Vincze L, Vanhaecke F. Quantitative Determination and Subcellular Imaging of Cu in Single Cells via Laser Ablation-ICP-Mass Spectrometry Using High-Density Microarray Gelatin Standards. Anal Chem 2016; 88:5783-9. [PMID: 27149342 DOI: 10.1021/acs.analchem.6b00334] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This manuscript describes the development and characterization of a high-density microarray calibration standard, manufactured in-house and designed to overcome the limitations in precision, accuracy, and throughput of current calibration approaches for the quantification of elemental concentrations on the cellular level using laser ablation-inductively coupled plasma-mass spectrometry (LA-ICPMS). As a case study, the accumulation of Cu in the model organism Scrippsiella trochoidea resulting from transition metal exposure (ranging from 0.5 to 100 μg/L) was evaluated. After the Cu exposure, cells of this photosynthetic dinoflagellate were treated with a critical point drying protocol, transferred to a carbon stub, and sputter-coated with a Au layer for scanning electron microscopy (SEM) analysis. In subsequent LA-ICPMS analysis, approximately 100 cells of each population were individually ablated. This approach permitted the evaluation of the mean concentration of Cu in the cell population across different exposure levels and also allowed the examination of the cellular distribution of Cu within the populations. In a cross-validation exercise, subcellular LA-ICPMS imaging was demonstrated to corroborate synchrotron radiation confocal X-ray fluorescence (SR-XRF) microimaging of single cells investigated under in vivo conditions.
Collapse
Affiliation(s)
- Stijn J M Van Malderen
- Department of Analytical Chemistry, Ghent University , Campus Sterre, Krijgslaan 281 - S12, 9000 Ghent, Belgium
| | - Eva Vergucht
- Department of Analytical Chemistry, Ghent University , Campus Sterre, Krijgslaan 281 - S12, 9000 Ghent, Belgium
| | - Maarten De Rijcke
- Department of Applied Ecology and Environmental Biology, Ghent University , Jozef Plateaustraat 22, 9000 Ghent, Belgium
| | - Colin Janssen
- Department of Applied Ecology and Environmental Biology, Ghent University , Jozef Plateaustraat 22, 9000 Ghent, Belgium
| | - Laszlo Vincze
- Department of Analytical Chemistry, Ghent University , Campus Sterre, Krijgslaan 281 - S12, 9000 Ghent, Belgium
| | - Frank Vanhaecke
- Department of Analytical Chemistry, Ghent University , Campus Sterre, Krijgslaan 281 - S12, 9000 Ghent, Belgium
| |
Collapse
|
47
|
Affiliation(s)
- Sanjin Hosic
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Shashi K. Murthy
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, USA
| | - Abigail N. Koppes
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
48
|
Design of a microfluidic strategy for trapping and screening single cells. Med Eng Phys 2016; 38:33-40. [DOI: 10.1016/j.medengphy.2015.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 10/08/2015] [Accepted: 10/26/2015] [Indexed: 01/09/2023]
|
49
|
Chen CC, Liu YJ, Yao DJ. Paper-based device for separation and cultivation of single microalga. Talanta 2015; 145:60-5. [DOI: 10.1016/j.talanta.2015.04.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/10/2015] [Accepted: 04/06/2015] [Indexed: 01/11/2023]
|
50
|
Singh M, Zhuo X, Choi DS, Gonzalez LE, Wang J, Hahm JI. Effects of crystallographic facet-specific peptide adsorption along single ZnO nanorods on the characteristic fluorescence intensification on nanorod ends (FINE) phenomenon. NANOSCALE 2015; 7:18813-26. [PMID: 26509316 PMCID: PMC4636451 DOI: 10.1039/c5nr05354d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The precise effect of crystallographically discriminating biomolecular adsorption on the fluorescence intensification profiles of individual zinc oxide nanorod (ZnO NR) platforms was elucidated in this study by employing peptide binding epitopes biased towards particular ZnO crystal surfaces and isolating the peptides on given crystalline facets of ZnO NRs. Subsequently, the fluorescence emission profiles of the preferentially bound peptide cases on the basal versus prismic planes of ZnO NRs were carefully evaluated both experimentally and via computer simulations. The phenomenon of fluorescence intensification on NR ends (FINE) was persistently observed on the individual ZnO NR platforms, regardless of the location of the bound peptides. In contrast to the consistent occurrence of FINE, the degree and magnitude of FINE were largely influenced by the discriminatory peptide adsorption to different ZnO NR facets. The temporal stability of the fluorescence signal was also greatly affected by the selectively located peptides on the ZnO NR crystal when spatially resolved on different NR facets. Similarities and differences in the spatial and temporal fluorescence signal of the crystalline NR facet-specific versus -nonspecific biomolecular adsorption events were then compared. To further illuminate the basis of our experimental findings, we also performed finite-difference-time-domain (FDTD) calculations and examined the different degrees of FINE by modelling the biased peptide adsorption cases. Our multifaceted efforts, providing combined insight into the spatial and temporal characteristics of the biomolecular fluorescence signal characteristically governed by the biomolecular location on the specific NR facets, will be valuable for novel applications and accurate signal interpretation of ZnO NR-based biosensors in many rapidly growing, highly miniaturized biodetection configurations.
Collapse
Affiliation(s)
- Manpreet Singh
- Department of Chemistry, Georgetown University, 37th & O Sts. NW, Washington, DC 20057, USA.
| | | | | | | | | | | |
Collapse
|