1
|
Farkas E, Dóra Kovács K, Szekacs I, Peter B, Lagzi I, Kitahata H, Suematsu NJ, Horvath R. Kinetic monitoring of molecular interactions during surfactant-driven self-propelled droplet motion by high spatial resolution waveguide sensing. J Colloid Interface Sci 2025; 677:352-364. [PMID: 39151228 DOI: 10.1016/j.jcis.2024.07.236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
HYPOTHESIS Self-driven actions, like motion, are fundamental characteristics of life. Today, intense research focuses on the kinetics of droplet motion. Quantifying macroscopic motion and exploring the underlying mechanisms are crucial in self-structuring and self-healing materials, advancements in soft robotics, innovations in self-cleaning environmental processes, and progress within the pharmaceutical industry. Usually, the driving forces inducing macroscopic motion act at the molecular scale, making their real-time and high-resolution investigation challenging. Label-free surface sensitive measurements with high lateral resolution could in situ measure both molecular-scale interactions and microscopic motion. EXPERIMENTS We employ surface-sensitive label-free sensors to investigate the kinetic changes in a self-assembled monolayer of the trimethyl(octadecyl)azanium chloride surfactant on a substrate surface during the self-propelled motion of nitrobenzene droplets. The adsorption-desorption of the surfactant at various concentrations, its removal due to the moving organic droplet, and rebuilding mechanisms at droplet-visited areas are all investigated with excellent time, spatial, and surface mass density resolution. FINDINGS We discovered concentration dependent velocity fluctuations, estimated the adsorbed amount of surfactant molecules, and revealed multilayer coverage at high concentrations. The desorption rate of surfactant (18.4 s-1) during the microscopic motion of oil droplets was determined by in situ differentiating between droplet visited and non-visited areas.
Collapse
Affiliation(s)
- Eniko Farkas
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, 1121 Budapest, Hungary
| | - Kinga Dóra Kovács
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, 1121 Budapest, Hungary; Department of Biological Physics, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Inna Szekacs
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, 1121 Budapest, Hungary
| | - Beatrix Peter
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, 1121 Budapest, Hungary
| | - István Lagzi
- Department of Physics, Institute of Physics, Budapest University of Technology and Economics, Muegyetem rkp. 3, 1111 Budapest, Hungary; HUN-REN-BME Condensed Matter Physics Research Group, Budapest University of Technology and Economics, Muegyetem rkp. 3, 1111 Budapest, Hungary
| | - Hiroyuki Kitahata
- Graduate School of Science, Chiba University, Yayoi-cho 1-33, Inage-ku, Chiba 263-8522, Japan
| | - Nobuhiko J Suematsu
- Meiji Institute of Advanced Study of Mathematical Sciences (MIMS), Meiji University, 4-21-1 Nakano, Tokyo 164-8525, Japan; Graduate School of Advanced Mathematical Sciences, Meiji University, 4-21-1 Nakano, Tokyo 164-8525, Japan.
| | - Robert Horvath
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, 1121 Budapest, Hungary; Nanobiosensorics Laboratory, Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary.
| |
Collapse
|
2
|
Beres B, Kovacs KD, Kanyo N, Peter B, Szekacs I, Horvath R. Label-Free Single-Cell Cancer Classification from the Spatial Distribution of Adhesion Contact Kinetics. ACS Sens 2024; 9:5815-5827. [PMID: 39082162 PMCID: PMC11590093 DOI: 10.1021/acssensors.4c01139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/03/2024]
Abstract
There is an increasing need for simple-to-use, noninvasive, and rapid tools to identify and separate various cell types or subtypes at the single-cell level with sufficient throughput. Often, the selection of cells based on their direct biological activity would be advantageous. These steps are critical in immune therapy, regenerative medicine, cancer diagnostics, and effective treatment. Today, live cell selection procedures incorporate some kind of biomolecular labeling or other invasive measures, which may impact cellular functionality or cause damage to the cells. In this study, we first introduce a highly accurate single-cell segmentation methodology by combining the high spatial resolution of a phase-contrast microscope with the adhesion kinetic recording capability of a resonant waveguide grating (RWG) biosensor. We present a classification workflow that incorporates the semiautomatic separation and classification of single cells from the measurement data captured by an RWG-based biosensor for adhesion kinetics data and a phase-contrast microscope for highly accurate spatial resolution. The methodology was tested with one healthy and six cancer cell types recorded with two functionalized coatings. The data set contains over 5000 single-cell samples for each surface and over 12,000 samples in total. We compare and evaluate the classification using these two types of surfaces (fibronectin and noncoated) with different segmentation strategies and measurement timespans applied to our classifiers. The overall classification performance reached nearly 95% with the best models showing that our proof-of-concept methodology could be adapted for real-life automatic diagnostics use cases. The label-free measurement technique has no impact on cellular functionality, directly measures cellular activity, and can be easily tuned to a specific application by varying the sensor coating. These features make it suitable for applications requiring further processing of selected cells.
Collapse
Affiliation(s)
- Balint Beres
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
- Department
of Automation and Applied Informatics, Faculty of Electrical Engineering
and Informatics, Budapest University of
Technology and Economics, Műegyetem rkp. 3, Budapest H-1111, Hungary
| | - Kinga Dora Kovacs
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
- Department
of Biological Physics, Eötvös
University, Pázmány Péter stny. 1/A, Budapest H-1117, Hungary
| | - Nicolett Kanyo
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
| | - Beatrix Peter
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
| | - Inna Szekacs
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
| | - Robert Horvath
- Nanobiosensorics
Laboratory, Institute of Technical Physics and Materials Science, HUN-REN Centre for Energy Research, Konkoly-Thege út 29-33, Budapest H-1121, Hungary
| |
Collapse
|
3
|
Gin A, Nguyen PD, Melzer JE, Li C, Strzelinski H, Liggett SB, Su J. Label-free, real-time monitoring of membrane binding events at zeptomolar concentrations using frequency-locked optical microresonators. Nat Commun 2024; 15:7445. [PMID: 39198447 PMCID: PMC11358326 DOI: 10.1038/s41467-024-51320-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
G-protein coupled receptors help regulate cellular function and communication, and are targets of small molecule drug discovery efforts. Conventional techniques to probe these interactions require labels and large amounts of receptor to achieve satisfactory sensitivity. Here, we use frequency-locked optical microtoroids for label-free characterization of membrane interactions in vitro at zeptomolar concentrations for the kappa opioid receptor and its native agonist dynorphin A 1-13, as well as big dynorphin (dynorphin A and dynorphin B) using a supported biomimetic membrane. The measured affinity of the agonist dynorphin A 1-13 to the κ-opioid receptor was also measured and found to be 3.1 nM. Radioligand assays revealed a dissociation constant in agreement with this value (1.1 nM). The limit of detection for the κOR/DynA 1-13 was calculated as 180 zM. The binding of Cholera Toxin B-monosialotetrahexosyl ganglioside was also monitored in real-time and an equilibrium dissociation constant of 1.53 nM was found. Our biosensing platform provides a method for highly sensitive real-time characterization of membrane embedded protein binding kinetics that is rapid and label-free, for drug discovery and toxin screening among other applications.
Collapse
Affiliation(s)
- Adley Gin
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, 85721, USA
| | - Phuong-Diem Nguyen
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jeffrey E Melzer
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, 85721, USA
| | - Cheng Li
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, 85721, USA
| | - Hannah Strzelinski
- Department of Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Stephen B Liggett
- Department of Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Judith Su
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, 85721, USA.
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
4
|
Arman S, Tilley RD, Gooding JJ. A review of electrochemical impedance as a tool for examining cell biology and subcellular mechanisms: merits, limits, and future prospects. Analyst 2024; 149:269-289. [PMID: 38015145 DOI: 10.1039/d3an01423a] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Herein the development of cellular impedance biosensors, electrochemical impedance spectroscopy, and the general principles and terms associated with the cell-electrode interface is reviewed. This family of techniques provides quantitative and sensitive information into cell responses to stimuli in real-time with high temporal resolution. The applications of cell-based impedance biosensors as a readout in cell biology is illustrated with a diverse range of examples. The current state of the field, its limitations, the possible available solutions, and the potential benefits of developing biosensors are discussed.
Collapse
Affiliation(s)
- Seyedyousef Arman
- School of Chemistry, The University of New South Wales, Sydney, New South Wales 2052, Australia.
- Australia Centre for Nanomedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Richard D Tilley
- School of Chemistry, The University of New South Wales, Sydney, New South Wales 2052, Australia.
- Electron Microscope Unit, Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - J Justin Gooding
- School of Chemistry, The University of New South Wales, Sydney, New South Wales 2052, Australia.
- Australia Centre for Nanomedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
5
|
Joseph S, Rajpal S, Kar D, Devinder S, Pandey S, Mishra P, Joseph J. Guided mode resonance immunosensor for label-free detection of pathogenic bacteria Pseudomonas aeruginosa. Biosens Bioelectron 2023; 241:115695. [PMID: 37776624 DOI: 10.1016/j.bios.2023.115695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/10/2023] [Accepted: 09/15/2023] [Indexed: 10/02/2023]
Abstract
Photonic biosensors are promising platforms for the rapid detection of pathogens with the potential to replace conventional diagnostics based on microbiological culturing methods. Intricately designed sensing elements with robust architectures can offer highly sensitive detection at minimal development cost enabling rapid adoption in low-resource settings. In this work, an optical detection scheme is developed by structuring guided mode resonance (GMR) on a highly stable, transparent silicon nitride (SiN) substrate and further biofunctionalized to identify a specific bacteria Pseudomonas aeruginosa. The resonance condition of the GMR chip is optimized to have relatively high bulk sensitivity with a good quality factor. The biofunctionalization aims at oriented immobilization of specific antibodies to allow maximum bacteria attachment and improved specificity. The sensitivity of the assays is evaluated for clinically relevant concentrations ranging from 102 to 108 CFU/mL. From the calibration curves, the sensitivity of the chip is extracted as 0.134nm/Log10 [concentration], and the detection modality possesses a favorably good limit of detection (LOD) 89 CFU/mL. The use of antibodies as a biorecognition element complemented with a good figure of merit of GMR sensing element allows selective bacteria identification compared to other non-specific pathogenic bacteria that are relevant for testing physiological samples. Our developed GMR biosensor is low-cost, easy to handle, and readily transformable into a portable handheld detection modality for remote usage.
Collapse
Affiliation(s)
- Shereena Joseph
- Department of Physics, Indian Institute of Technology Delhi, New Delhi, India
| | - Soumya Rajpal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Debashree Kar
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Shital Devinder
- Centre for Sensors, Instruments and Cyber Physical System Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Saurabh Pandey
- Department of Physics, Indian Institute of Technology Delhi, New Delhi, India
| | - Prashant Mishra
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Joby Joseph
- Department of Physics, Indian Institute of Technology Delhi, New Delhi, India; Optics and Photonics Centre, Indian Institute of Technology Delhi, New Delhi, India.
| |
Collapse
|
6
|
Shih CW, Rodt S, Reitzenstein S. Universal design method for bright quantum light sources based on circular Bragg grating cavities. OPTICS EXPRESS 2023; 31:35552-35564. [PMID: 38017723 DOI: 10.1364/oe.501495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/26/2023] [Indexed: 11/30/2023]
Abstract
We theoretically develop an efficient and universal design scheme of quantum light sources based on hybrid circular Bragg grating (CBG) cavity with and without electrical contact bridges. As the proposed design scheme strongly alleviates the computational cost of numerical simulation, we present high-performance CBG designs based on the GaAs/SiO2/Au material system for emission wavelengths ranging from 900 nm to 1600 nm, covering the whole telecom O-band and C-band. All designs achieve remarkable Purcell factors surpassing a value of 26 and extraction efficiencies (into a numerical aperture of 0.8) exceeding 92% without contact bridges and 86% with contact bridges. Additionally, we show that our design approach easily deals with realistic structural constraints, such as preset thicknesses of a semiconductor membrane or SiO2 layers or with a different material system. The high design flexibility greatly supports the experimental deterministic fabrication approaches, allowing one to perform in-situ design adaptation and to integrate single quantum emitters of an inhomogeneously broadened ensemble on the same chip into wavelength-adapted structures without spectral constraints, which highly increase the yield of quantum device fabrication.
Collapse
|
7
|
Gin A, Nguyen PD, Melzer JE, Li C, Strzelinski H, Liggett SB, Su J. Label-free, real-time monitoring of membrane binding events at zeptomolar concentrations using frequency-locked optical microresonators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.20.558657. [PMID: 37786702 PMCID: PMC10541581 DOI: 10.1101/2023.09.20.558657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Binding events to elements of the cell membrane act as receptors which regulate cellular function and communication and are the targets of many small molecule drug discovery efforts for agonists and antagonists. Conventional techniques to probe these interactions generally require labels and large amounts of receptor to achieve satisfactory sensitivity. Whispering gallery mode microtoroid optical resonators have demonstrated sensitivity to detect single-molecule binding events. Here, we demonstrate the use of frequency-locked optical microtoroids for characterization of membrane interactions in vitro at zeptomolar concentrations using a supported biomimetic membrane. Arrays of microtoroids were produced using photolithography and subsequently modified with a biomimetic membrane, providing high quality (Q) factors (> 10 6 ) in aqueous environments. Fluorescent recovery after photobleaching (FRAP) experiments confirmed the retained fluidity of the microtoroid supported-lipid membrane with a diffusion coefficient of 3.38 ± 0.26 μm 2 ⋅ s - 1 . Utilizing this frequency-locked membrane-on-a-chip model combined with auto-balanced detection and non-linear post-processing techniques, we demonstrate zeptomolar detection levels The binding of Cholera Toxin B- monosialotetrahexosyl ganglioside (GM1) was monitored in real-time, with an apparent equilibrium dissociation constant k d = 1.53 nM . The measured affiny of the agonist dynorphin A 1-13 to the κ -opioid receptor revealed a k d = 3.1 nM using the same approach. Radioligand binding competition with dynorphin A 1-13 revealed a k d in agreement (1.1 nM) with the unlabeled method. The biosensing platform reported herein provides a highly sensitive real-time characterization of membrane embedded protein binding kinetics, that is rapid and label-free, for toxin screening and drug discovery, among other applications.
Collapse
Affiliation(s)
- Adley Gin
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ 85721
| | - Phuong-Diem Nguyen
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ 85721
| | - Jeffrey E. Melzer
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ 85721
| | - Cheng Li
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ 85721
| | - Hannah Strzelinski
- Department of Medicine, University of South Florida Morsani College of Medicine, Tampa, FL 33612
| | - Stephen B. Liggett
- Department of Medicine, University of South Florida Morsani College of Medicine, Tampa, FL 33612
| | - Judith Su
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ 85721
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ 85721
| |
Collapse
|
8
|
Bruhier H, Dutems J, Laffont E, Crespo-Monteiro N, Verrier I, Parriaux O, Berini P, Jourlin Y. Common-mode plasmon sensing scheme as a high-sensitivity compact SPR sensor. OPTICS LETTERS 2023; 48:3733-3736. [PMID: 37450737 DOI: 10.1364/ol.483692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 06/05/2023] [Indexed: 07/18/2023]
Abstract
A deep metal grating enables quasi-phase-matched simultaneous excitation of two counterpropagating surface plasmon modes by means of its +1st and -2nd diffraction orders. The resulting angular reflection spectra of the scattered -1st and zeroth orders exhibit three interleaved zeros and maxima in a range centered around the Littrow angle. The spectra differ thoroughly from the usual reflection dip resulting from single-order plasmon coupling that produces strong absorption. The zeroth and -1st orders exhibit two crossing angles enabling high-sensitivity common-mode detection schemes designed to reject variations in source power and environmental noise. The proof of concept and experimental assessment of this new surface plasmon resonance (SPR) sensing scheme are demonstrated by monitoring gases in a pressure-controlled chamber. A limit of detection (LOD) of 2 × 10-7 refractive index unit (RIU) was achieved.
Collapse
|
9
|
Shakurov R, Sizova S, Dudik S, Serkina A, Bazhutov M, Stanaityte V, Tulyagin P, Konopsky V, Alieva E, Sekatskii S, Bespyatykh J, Basmanov D. Dendrimer-Based Coatings on a Photonic Crystal Surface for Ultra-Sensitive Small Molecule Detection. Polymers (Basel) 2023; 15:2607. [PMID: 37376252 DOI: 10.3390/polym15122607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
We propose and demonstrate dendrimer-based coatings for a sensitive biochip surface that enhance the high-performance sorption of small molecules (i.e., biomolecules with low molecular weights) and the sensitivity of a label-free, real-time photonic crystal surface mode (PC SM) biosensor. Biomolecule sorption is detected by measuring changes in the parameters of optical modes on the surface of a photonic crystal (PC). We describe the step-by-step biochip fabrication process. Using oligonucleotides as small molecules and PC SM visualization in a microfluidic mode, we show that the PAMAM (poly-amidoamine)-modified chip's sorption efficiency is almost 14 times higher than that of the planar aminosilane layer and 5 times higher than the 3D epoxy-dextran matrix. The results obtained demonstrate a promising direction for further development of the dendrimer-based PC SM sensor method as an advanced label-free microfluidic tool for detecting biomolecule interactions. Current label-free methods for small biomolecule detection, such as surface plasmon resonance (SPR), have a detection limit down to pM. In this work, we achieved for a PC SM biosensor a Limit of Quantitation of up to 70 fM, which is comparable with the best label-using methods without their inherent disadvantages, such as changes in molecular activity caused by labeling.
Collapse
Affiliation(s)
- Ruslan Shakurov
- Lopukhin Federal Research and Clinical Center of Physical Chemical Medicine of Federal Medical Biological Agency, 1A Malaya Pirogovskaya Street, 119435 Moscow, Russia
- Research Institute for Systems Biology and Medicine (RISBM), Nauchniy Proezd 18, 117246 Moscow, Russia
| | - Svetlana Sizova
- Lopukhin Federal Research and Clinical Center of Physical Chemical Medicine of Federal Medical Biological Agency, 1A Malaya Pirogovskaya Street, 119435 Moscow, Russia
- Research Institute for Systems Biology and Medicine (RISBM), Nauchniy Proezd 18, 117246 Moscow, Russia
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry RAS, 16/10 Miklukho-Maklaya Street, 117997 Moscow, Russia
| | - Stepan Dudik
- Lopukhin Federal Research and Clinical Center of Physical Chemical Medicine of Federal Medical Biological Agency, 1A Malaya Pirogovskaya Street, 119435 Moscow, Russia
- Research Institute for Systems Biology and Medicine (RISBM), Nauchniy Proezd 18, 117246 Moscow, Russia
| | - Anna Serkina
- Lopukhin Federal Research and Clinical Center of Physical Chemical Medicine of Federal Medical Biological Agency, 1A Malaya Pirogovskaya Street, 119435 Moscow, Russia
| | - Mark Bazhutov
- Lopukhin Federal Research and Clinical Center of Physical Chemical Medicine of Federal Medical Biological Agency, 1A Malaya Pirogovskaya Street, 119435 Moscow, Russia
| | - Viktorija Stanaityte
- Lopukhin Federal Research and Clinical Center of Physical Chemical Medicine of Federal Medical Biological Agency, 1A Malaya Pirogovskaya Street, 119435 Moscow, Russia
| | - Petr Tulyagin
- Research Institute for Systems Biology and Medicine (RISBM), Nauchniy Proezd 18, 117246 Moscow, Russia
| | - Valery Konopsky
- Institute of Spectroscopy RAS, 5 Fizicheskaya Street, Troitsk, 108840 Moscow, Russia
| | - Elena Alieva
- Institute of Spectroscopy RAS, 5 Fizicheskaya Street, Troitsk, 108840 Moscow, Russia
| | - Sergey Sekatskii
- Laboratory of Biological Electron Microscopy, Institute of Physics (IPHYS), BSP 419, Ecole Polytechnique Fédérale de Lausanne, and Department of Fundamental Biology, Faculty of Biology and Medicine, University of Lausanne, CH1015 Lausanne, Switzerland
| | - Julia Bespyatykh
- Lopukhin Federal Research and Clinical Center of Physical Chemical Medicine of Federal Medical Biological Agency, 1A Malaya Pirogovskaya Street, 119435 Moscow, Russia
- Expertise Department in Anti-Doping and Drug Control, Mendeleev University of Chemical Technology of Russia, 9, Miusskaya Square, 125047 Moscow, Russia
- Institute of Physics and Technology, 9 Institutskiy Pereulok, 141701 Dolgoprudny, Russia
| | - Dmitry Basmanov
- Lopukhin Federal Research and Clinical Center of Physical Chemical Medicine of Federal Medical Biological Agency, 1A Malaya Pirogovskaya Street, 119435 Moscow, Russia
- Research Institute for Systems Biology and Medicine (RISBM), Nauchniy Proezd 18, 117246 Moscow, Russia
- Institute of Physics and Technology, 9 Institutskiy Pereulok, 141701 Dolgoprudny, Russia
| |
Collapse
|
10
|
Huang SH, Sartorello G, Shen PT, Xu C, Elemento O, Shvets G. Metasurface-enhanced infrared spectroscopy in multiwell format for real-time assaying of live cells. LAB ON A CHIP 2023; 23:2228-2240. [PMID: 37010356 PMCID: PMC10159923 DOI: 10.1039/d3lc00017f] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Fourier transform infrared (FTIR) spectroscopy is a popular technique for the analysis of biological samples, yet its application in characterizing live cells is limited due to the strong attenuation of mid-IR light in water. Special thin flow cells and attenuated total reflection (ATR) FTIR spectroscopy have been used to mitigate this problem, but these techniques are difficult to integrate into a standard cell culture workflow. In this work, we demonstrate that the use of a plasmonic metasurface fabricated on planar substrates and the probing of cellular IR spectra through metasurface-enhanced infrared spectroscopy (MEIRS) can be an effective technique to characterize the IR spectra of live cells in a high-throughput manner. Cells are cultured on metasurfaces integrated with multiwell cell culture chambers and are probed from the bottom using an inverted FTIR micro-spectrometer. To demonstrate the use of MEIRS as a cellular assay, cellular adhesion on metasurfaces with different surface coatings and cellular response to the activation of the protease-activated receptor (PAR) signaling pathway were characterized through the changes in cellular IR spectra.
Collapse
Affiliation(s)
- Steven H Huang
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York, 14853, USA.
| | - Giovanni Sartorello
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York, 14853, USA.
| | - Po-Ting Shen
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York, 14853, USA.
| | - Chengqi Xu
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, 10021, USA.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, 10021, USA.
| | - Gennady Shvets
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York, 14853, USA.
| |
Collapse
|
11
|
Masterson AN, Chowdhury NN, Fang Y, Yip-Schneider MT, Hati S, Gupta P, Cao S, Wu H, Schmidt CM, Fishel ML, Sardar R. Amplification-Free, High-Throughput Nanoplasmonic Quantification of Circulating MicroRNAs in Unprocessed Plasma Microsamples for Earlier Pancreatic Cancer Detection. ACS Sens 2023; 8:1085-1100. [PMID: 36853001 DOI: 10.1021/acssensors.2c02105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly malignancy that is often detected at an advanced stage. Earlier diagnosis of PDAC is key to reducing mortality. Circulating biomarkers such as microRNAs are gaining interest, but existing technologies require large sample volumes, amplification steps, extensive biofluid processing, lack sensitivity, and are low-throughput. Here, we present an advanced nanoplasmonic sensor for the highly sensitive, amplification-free detection and quantification of microRNAs (microRNA-10b, microRNA-let7a) from unprocessed plasma microsamples. The sensor construct utilizes uniquely designed -ssDNA receptors attached to gold triangular nanoprisms, which display unique localized surface plasmon resonance (LSPR) properties, in a multiwell plate format. The formation of -ssDNA/microRNA duplex controls the nanostructure-biomolecule interfacial electronic interactions to promote the charge transfer/exciton delocalization processes and enhance the LSPR responses to achieve attomolar (10-18 M) limit of detection (LOD) in human plasma. This improve LOD allows the fabrication of a high-throughput assay in a 384-well plate format. The performance of nanoplasmonic sensors for microRNA detection was further assessed by comparing with the qRT-PCR assay of 15 PDAC patient plasma samples that shows a positive correlation between these two assays with the Pearson correlation coefficient value >0.86. Evaluation of >170 clinical samples reveals that oncogenic microRNA-10b and tumor suppressor microRNA-let7a levels can individually differentiate PDAC from chronic pancreatitis and normal controls with >94% sensitivity and >94% specificity at a 95% confidence interval (CI). Furthermore, combining both oncogenic and tumor suppressor microRNA levels significantly improves differentiation of PDAC stages I and II versus III and IV with >91% and 87% sensitivity and specificity, respectively, in comparison to the sensitivity and specificity values for individual microRNAs. Moreover, we show that the level of microRNAs varies substantially in pre- and post-surgery PDAC patients (n = 75). Taken together, this ultrasensitive nanoplasmonic sensor with excellent sensitivity and specificity is capable of assaying multiple biomarkers simultaneously and may facilitate early detection of PDAC to improve patient care.
Collapse
Affiliation(s)
- Adrianna N Masterson
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, Indiana 46202, United States
| | - Nayela N Chowdhury
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana 46202, United States
| | - Yue Fang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Michele T Yip-Schneider
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Sumon Hati
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, Indiana 46202, United States
| | - Prashant Gupta
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri 63130, United States
| | - Sha Cao
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Huangbing Wu
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - C Max Schmidt
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana 46202, United States
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Melissa L Fishel
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana 46202, United States
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Rajesh Sardar
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, Indiana 46202, United States
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana 46202, United States
| |
Collapse
|
12
|
Khandekar C, Jin W, Fan S. Nanophotonic detector array to enable direct thermal infrared vision. OPTICS EXPRESS 2022; 30:39222-39233. [PMID: 36258467 DOI: 10.1364/oe.475296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
Detection of infrared (IR) photons in a room-temperature IR camera is carried out by a two-dimensional array of microbolometer pixels which exhibit temperature-sensitive resistivity. When IR light coming from the far-field is focused onto this array, microbolometer pixels are heated up in proportion to the temperatures of the far-field objects. The resulting resistivity change of each pixel is measured via on-chip electronic readout circuit followed by analog to digital (A/D) conversion, image processing, and presentation of the final IR image on a separate information display screen. In this work, we introduce a new nanophotonic detector as a minimalist alternative to microbolometer such that the final IR image can be presented without using the components required for A/D conversion, image processing and display. In our design, the detector array is illuminated with visible laser light and the reflected light itself carries the IR image which can be directly viewed. We numerically demonstrate this functionality using a resonant waveguide grating structure made of typical materials such as silicon carbide, silicon nitride, and silica for which lithography techniques are well-developed. We clarify the requirements to tackle the issues of fabrication nonuniformities and temperature drifts in the detector array. We envision a potential near-eye display device for direct IR vision based on timely use of diffractive optical waveguides in augmented reality headsets and tunable visible laser sources. Our work indicates a way to achieve thermal IR vision for suitable use cases with lower cost, smaller form factor, and reduced power consumption compared to the existing thermal IR cameras.
Collapse
|
13
|
Janik M, Sosnowska M, Gabler T, Koba M, Myśliwiec A, Kutwin M, Chwalibóg ES, Śmietana M. Life in an optical fiber: Monitoring of cell cultures with microcavity in-line Mach-Zehnder interferometer. Biosens Bioelectron 2022; 217:114718. [PMID: 36174357 DOI: 10.1016/j.bios.2022.114718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/23/2022] [Accepted: 09/10/2022] [Indexed: 11/02/2022]
Abstract
Monitoring cell adhesion and growth are crucial for various applications involving drug screening, cytotoxicity, and cytocompatibility studies. However, acquiring accurate information about the growing state and responsiveness to a treatment of a cell system in a real-time and label-free manner is still a challenge. This work presents the first research on direct, real-time, and label-free adherent cell culture monitoring using a microcavity in-line Mach-Zehnder interferometer (μIMZI) fabricated in an optical fiber. The sensing solution based on μIMZI offers a great advantage over many other monitoring concepts tracking the changes taking place on the microcavity's bottom surface and within its volume, thus offering a greater penetration depth. In this study, we verified performance of the approach using a non-cancer bone marrow stromal cell line HS-5. The results demonstrate that the changes of the acquired signal are closely related to the different states of cells' adhesion, proliferation, morphology, and variation of mass. Thus, this label-free, real-time μIMZI-based monitoring technique gives a great promise to the analysis or monitoring of relevant new treatments in future scientific, as well as clinical applications.
Collapse
Affiliation(s)
- Monika Janik
- Warsaw University of Technology, Institute of Microelectronics and Optoelectronics,Koszykowa 75, 00-662, Warsaw, Poland.
| | - Malwina Sosnowska
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Lzife Sciences, Ciszewskiego 8, 02-786, Warsaw, Poland
| | - Tomasz Gabler
- Warsaw University of Technology, Institute of Microelectronics and Optoelectronics,Koszykowa 75, 00-662, Warsaw, Poland
| | - Marcin Koba
- Warsaw University of Technology, Institute of Microelectronics and Optoelectronics,Koszykowa 75, 00-662, Warsaw, Poland; National Institute of Telecommunications, Szachowa 1, 04-894, Warsaw, Poland
| | - Anna Myśliwiec
- Warsaw University of Technology, Institute of Microelectronics and Optoelectronics,Koszykowa 75, 00-662, Warsaw, Poland
| | - Marta Kutwin
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Lzife Sciences, Ciszewskiego 8, 02-786, Warsaw, Poland
| | - Ewa Sawosz Chwalibóg
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Lzife Sciences, Ciszewskiego 8, 02-786, Warsaw, Poland
| | - Mateusz Śmietana
- Warsaw University of Technology, Institute of Microelectronics and Optoelectronics,Koszykowa 75, 00-662, Warsaw, Poland
| |
Collapse
|
14
|
Seyedabadi M, Gharghabi M, Gurevich EV, Gurevich VV. Structural basis of GPCR coupling to distinct signal transducers: implications for biased signaling. Trends Biochem Sci 2022; 47:570-581. [PMID: 35396120 PMCID: PMC9189013 DOI: 10.1016/j.tibs.2022.03.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/10/2022] [Accepted: 03/11/2022] [Indexed: 02/06/2023]
Abstract
Three classes of G-protein-coupled receptor (GPCR) partners - G proteins, GPCR kinases, and arrestins - preferentially bind active GPCRs. Our analysis suggests that the structures of GPCRs bound to these interaction partners available today do not reveal a clear conformational basis for signaling bias, which would have enabled the rational design of biased GRCR ligands. In view of this, three possibilities are conceivable: (i) there are no generalizable GPCR conformations conducive to binding a particular type of partner; (ii) subtle differences in the orientation of individual residues and/or their interactions not easily detectable in the receptor-transducer structures determine partner preference; or (iii) the dynamics of GPCR binding to different types of partners rather than the structures of the final complexes might underlie transducer bias.
Collapse
Affiliation(s)
- Mohammad Seyedabadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehdi Gharghabi
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | | |
Collapse
|
15
|
Szittner Z, Péter B, Kurunczi S, Székács I, Horváth R. Functional blood cell analysis by label-free biosensors and single-cell technologies. Adv Colloid Interface Sci 2022; 308:102727. [DOI: 10.1016/j.cis.2022.102727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/25/2022] [Accepted: 06/27/2022] [Indexed: 11/01/2022]
|
16
|
Investigating the ligand agonism and antagonism at the D 2long receptor by dynamic mass redistribution. Sci Rep 2022; 12:9637. [PMID: 35688965 PMCID: PMC9187652 DOI: 10.1038/s41598-022-14311-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/06/2022] [Indexed: 11/09/2022] Open
Abstract
The signalling of the D2 receptor (D2R), a G protein-coupled receptor (GPCR), is a complex process consisting of various components. For the screening of D2R ligands, methods quantifying distinct second messengers such as cAMP or the interaction of the receptor with β-arrestin, are commonly employed. In contrast, a label-free biosensor technology like dynamic mass redistribution (DMR), where it is mostly unknown how the individual signalling pathways contribute to the DMR signal, provides a holistic readout of the complex cellular response. In this study, we report the successful application of the DMR technology to CHO-K1 cells stably expressing the human dopamine D2long receptor. In real-time kinetic experiments, studies of D2R reference compounds yielded results for agonists and antagonists that were consistent with those obtained by conventional methods and also allowed a discrimination between partial and full agonists. Furthermore, investigations on the signalling pathway in CHO-K1 hD2longR cells identified the Gαi/o protein as the main proximal trigger of the observed DMR response. The present study has shown that the DMR technology is a valuable method for the characterisation of putative new ligands and, due to its label-free nature, suggests its use for deorphanisation studies of GPCRs.
Collapse
|
17
|
Kanyo N, Kovács K, Kovács S, Béres B, Peter B, Székács I, Horvath R. Single-cell adhesivity distribution of glycocalyx digested cancer cells from high spatial resolution label-free biosensor measurements. Matrix Biol Plus 2022; 14:100103. [PMID: 35243300 PMCID: PMC8857652 DOI: 10.1016/j.mbplus.2022.100103] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/26/2022] [Accepted: 01/30/2022] [Indexed: 12/01/2022] Open
Abstract
The glycocalyx is a cell surface sugar layer of most cell types that greatly influences the interaction of cells with their environment. Its components are glycolipids, glycoproteins, and oligosaccharides. Interestingly, cancer cells have a thicker glycocalyx layer compared to healthy cells, but to date, there has been no consensus in the literature on the exact role of cell surface polysaccharides and their derivatives in cellular adhesion and signaling. In our previous work we discovered that specific glycocalyx components of cancer cells regulate the kinetics and strength of adhesion on RGD (arginine-glycine-aspartic acid) peptide-coated surfaces [1]. Depending on the employed enzyme concentration digesting specific components both adhesion strengthening and weakening could be observed by monitoring the averaged behavior of thousands of cells. The enzyme chondroitinase ABC (ChrABC) was used to digest the chondroitin-4-sulfate, chondroitin-6-sulfate, and dermatan sulfate components in the glycocalyx of cancer cells. In the present work, a high spatial resolution label-free optical biosensor was employed to monitor the adhesivity of cancer cells both at the single-cell and population level. Population-level distributions of single-cell adhesivity were first recorded and analyzed when ChrABC was added to the adhering cells. At relatively low and high ChrABC concentrations subpopulations with remarkably large and weak adhesivity were identified. The changes in the adhesivity distribution due to the enzyme treatment were analyzed and the subpopulations most affected by the enzyme treatment were highlighted. The presented results open up new directions in glycocalyx related cell adhesion research and in the development of more meaningful targeted cancer treatments affecting adhesion.
Collapse
Affiliation(s)
- N. Kanyo
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary
| | - K.D. Kovács
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary
- ELTE Eötvös Loránd University, Department of Biological Physics, Budapest, Hungary
| | - S.V. Kovács
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary
| | - B. Béres
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary
| | - B. Peter
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary
| | - I. Székács
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary
| | - R. Horvath
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary
| |
Collapse
|
18
|
Shen PT, Huang SH, Huang Z, Wilson JJ, Shvets G. Probing the Drug Dynamics of Chemotherapeutics Using Metasurface-Enhanced Infrared Reflection Spectroscopy of Live Cells. Cells 2022; 11:1600. [PMID: 35626636 PMCID: PMC9139550 DOI: 10.3390/cells11101600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 11/25/2022] Open
Abstract
Infrared spectroscopy has drawn considerable interest in biological applications, but the measurement of live cells is impeded by the attenuation of infrared light in water. Metasurface-enhanced infrared reflection spectroscopy (MEIRS) had been shown to mitigate the problem, enhance the cellular infrared signal through surface-enhanced infrared absorption, and encode the cellular vibrational signatures in the reflectance spectrum at the same time. In this study, we used MEIRS to study the dynamic response of live cancer cells to a newly developed chemotherapeutic metal complex with distinct modes of action (MoAs): tricarbonyl rhenium isonitrile polypyridyl (TRIP). MEIRS measurements demonstrated that administering TRIP resulted in long-term (several hours) reduction in protein, lipid, and overall refractive index signals, and in short-term (tens of minutes) increase in these signals, consistent with the induction of endoplasmic reticulum stress. The unique tricarbonyl IR signature of TRIP in the bioorthogonal spectral window was monitored in real time, and was used as an infrared tag to detect the precise drug delivery time that was shown to be closely correlated with the onset of the phenotypic response. These results demonstrate that MEIRS is an effective label-free real-time cellular assay capable of detecting and interpreting the early phenotypic responses of cells to IR-tagged chemotherapeutics.
Collapse
Affiliation(s)
- Po-Ting Shen
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853, USA; (P.-T.S.); (S.H.H.)
| | - Steven H. Huang
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853, USA; (P.-T.S.); (S.H.H.)
| | - Zhouyang Huang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA; (Z.H.); (J.J.W.)
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA; (Z.H.); (J.J.W.)
| | - Gennady Shvets
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853, USA; (P.-T.S.); (S.H.H.)
| |
Collapse
|
19
|
Kavand H, Nasiri R, Herland A. Advanced Materials and Sensors for Microphysiological Systems: Focus on Electronic and Electrooptical Interfaces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107876. [PMID: 34913206 DOI: 10.1002/adma.202107876] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/07/2021] [Indexed: 06/14/2023]
Abstract
Advanced in vitro cell culture systems or microphysiological systems (MPSs), including microfluidic organ-on-a-chip (OoC), are breakthrough technologies in biomedicine. These systems recapitulate features of human tissues outside of the body. They are increasingly being used to study the functionality of different organs for applications such as drug evolutions, disease modeling, and precision medicine. Currently, developers and endpoint users of these in vitro models promote how they can replace animal models or even be a better ethically neutral and humanized alternative to study pathology, physiology, and pharmacology. Although reported models show a remarkable physiological structure and function compared to the conventional 2D cell culture, they are almost exclusively based on standard passive polymers or glass with none or minimal real-time stimuli and readout capacity. The next technology leap in reproducing in vivo-like functionality and real-time monitoring of tissue function could be realized with advanced functional materials and devices. This review describes the currently reported electronic and optical advanced materials for sensing and stimulation of MPS models. In addition, an overview of multi-sensing for Body-on-Chip platforms is given. Finally, one gives the perspective on how advanced functional materials could be integrated into in vitro systems to precisely mimic human physiology.
Collapse
Affiliation(s)
- Hanie Kavand
- Division of Micro- and Nanosystems, Department of Intelligent Systems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
| | - Rohollah Nasiri
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solnavägen 9/B8, Solna, 171 65, Sweden
- Division of Nanobiotechnology, Department of Protein Science, KTH Royal Institute of Technology, Tomtebodavägen 23a, Solna, 171 65, Sweden
| | - Anna Herland
- Division of Micro- and Nanosystems, Department of Intelligent Systems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solnavägen 9/B8, Solna, 171 65, Sweden
- Division of Nanobiotechnology, Department of Protein Science, KTH Royal Institute of Technology, Tomtebodavägen 23a, Solna, 171 65, Sweden
| |
Collapse
|
20
|
Servant NB, Williams ME, Brust PF, Tang H, Wong MS, Chen Q, Lebl-Rinnova M, Adamski-Werner SL, Tachdjian C, Servant G. A Dynamic Mass Redistribution Assay for the Human Sweet Taste Receptor Uncovers G-Protein Dependent Biased Ligands. Front Pharmacol 2022; 13:832529. [PMID: 35250580 PMCID: PMC8893300 DOI: 10.3389/fphar.2022.832529] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/24/2022] [Indexed: 12/26/2022] Open
Abstract
The sweet taste receptor is rather unique, recognizing a diverse repertoire of natural or synthetic ligands, with a surprisingly large structural diversity, and with potencies stretching over more than six orders of magnitude. Yet, it is not clear if different cell-based assays can faithfully report the relative potencies and efficacies of these molecules. Indeed, up to now, sweet taste receptor agonists have been almost exclusively characterized using cell-based assays developed with overexpressed and promiscuous G proteins. This non-physiological coupling has allowed the quantification of receptor activity via phospholipase C activation and calcium mobilization measurements in heterologous cells on a FLIPR system, for example. Here, we developed a novel assay for the human sweet taste receptor where endogenous G proteins and signaling pathways are recruited by the activated receptor. The effects of several sweet taste receptor agonists and other types of modulators were recorded by measuring changes in dynamic mass redistribution (DMR) using an Epic® reader. Potency and efficacy values obtained in the DMR assay were compared to those results obtained with the classical FLIPR assay. Results demonstrate that for some ligands, the two assay systems provide similar information. However, a clear bias for the FLIPR assay was observed for one third of the agonists evaluated, suggesting that the use of non-physiological coupling may influence the potency and efficacy of sweet taste receptor ligands. Replacing the promiscuous G protein with a chimeric G protein containing the C-terminal tail 25 residues of the physiologically relevant G protein subunit Gαgustducin reduced or abrogated bias.
Collapse
|
21
|
Zarrintaj P, Saeb MR, Stadler FJ, Yazdi MK, Nezhad MN, Mohebbi S, Seidi F, Ganjali MR, Mozafari M. Human Organs-on-Chips: A Review of the State-of-the-Art, Current Prospects, and Future Challenges. Adv Biol (Weinh) 2022; 6:e2000526. [PMID: 34837667 DOI: 10.1002/adbi.202000526] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 08/03/2021] [Indexed: 01/09/2023]
Abstract
New emerging technologies, remarkably miniaturized 3D organ models and microfluidics, enable simulation of the real in vitro microenvironment ex vivo more closely. There are many fascinating features of innovative organ-on-a-chip (OOC) technology, including the possibility of integrating semipermeable and/or stretchable membranes, creating continuous perfusion of fluids into microchannels and chambers (while maintaining laminar flow regime), embedding microdevices like microsensors, microstimulators, micro heaters, or different cell lines, along with other 3D cell culture technologies. OOC systems are designed to imitate the structure and function of human organs, ranging from breathing lungs to beating hearts. This technology is expected to be able to revolutionize cell biology studies, personalized precision medicine, drug development process, and cancer diagnosis/treatment. OOC systems can significantly reduce the cost associated with tedious drug development processes and the risk of adverse drug reactions in the body, which makes drug screening more effective. The review mainly focus on presenting an overview of the several previously developed OOC systems accompanied by subjects relevant to pharmacy-, cancer-, and placenta-on-a-chip. The challenging issues and opportunities related to these systems are discussed, along with a future perspective for this technology.
Collapse
Affiliation(s)
- Payam Zarrintaj
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK, 74078, USA
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, G. Narutowicza 11/12, Gdańsk, 80-233, Poland
| | - Florian J Stadler
- College of Materials Science and Engineering, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, Shenzhen University, Shenzhen, 518060, China
| | - Mohsen Khodadadi Yazdi
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran, 1417466191, Iran
| | - Mojtaba Nasiri Nezhad
- Department of Chemical Engineering, Urmia University of Technology, Urmia, 57166-419, Iran
| | - Shabnam Mohebbi
- Department of Chemical Engineering, Tabriz University, Tabriz, 51335-1996, Iran
| | - Farzad Seidi
- Joint International Research Lab of Lignocellulosic Functional Materials, Nanjing Forestry University, Nanjing, 210037, China
| | - Mohammad Reza Ganjali
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran, 1417466191, Iran
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, 14395-1179, Iran
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| |
Collapse
|
22
|
Hauser AS. Personalized Medicine Through GPCR Pharmacogenomics. COMPREHENSIVE PHARMACOLOGY 2022:191-219. [DOI: 10.1016/b978-0-12-820472-6.00100-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
23
|
Yeh CT, Barshilia D, Hsieh CJ, Li HY, Hsieh WH, Chang GE. Rapid and Highly Sensitive Detection of C-Reaction Protein Using Robust Self-Compensated Guided-Mode Resonance BioSensing System for Point-of-Care Applications. BIOSENSORS 2021; 11:523. [PMID: 34940280 PMCID: PMC8699450 DOI: 10.3390/bios11120523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 05/24/2023]
Abstract
The rapid and sensitive detection of human C-reactive protein (CRP) in a point-of-care (POC) may be conducive to the early diagnosis of various diseases. Biosensors have emerged as a new technology for rapid and accurate detection of CRP for POC applications. Here, we propose a rapid and highly stable guided-mode resonance (GMR) optofluidic biosensing system based on intensity detection with self-compensation, which substantially reduces the instability caused by environmental factors for a long detection time. In addition, a low-cost LED serving as the light source and a photodetector are used for intensity detection and real-time biosensing, and the system compactness facilitates POC applications. Self-compensation relies on a polarizing beam splitter to separate the transverse-magnetic-polarized light and transverse-electric-polarized light from the light source. The transverse-electric-polarized light is used as a background signal for compensating noise, while the transverse-magnetic-polarized light is used as the light source for the GMR biosensor. After compensation, noise is drastically reduced, and both the stability and performance of the system are enhanced over a long period. Refractive index experiments revealed a resolution improvement by 181% when using the proposed system with compensation. In addition, the system was successfully applied to CRP detection, and an outstanding limit of detection of 1.95 × 10-8 g/mL was achieved, validating the proposed measurement system for biochemical reaction detection. The proposed GMR biosensing sensing system can provide a low-cost, compact, rapid, sensitive, and highly stable solution for a variety of point-of-care applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Guo-En Chang
- Department of Mechanical Engineering, Advanced Institute of Manufacturing with High-Tech Innovations (AIM-HI), National Chung Cheng University, Minxiong Township 62102, Taiwan; (C.-T.Y.); (D.B.); (C.-J.H.); (H.-Y.L.); (W.-H.H.)
| |
Collapse
|
24
|
Huang SH, Li J, Fan Z, Delgado R, Shvets G. Monitoring the effects of chemical stimuli on live cells with metasurface-enhanced infrared reflection spectroscopy. LAB ON A CHIP 2021; 21:3991-4004. [PMID: 34474459 PMCID: PMC8511245 DOI: 10.1039/d1lc00580d] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Infrared spectroscopy has found wide applications in the analysis of biological materials. A more recent development is the use of engineered nanostructures - plasmonic metasurfaces - as substrates for metasurface-enhanced infrared reflection spectroscopy (MEIRS). Here, we demonstrate that strong field enhancement from plasmonic metasurfaces enables the use of MEIRS as a highly informative analytic technique for real-time monitoring of cells. By exposing live cells cultured on a plasmonic metasurface to chemical compounds, we show that MEIRS can be used as a label-free phenotypic assay for detecting multiple cellular responses to external stimuli: changes in cell morphology, adhesion, and lipid composition of the cellular membrane, as well as intracellular signaling. Using a focal plane array detection system, we show that MEIRS also enables spectro-chemical imaging at the single-cell level. The described metasurface-based all-optical sensor opens the way to a scalable, high-throughput spectroscopic assay for live cells.
Collapse
Affiliation(s)
- Steven H Huang
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York, 14853 USA.
| | - Jiaruo Li
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York, 14853 USA.
| | - Zhiyuan Fan
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York, 14853 USA.
| | - Robert Delgado
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York, 14853 USA.
| | - Gennady Shvets
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York, 14853 USA.
| |
Collapse
|
25
|
Tirumala MG, Anchi P, Raja S, Rachamalla M, Godugu C. Novel Methods and Approaches for Safety Evaluation of Nanoparticle Formulations: A Focus Towards In Vitro Models and Adverse Outcome Pathways. Front Pharmacol 2021; 12:612659. [PMID: 34566630 PMCID: PMC8458898 DOI: 10.3389/fphar.2021.612659] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 08/05/2021] [Indexed: 12/18/2022] Open
Abstract
Nanotoxicology is an emerging field employed in the assessment of unintentional hazardous effects produced by nanoparticles (NPs) impacting human health and the environment. The nanotoxicity affects the range between induction of cellular stress and cytotoxicity. The reasons so far reported for these toxicological effects are due to their variable sizes with high surface areas, shape, charge, and physicochemical properties, which upon interaction with the biological components may influence their functioning and result in adverse outcomes (AO). Thus, understanding the risk produced by these materials now is an important safety concern for the development of nanotechnology and nanomedicine. Since the time nanotoxicology has evolved, the methods employed have been majorly relied on in vitro cell-based evaluations, while these simple methods may not predict the complexity involved in preclinical and clinical conditions concerning pharmacokinetics, organ toxicity, and toxicities evidenced through multiple cellular levels. The safety profiles of nanoscale nanomaterials and nanoformulations in the delivery of drugs and therapeutic applications are of considerable concern. In addition, the safety assessment for new nanomedicine formulas lacks regulatory standards. Though the in vivo studies are greatly needed, the end parameters used for risk assessment are not predicting the possible toxic effects produced by various nanoformulations. On the other side, due to increased restrictions on animal usage and demand for the need for high-throughput assays, there is a need for developing and exploring novel methods to evaluate NPs safety concerns. The progress made in molecular biology and the availability of several modern techniques may offer novel and innovative methods to evaluate the toxicological behavior of different NPs by using single cells, cell population, and whole organisms. This review highlights the recent novel methods developed for the evaluation of the safety impacts of NPs and attempts to solve the problems that come with risk assessment. The relevance of investigating adverse outcome pathways (AOPs) in nanotoxicology has been stressed in particular.
Collapse
Affiliation(s)
- Mounika Gayathri Tirumala
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pratibha Anchi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Susmitha Raja
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
26
|
Single-cell adhesion strength and contact density drops in the M phase of cancer cells. Sci Rep 2021; 11:18500. [PMID: 34531409 PMCID: PMC8445979 DOI: 10.1038/s41598-021-97734-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 08/27/2021] [Indexed: 02/08/2023] Open
Abstract
The high throughput, cost effective and sensitive quantification of cell adhesion strength at the single-cell level is still a challenging task. The adhesion force between tissue cells and their environment is crucial in all multicellular organisms. Integrins transmit force between the intracellular cytoskeleton and the extracellular matrix. This force is not only a mechanical interaction but a way of signal transduction as well. For instance, adhesion-dependent cells switch to an apoptotic mode in the lack of adhesion forces. Adhesion of tumor cells is a potential therapeutic target, as it is actively modulated during tissue invasion and cell release to the bloodstream resulting in metastasis. We investigated the integrin-mediated adhesion between cancer cells and their RGD (Arg-Gly-Asp) motif displaying biomimetic substratum using the HeLa cell line transfected by the Fucci fluorescent cell cycle reporter construct. We employed a computer-controlled micropipette and a high spatial resolution label-free resonant waveguide grating-based optical sensor calibrated to adhesion force and energy at the single-cell level. We found that the overall adhesion strength of single cancer cells is approximately constant in all phases except the mitotic (M) phase with a significantly lower adhesion. Single-cell evanescent field based biosensor measurements revealed that at the mitotic phase the cell material mass per unit area inside the cell-substratum contact zone is significantly less, too. Importantly, the weaker mitotic adhesion is not simply a direct consequence of the measured smaller contact area. Our results highlight these differences in the mitotic reticular adhesions and confirm that cell adhesion is a promising target of selective cancer drugs as the vast majority of normal, differentiated tissue cells do not enter the M phase and do not divide.
Collapse
|
27
|
Saftics A, Kurunczi S, Peter B, Szekacs I, Ramsden JJ, Horvath R. Data evaluation for surface-sensitive label-free methods to obtain real-time kinetic and structural information of thin films: A practical review with related software packages. Adv Colloid Interface Sci 2021; 294:102431. [PMID: 34330074 DOI: 10.1016/j.cis.2021.102431] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 02/07/2023]
Abstract
Interfacial layers are important in a wide range of applications in biomedicine, biosensing, analytical chemistry and the maritime industries. Given the growing number of applications, analysis of such layers and understanding their behavior is becoming crucial. Label-free surface sensitive methods are excellent for monitoring the formation kinetics, structure and its evolution of thin layers, even at the nanoscale. In this paper, we review existing and commercially available label-free techniques and demonstrate how the experimentally obtained data can be utilized to extract kinetic and structural information during and after formation, and any subsequent adsorption/desorption processes. We outline techniques, some traditional and some novel, based on the principles of optical and mechanical transduction. Our special focus is the current possibilities of combining label-free methods, which is a powerful approach to extend the range of detected and deduced parameters. We summarize the most important theoretical considerations for obtaining reliable information from measurements taking place in liquid environments and, hence, with layers in a hydrated state. A thorough treamtmaent of the various kinetic and structural quantities obtained from evaluation of the raw label-free data are provided. Such quantities include layer thickness, refractive index, optical anisotropy (and molecular orientation derived therefrom), degree of hydration, viscoelasticity, as well as association and dissociation rate constants and occupied area of subsequently adsorbed species. To demonstrate the effect of variations in model conditions on the observed data, simulations of kinetic curves at various model settings are also included. Based on our own extensive experience with optical waveguide lightmode spectroscopy (OWLS) and the quartz crystal microbalance (QCM), we have developed dedicated software packages for data analysis, which are made available to the scientific community alongside this paper.
Collapse
|
28
|
Kliment K, Szekacs I, Peter B, Erdei A, Kurucz I, Horvath R. Label-free real-time monitoring of the BCR-triggered activation of primary human B cells modulated by the simultaneous engagement of inhibitory receptors. Biosens Bioelectron 2021; 191:113469. [PMID: 34229298 DOI: 10.1016/j.bios.2021.113469] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/26/2022]
Abstract
Today, there is an intense demand for lab-on-a-chip and tissue-on-a-chip applications in basic cell biological research and medical diagnostics. A particular challenge is the implementation of advanced biosensor techniques in point-of-care testing utilizing human primary cells. In this study, a resonant waveguide grating (RWG)-based label-free optical biosensor technique has been applied for real-time monitoring of the integrated responses of primary human tonsillar B cells initiated by B cell receptor (BCR) and modified by FcγRIIb and CR1 engagement. The BCR-triggered biosensor responses of resting and activated B cells were revealed to be specific and dose-dependent, in some cases with strong donor dependency. Targeted inhibition of Syk attenuated the label-free biosensor response upon BCR stimulation. Indifferent protein human serum albumin (HSA) did not interfere with the recorded signal to BCR stimulation. Simultaneous engagement of BCR and FcγRIIb modulated the kinetic signal of the cells. Activated and resting B cells exhibited different response profiles upon simultaneous engagement of BCR and CR1. This advanced approach has the potential to decipher interfering signaling events in human B cells, manage differences between activated and resting B cell states, helping to understand the actual integrated response of these immune cells, and could be useful in the point-of-care diagnostic testing on human primary cells.
Collapse
Affiliation(s)
- Kristof Kliment
- Department of Immunology, Eotvos Lorand University, Budapest, Hungary; Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, 29-33 Konkoly-Thege Miklós út, Budapest, Hungary
| | - Inna Szekacs
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, 29-33 Konkoly-Thege Miklós út, Budapest, Hungary.
| | - Beatrix Peter
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, 29-33 Konkoly-Thege Miklós út, Budapest, Hungary
| | - Anna Erdei
- Department of Immunology, Eotvos Lorand University, Budapest, Hungary; MTA-ELTE Immunology Research Group, Eotvos Lorand University, Budapest, Hungary
| | - Istvan Kurucz
- MTA-ELTE Immunology Research Group, Eotvos Lorand University, Budapest, Hungary
| | - Robert Horvath
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, 29-33 Konkoly-Thege Miklós út, Budapest, Hungary
| |
Collapse
|
29
|
Kovacs B, Kraft FA, Szabo Z, Nazirizadeh Y, Gerken M, Horvath R. Near cut-off wavelength operation of resonant waveguide grating biosensors. Sci Rep 2021; 11:13091. [PMID: 34158570 PMCID: PMC8219702 DOI: 10.1038/s41598-021-92327-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/07/2021] [Indexed: 02/05/2023] Open
Abstract
Numerical simulations and analytical calculations are performed to support the design of grating-coupled planar optical waveguides for biological sensing. Near cut-off and far from cut-off modes are investigated, and their characteristics and suitability for sensing are compared. The numerical simulations reveal the high sensitivity of the guided mode intensity near the cut-off wavelength for any refractive index change along the waveguide. Consequently, it is sufficient to monitor the intensity change of the near cut-off sensing mode, which leads to a simpler sensor design compared to those setups where the resonant wavelength shift of the guided mode is monitored with high precision. The operating wavelength and the sensitivity of the proposed device can be tuned by varying the geometrical parameters of the corrugated waveguide. These results may lead to the development of highly sensitive integrated sensors, which have a simple design and therefore are cost-effective for a wide range of applications. These numerical findings are supported with experimental results, where the cut-off sensing mode was identified.
Collapse
Affiliation(s)
- Balint Kovacs
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary.
- Division of Medical Image Computing (MIC), German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany.
| | - Fabio Aldo Kraft
- Institute of Electrical Engineering and Information Technology, Kiel University, Kiel, Germany
| | - Zsolt Szabo
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | | | - Martina Gerken
- Institute of Electrical Engineering and Information Technology, Kiel University, Kiel, Germany
| | - Robert Horvath
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary
| |
Collapse
|
30
|
Incaviglia I, Frutiger A, Blickenstorfer Y, Treindl F, Ammirati G, Lüchtefeld I, Dreier B, Plückthun A, Vörös J, Reichmuth AM. An Approach for the Real-Time Quantification of Cytosolic Protein-Protein Interactions in Living Cells. ACS Sens 2021; 6:1572-1582. [PMID: 33759497 DOI: 10.1021/acssensors.0c02480] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In recent years, cell-based assays have been frequently used in molecular interaction analysis. Cell-based assays complement traditional biochemical and biophysical methods, as they allow for molecular interaction analysis, mode of action studies, and even drug screening processes to be performed under physiologically relevant conditions. In most cellular assays, biomolecules are usually labeled to achieve specificity. In order to overcome some of the drawbacks associated with label-based assays, we have recently introduced "cell-based molography" as a biosensor for the analysis of specific molecular interactions involving native membrane receptors in living cells. Here, we expand this assay to cytosolic protein-protein interactions. First, we created a biomimetic membrane receptor by tethering one cytosolic interaction partner to the plasma membrane. The artificial construct is then coherently arranged into a two-dimensional pattern within the cytosol of living cells. Thanks to the molographic sensor, the specific interactions between the coherently arranged protein and its endogenous interaction partners become visible in real time without the use of a fluorescent label. This method turns out to be an important extension of cell-based molography because it expands the range of interactions that can be analyzed by molography to those in the cytosol of living cells.
Collapse
Affiliation(s)
- Ilaria Incaviglia
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Andreas Frutiger
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Yves Blickenstorfer
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Fridolin Treindl
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Giulia Ammirati
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Ines Lüchtefeld
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Birgit Dreier
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Janos Vörös
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Andreas M Reichmuth
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
31
|
Kovács KD, Novák M, Hajnal Z, Hős C, Szabó B, Székács I, Fang Y, Bonyár A, Horvath R. Label-free tracking of whole-cell response on RGD functionalized surfaces to varied flow velocities generated by fluidic rotation. J Colloid Interface Sci 2021; 599:620-630. [PMID: 33984760 DOI: 10.1016/j.jcis.2021.04.091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/11/2021] [Accepted: 04/18/2021] [Indexed: 10/21/2022]
Abstract
Fluidic flow plays important roles in colloid and interface sciences. Measuring adsorption, aggregation processes and living cell behavior under a fluidic environment with varied flow velocities in a parallel and high-throughput manner remains to be a challenging task. Here a method is introduced to monitor cell response to well-defined flow with varied velocities over an array of label-free resonant waveguide grating (RWG) based optical biosensors. The arrangement consists of a circular well with an array of biosensors at the bottom surface. By rotating the liquid over the biosensor array using a magnetic stirrer bar, flow velocities from zero to a predefined maximum can be easily established over different locations within the biosensor array as characterized in detail by numerical simulations. Cell adhesion and detachment measurements on an Arg-Gly-Asp (RGD) peptide functionalized surface were performed to demonstrate i) measurements at a wide range of simultaneous flow velocities over the same interface; ii) the possibility of parallel measurements at the same flow conditions in one run; and iii) the simple tuning of the employed range of flow velocities. Our setup made it possible to analyze the magnitude and rate of cell detachment at various flow velocities in parallel and determine the critical velocity and force where cells start to detach from the RGD motif displaying biomimetic surface. Furthermore, cellular response to simultaneous mechanical (flow) and chemical stimulation was also investigated using trypsin as a model. This study opens a new possibility to investigate interface phenomena under predefined and conveniently varied flow conditions.
Collapse
Affiliation(s)
- Kinga Dóra Kovács
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary; Department of Biological Physics, Eötvös University, Budapest, Hungary
| | - Martin Novák
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary
| | - Zoltán Hajnal
- Microsystems Laboratory, ELKH EK MFA, Budapest, Hungary
| | - Csaba Hős
- Department of Hydrodynamic Systems, Budapest University of Technology and Economics, Budapest, Hungary
| | - Bálint Szabó
- Department of Biological Physics, Eötvös University, Budapest, Hungary
| | - Inna Székács
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary
| | - Ye Fang
- EIG New Programs, Corning Research and Development Corporation, Corning Incorporated, NY, USA
| | - Attila Bonyár
- Department of Electronics Technology, Faculty of Electrical Engineering and Informatics, Budapest University of Technology and Economics, Budapest, Hungary.
| | - Robert Horvath
- Nanobiosensorics Laboratory, ELKH EK MFA, Budapest, Hungary.
| |
Collapse
|
32
|
Recent progress in surface plasmon resonance based sensors: A comprehensive review. Heliyon 2021; 7:e06321. [PMID: 33869818 PMCID: PMC8035490 DOI: 10.1016/j.heliyon.2021.e06321] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/21/2021] [Accepted: 02/16/2021] [Indexed: 12/17/2022] Open
Abstract
In the recent years, researchers have contributed substantially in the field of Surface Plasmon Resonance (SPR) sensors and its applications. SPR sensors show the salient features, such as label-free detection, real-time monitoring, small sample size, furnish accurate outcomes at low cost, and smooth handling. Moreover, the SPR sensors are also well-known because of its quantitative and qualitative excellent performance in real-time applications, including drug discovery, environment monitoring, food safety, medical diagnosis, clinical diagnosis, biological studies, and biomolecule interactions. This paper exhibits a comprehensive review of SPR based sensors, such as prism-based SPR with the applications (e.g., biomolecule interaction, medical diagnostic, etc.), fiber-based SPR, and waveguide-based SPR. Furthermore, we summarized the modern designs and techniques with their limitations and challenges in detail. The erudition outlined in this paper can be given an exceptional benefit for the researchers and industry people in the field of SPR based sensors.
Collapse
|
33
|
Shinde A, Illath K, Gupta P, Shinde P, Lim KT, Nagai M, Santra TS. A Review of Single-Cell Adhesion Force Kinetics and Applications. Cells 2021; 10:577. [PMID: 33808043 PMCID: PMC8000588 DOI: 10.3390/cells10030577] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Cells exert, sense, and respond to the different physical forces through diverse mechanisms and translating them into biochemical signals. The adhesion of cells is crucial in various developmental functions, such as to maintain tissue morphogenesis and homeostasis and activate critical signaling pathways regulating survival, migration, gene expression, and differentiation. More importantly, any mutations of adhesion receptors can lead to developmental disorders and diseases. Thus, it is essential to understand the regulation of cell adhesion during development and its contribution to various conditions with the help of quantitative methods. The techniques involved in offering different functionalities such as surface imaging to detect forces present at the cell-matrix and deliver quantitative parameters will help characterize the changes for various diseases. Here, we have briefly reviewed single-cell mechanical properties for mechanotransduction studies using standard and recently developed techniques. This is used to functionalize from the measurement of cellular deformability to the quantification of the interaction forces generated by a cell and exerted on its surroundings at single-cell with attachment and detachment events. The adhesive force measurement for single-cell microorganisms and single-molecules is emphasized as well. This focused review should be useful in laying out experiments which would bring the method to a broader range of research in the future.
Collapse
Affiliation(s)
- Ashwini Shinde
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Pallavi Shinde
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon-Si, Gangwon-Do 24341, Korea;
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempaku-cho, Toyohashi, Aichi 441-8580, Japan;
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| |
Collapse
|
34
|
Hwang RB. A theoretical design of evanescent wave biosensors based on gate-controlled graphene surface plasmon resonance. Sci Rep 2021; 11:1999. [PMID: 33479396 PMCID: PMC7820429 DOI: 10.1038/s41598-021-81595-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/11/2021] [Indexed: 11/10/2022] Open
Abstract
A surface plasmon resonance (SPR) sensor based on gate-controlled periodic graphene ribbons array is reported. Different from the conventional methods by monitoring reflectivity variations with respect to incident angle or wavelength, this approach measures the change in SPR curve against the variation of graphene chemical potential (via dynamically tuning the gate voltage) at both fixed incident angle and wavelength without the need of rotating mirror, tunable filter or spectrometer for angular or wavelength interrogation. Theoretical calculations show that the sensitivities are 36,401.1 mV/RIU, 40,676.5 mV/RIU, 40,918.2 mV/RIU, and 41,160 mV/RIU for analyte refractive index (RI) equal to 1.33, 1.34, 1.35 and 1.36; their figure of merit (1/RIU) are 21.84, 24, 23.74 and 23.69, respectively. Significantly, the enhancement in the non-uniform local field due to the subwavelength graphene ribbon resonator can facilitate the detection in redistribution of protein monolayers modeled as dielectric bricks.
Collapse
Affiliation(s)
- Ruey-Bing Hwang
- Institute of Communications Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu, 30050, Taiwan. .,Center for mmWave Smart Radar Systems and Technologies, National Chiao Tung University, Hsinchu, 30050, Taiwan.
| |
Collapse
|
35
|
Yang NZ, Hsiung CT, Huang CS. Biosensor based on two-dimensional gradient guided-mode resonance filter. OPTICS EXPRESS 2021; 29:1320-1332. [PMID: 33726350 DOI: 10.1364/oe.408597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/21/2020] [Indexed: 06/12/2023]
Abstract
A novel biosensor based on a two-dimensional gradient (TDG) guided-mode resonance (GMR) filter was introduced in this study. The TDG-GMR is demarcated in terms of the gradient grating period (GGP) in one dimension and gradient waveguide thickness (GWT) in the other dimension. A single compact sensor can combine these two features to simultaneously provide a broad detection range through GGP and high resolution through GWT. A detection range of 0.109 RIU (0%-60% sucrose content) with a limit of detection of 5.62 × 10-4 was demonstrated in this study by using a TDG-GMR with a size of 140.8 × 125.4 µm2. This value cannot be achieved using one dimensional gradient GMR sensor. Label-free (LF) biomolecule detection through TDG-GMR was also experimentally demonstrated in a model assay of albumin. The result confirms that the GWT-GMR provides a better resolution, whereas the GGP-GMR provides a broader detection range. A device for multiplex measurement could be easily implemented with a compact sensor chip and a simple readout directly from a charge-coupled device. This system would require a narrow-band source such as a light emitting diode or a laser diode, in addition to a limited number of other components such as a polarizer and a collimator. The proposed TDG-GMR could easily be integrated with smartphones and portable devices.
Collapse
|
36
|
Sijben HJ, van den Berg JJE, Broekhuis JD, IJzerman AP, Heitman LH. A study of the dopamine transporter using the TRACT assay, a novel in vitro tool for solute carrier drug discovery. Sci Rep 2021; 11:1312. [PMID: 33446713 PMCID: PMC7809260 DOI: 10.1038/s41598-020-79218-w] [Citation(s) in RCA: 7] [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: 09/17/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
Members of the solute carrier (SLC) transporter protein family are increasingly recognized as therapeutic drug targets. The majority of drug screening assays for SLCs are based on the uptake of radiolabeled or fluorescent substrates. Thus, these approaches often have limitations that compromise on throughput or the physiological environment of the SLC. In this study, we report a novel application of an impedance-based biosensor, xCELLigence, to investigate dopamine transporter (DAT) activity via substrate-induced activation of G protein-coupled receptors (GPCRs). The resulting assay, which is coined the 'transporter activity through receptor activation' (TRACT) assay, is based on the hypothesis that DAT-mediated removal of extracellular dopamine directly affects the ability of dopamine to activate cognate membrane-bound GPCRs. In two human cell lines with heterologous DAT expression, dopamine-induced GPCR signaling was attenuated. Pharmacological inhibition or the absence of DAT restored the apparent potency of dopamine for GPCR activation. The inhibitory potencies for DAT inhibitors GBR12909 (pIC50 = 6.2, 6.6) and cocaine (pIC50 = 6.3) were in line with values from reported orthogonal transport assays. Conclusively, this study demonstrates the novel use of label-free whole-cell biosensors to investigate DAT activity using GPCR activation as a readout. This holds promise for other SLCs that share their substrate with a GPCR.
Collapse
Affiliation(s)
- Hubert J Sijben
- Division of Drug Discovery and Safety, LACDR, Leiden University, P.O. Box 9502, 2300RA, Leiden, The Netherlands
| | - Julie J E van den Berg
- Division of Drug Discovery and Safety, LACDR, Leiden University, P.O. Box 9502, 2300RA, Leiden, The Netherlands
| | - Jeremy D Broekhuis
- Division of Drug Discovery and Safety, LACDR, Leiden University, P.O. Box 9502, 2300RA, Leiden, The Netherlands
| | - Adriaan P IJzerman
- Division of Drug Discovery and Safety, LACDR, Leiden University, P.O. Box 9502, 2300RA, Leiden, The Netherlands
| | - Laura H Heitman
- Division of Drug Discovery and Safety, LACDR, Leiden University, P.O. Box 9502, 2300RA, Leiden, The Netherlands.
- Oncode Institute, Leiden, The Netherlands.
| |
Collapse
|
37
|
Kanyo N, Kovacs KD, Saftics A, Szekacs I, Peter B, Santa-Maria AR, Walter FR, Dér A, Deli MA, Horvath R. Glycocalyx regulates the strength and kinetics of cancer cell adhesion revealed by biophysical models based on high resolution label-free optical data. Sci Rep 2020; 10:22422. [PMID: 33380731 PMCID: PMC7773743 DOI: 10.1038/s41598-020-80033-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
The glycocalyx is thought to perform a potent, but not yet defined function in cellular adhesion and signaling. Since 95% of cancer cells have altered glycocalyx structure, this role can be especially important in cancer development and metastasis. The glycocalyx layer of cancer cells directly influences cancer progression, involving the complicated kinetic process of cellular adhesion at various levels. In the present work, we investigated the effect of enzymatic digestion of specific glycocalyx components on cancer cell adhesion to RGD (arginine-glycine-aspartic acid) peptide motif displaying surfaces. High resolution kinetic data of cell adhesion was recorded by the surface sensitive label-free resonant waveguide grating (RWG) biosensor, supported by fluorescent staining of the cells and cell surface charge measurements. We found that intense removal of chondroitin sulfate (CS) and dermatan sulfate chains by chondroitinase ABC reduced the speed and decreased the strength of adhesion of HeLa cells. In contrast, mild digestion of glycocalyx resulted in faster and stronger adhesion. Control experiments on a healthy and another cancer cell line were also conducted, and the discrepancies were analysed. We developed a biophysical model which was fitted to the kinetic data of HeLa cells. Our analysis suggests that the rate of integrin receptor transport to the adhesion zone and integrin-RGD binding is strongly influenced by the presence of glycocalyx components, but the integrin-RGD dissociation is not. Moreover, based on the kinetic data we calculated the dependence of the dissociation constant of integrin-RGD binding on the enzyme concentration. We also determined the dissociation constant using a 2D receptor binding model based on saturation level static data recorded at surfaces with tuned RGD densities. We analyzed the discrepancies of the kinetic and static dissociation constants, further illuminating the role of cancer cell glycocalyx during the adhesion process. Altogether, our experimental results and modelling demonstrated that the chondroitin sulfate and dermatan sulfate chains of glycocalyx have an important regulatory function during the cellular adhesion process, mainly controlling the kinetics of integrin transport and integrin assembly into mature adhesion sites. Our results potentially open the way for novel type of cancer treatments affecting these regulatory mechanisms of cellular glycocalyx.
Collapse
Affiliation(s)
- Nicolett Kanyo
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege M. út 29-33, 1120, Budapest, Hungary
| | - Kinga Dora Kovacs
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege M. út 29-33, 1120, Budapest, Hungary
| | - Andras Saftics
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege M. út 29-33, 1120, Budapest, Hungary
| | - Inna Szekacs
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege M. út 29-33, 1120, Budapest, Hungary
| | - Beatrix Peter
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege M. út 29-33, 1120, Budapest, Hungary
| | - Ana R Santa-Maria
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62., 6726, Szeged, Hungary.,Doctoral School of Biology, University of Szeged, Közép fasor 52., 6726, Szeged, Hungary.,Department of Biotechnology, University of Szeged, Közép fasor 52., 6726, Szeged, Hungary
| | - Fruzsina R Walter
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62., 6726, Szeged, Hungary.,Department of Biotechnology, University of Szeged, Közép fasor 52., 6726, Szeged, Hungary
| | - András Dér
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62., 6726, Szeged, Hungary
| | - Mária A Deli
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62., 6726, Szeged, Hungary
| | - Robert Horvath
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege M. út 29-33, 1120, Budapest, Hungary.
| |
Collapse
|
38
|
Chen YT, Lee YC, Lai YH, Lim JC, Huang NT, Lin CT, Huang JJ. Review of Integrated Optical Biosensors for Point-Of-Care Applications. BIOSENSORS-BASEL 2020; 10:bios10120209. [PMID: 33353033 PMCID: PMC7766912 DOI: 10.3390/bios10120209] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/12/2020] [Accepted: 12/16/2020] [Indexed: 12/25/2022]
Abstract
This article reviews optical biosensors and their integration with microfluidic channels. The integrated biosensors have the advantages of higher accuracy and sensitivity because they can simultaneously monitor two or more parameters. They can further incorporate many functionalities such as electrical control and signal readout monolithically in a single semiconductor chip, making them ideal candidates for point-of-care testing. In this article, we discuss the applications by specifically looking into point-of-care testing (POCT) using integrated optical sensors. The requirement and future perspective of integrated optical biosensors for POC is addressed.
Collapse
Affiliation(s)
- Yung-Tsan Chen
- Graduate Institute of Photonics and Optoelectronics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (Y.-T.C.); (Y.-C.L.); (Y.-H.L.); (J.-C.L.)
| | - Ya-Chu Lee
- Graduate Institute of Photonics and Optoelectronics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (Y.-T.C.); (Y.-C.L.); (Y.-H.L.); (J.-C.L.)
| | - Yao-Hsuan Lai
- Graduate Institute of Photonics and Optoelectronics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (Y.-T.C.); (Y.-C.L.); (Y.-H.L.); (J.-C.L.)
| | - Jin-Chun Lim
- Graduate Institute of Photonics and Optoelectronics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (Y.-T.C.); (Y.-C.L.); (Y.-H.L.); (J.-C.L.)
| | - Nien-Tsu Huang
- Department of Electrical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (N.-T.H.); (C.-T.L.)
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
| | - Chih-Ting Lin
- Department of Electrical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (N.-T.H.); (C.-T.L.)
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
- Graduate Institute of Electronics Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan
| | - Jian-Jang Huang
- Graduate Institute of Photonics and Optoelectronics, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (Y.-T.C.); (Y.-C.L.); (Y.-H.L.); (J.-C.L.)
- Department of Electrical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan; (N.-T.H.); (C.-T.L.)
- Correspondence:
| |
Collapse
|
39
|
Jankovics H, Kovacs B, Saftics A, Gerecsei T, Tóth É, Szekacs I, Vonderviszt F, Horvath R. Grating-coupled interferometry reveals binding kinetics and affinities of Ni ions to genetically engineered protein layers. Sci Rep 2020; 10:22253. [PMID: 33335217 PMCID: PMC7746762 DOI: 10.1038/s41598-020-79226-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/03/2020] [Indexed: 12/18/2022] Open
Abstract
Reliable measurement of the binding kinetics of low molecular weight analytes to their targets is still a challenging task. Often, the introduction of labels is simply impossible in such measurements, and the application of label-free methods is the only reliable choice. By measuring the binding kinetics of Ni(II) ions to genetically modified flagellin layers, we demonstrate that: (1) Grating-Coupled Interferometry (GCI) is well suited to resolve the binding of ions, even at very low protein immobilization levels; (2) it supplies high quality kinetic data from which the number and strength of available binding sites can be determined, and (3) the rate constants of the binding events can also be obtained with high accuracy. Experiments were performed using a flagellin variant incorporating the C-terminal domain of the nickel-responsive transcription factor NikR. GCI results were compared to affinity data from titration calorimetry. We found that besides the low-affinity binding sites characterized by a micromolar dissociation constant (Kd), tetrameric FliC-NikRC molecules possess high-affinity binding sites with Kd values in the nanomolar range. GCI enabled us to obtain real-time kinetic data for the specific binding of an analyte with molar mass as low as 59 Da, even at signals lower than 1 pg/mm2.
Collapse
Affiliation(s)
- Hajnalka Jankovics
- Bio-Nanosystems Laboratory, Research Institute of Biomolecular and Chemical Engineering, University of Pannonia, Egyetem u. 10, Veszprém, Hungary
| | - Boglarka Kovacs
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege Miklós út 29-33, Budapest, Hungary
| | - Andras Saftics
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege Miklós út 29-33, Budapest, Hungary
| | - Tamas Gerecsei
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege Miklós út 29-33, Budapest, Hungary
| | - Éva Tóth
- Bio-Nanosystems Laboratory, Research Institute of Biomolecular and Chemical Engineering, University of Pannonia, Egyetem u. 10, Veszprém, Hungary
| | - Inna Szekacs
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege Miklós út 29-33, Budapest, Hungary
| | - Ferenc Vonderviszt
- Bio-Nanosystems Laboratory, Research Institute of Biomolecular and Chemical Engineering, University of Pannonia, Egyetem u. 10, Veszprém, Hungary
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege Miklós út 29-33, Budapest, Hungary
| | - Robert Horvath
- Nanobiosensorics Laboratory, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege Miklós út 29-33, Budapest, Hungary.
| |
Collapse
|
40
|
Suutari T, Rahman SN, Vischer HF, van Iperen D, Merivaara A, Yliperttula M, Leurs R, Kool J, Viitala T. Label-Free Analysis with Multiple Parameters Separates G Protein-Coupled Receptor Signaling Pathways. Anal Chem 2020; 92:14509-14516. [PMID: 33054153 DOI: 10.1021/acs.analchem.0c02652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Real-time label-free techniques are used to profile G protein-coupled receptor (GPCR) signaling pathways in living cells. However, interpreting the label-free signal responses is challenging, and previously reported methods do not reliably separate pathways from each other. In this study, a continuous angular-scanning surface plasmon resonance (SPR) technique is utilized for measuring label-free GPCR signal profiles. We show how the continuous angular-scanning ability, measuring up to nine real-time label-free parameters simultaneously, results in more information-rich label-free signal profiles for different GPCR pathways, providing a more accurate pathway separation. For this, we measured real-time full-angular SPR response curves for Gs, Gq, and Gi signaling pathways in living cells. By selecting two of the most prominent label-free parameters: the full SPR curve angular and intensity shifts, we present how this analysis approach can separate each of the three signaling pathways in a straightforward single-step analysis setup, without concurrent use of signal inhibitors or other response modulating compounds.
Collapse
Affiliation(s)
- Teemu Suutari
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland.,Division of BioAnalytical Chemistry, Amsterdam Institute for Medicines, Molecules and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Sabrina N Rahman
- Division of Medicinal Chemistry, Amsterdam Institute for Medicines, Molecules and Systems, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Henry F Vischer
- Division of Medicinal Chemistry, Amsterdam Institute for Medicines, Molecules and Systems, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Dick van Iperen
- Precision Mechanics and Engineering Bèta, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Arto Merivaara
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Marjo Yliperttula
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Rob Leurs
- Division of Medicinal Chemistry, Amsterdam Institute for Medicines, Molecules and Systems, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Jeroen Kool
- Division of BioAnalytical Chemistry, Amsterdam Institute for Medicines, Molecules and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands.,Centre for Analytical Sciences Amsterdam (CASA), 1098 XH Amsterdam, The Netherlands
| | - Tapani Viitala
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland.,Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
41
|
Li YCE, Lee IC. The Current Trends of Biosensors in Tissue Engineering. BIOSENSORS 2020; 10:E88. [PMID: 32756393 PMCID: PMC7459738 DOI: 10.3390/bios10080088] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 12/30/2022]
Abstract
Biosensors constitute selective, sensitive, and rapid tools for disease diagnosis in tissue engineering applications. Compared to standard enzyme-linked immunosorbent assay (ELISA) analytical technology, biosensors provide a strategy to real-time and on-site monitor micro biophysiological signals via a combination of biological, chemical, and physical technologies. This review summarizes the recent and significant advances made in various biosensor technologies for different applications of biological and biomedical interest, especially on tissue engineering applications. Different fabrication techniques utilized for tissue engineering purposes, such as computer numeric control (CNC), photolithographic, casting, and 3D printing technologies are also discussed. Key developments in the cell/tissue-based biosensors, biomolecular sensing strategies, and the expansion of several biochip approaches such as organs-on-chips, paper based-biochips, and flexible biosensors are available. Cell polarity and cell behaviors such as proliferation, differentiation, stimulation response, and metabolism detection are included. Biosensors for diagnosing tissue disease modes such as brain, heart, lung, and liver systems and for bioimaging are discussed. Finally, we discuss the challenges faced by current biosensing techniques and highlight future prospects of biosensors for tissue engineering applications.
Collapse
Affiliation(s)
- Yi-Chen Ethan Li
- Department of Chemical Engineering, Feng Chia University, Taichung 40724, Taiwan
| | - I-Chi Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| |
Collapse
|
42
|
Is GPR146 really the receptor for proinsulin C-peptide? Bioorg Med Chem Lett 2020; 30:127208. [DOI: 10.1016/j.bmcl.2020.127208] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 01/01/2023]
|
43
|
Lee KL, Hou HS, Cheng JY, Wei PK. High-Throughput and Dynamic Study of Drug and Cell Interactions Using Contrast Images in Aluminum-Based Nanoslit Arrays. Anal Chem 2020; 92:9674-9681. [DOI: 10.1021/acs.analchem.0c00972] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Kuang-Li Lee
- Research Center for Applied Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Hsien-San Hou
- Research Center for Applied Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Ji-Yen Cheng
- Research Center for Applied Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Pei-Kuen Wei
- Research Center for Applied Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
- Institute of Optoelectronic Sciences, National Taiwan Ocean University, Keelung 20224, Taiwan
- Institute of Biophotonics, National Yang-Ming University, Taipei 11221, Taiwan
| |
Collapse
|
44
|
Meyrath M, Szpakowska M, Zeiner J, Massotte L, Merz MP, Benkel T, Simon K, Ohnmacht J, Turner JD, Krüger R, Seutin V, Ollert M, Kostenis E, Chevigné A. The atypical chemokine receptor ACKR3/CXCR7 is a broad-spectrum scavenger for opioid peptides. Nat Commun 2020; 11:3033. [PMID: 32561830 PMCID: PMC7305236 DOI: 10.1038/s41467-020-16664-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 05/15/2020] [Indexed: 12/29/2022] Open
Abstract
Endogenous opioid peptides and prescription opioid drugs modulate pain, anxiety and stress by activating opioid receptors, currently classified into four subtypes. Here we demonstrate that ACKR3/CXCR7, hitherto known as an atypical scavenger receptor for chemokines, is a broad-spectrum scavenger of opioid peptides. Phylogenetically, ACKR3 is intermediate between chemokine and opioid receptors and is present in various brain regions together with classical opioid receptors. Functionally, ACKR3 is a scavenger receptor for a wide variety of opioid peptides, especially enkephalins and dynorphins, reducing their availability for the classical opioid receptors. ACKR3 is not modulated by prescription opioids, but we show that an ACKR3-selective subnanomolar competitor peptide, LIH383, can restrain ACKR3’s negative regulatory function on opioid peptides in rat brain and potentiate their activity towards classical receptors, which may open alternative therapeutic avenues for opioid-related disorders. Altogether, our results reveal that ACKR3 is an atypical opioid receptor with cross-family ligand selectivity. Opioids modulate pain, anxiety and stress by activating four subtypes of opioid receptors. The authors show that atypical chemokine receptor 3 (ACKR3) is a scavenger for various endogenous opioid peptides regulating their availability without activating downstream signaling.
Collapse
Affiliation(s)
- Max Meyrath
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Martyna Szpakowska
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Julian Zeiner
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Laurent Massotte
- Neurophysiology Unit, GIGA Neurosciences, University of Liège, avenue de l'hopital, B-4000, Liège, Belgium
| | - Myriam P Merz
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Tobias Benkel
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany.,Research Training Group 1873, University of Bonn, Bonn, Germany
| | - Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Jochen Ohnmacht
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg.,Department of Life Sciences and Medicine, University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg
| | - Jonathan D Turner
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg.,Transversal Translational Medicine, Luxembourg Institute of Health (LIH), rue Thomas Edison 1A-B, L-1445, Strassen, Luxembourg
| | - Vincent Seutin
- Neurophysiology Unit, GIGA Neurosciences, University of Liège, avenue de l'hopital, B-4000, Liège, Belgium
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, 5000, Odense, Denmark
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Andy Chevigné
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
45
|
Reichmuth AM, Zimmermann M, Wilhelm F, Frutiger A, Blickenstorfer Y, Fattinger C, Waldhoer M, Vörös J. Quantification of Molecular Interactions in Living Cells in Real Time using a Membrane Protein Nanopattern. Anal Chem 2020; 92:8983-8991. [DOI: 10.1021/acs.analchem.0c00987] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Andreas Michael Reichmuth
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, University and ETH Zurich, 8092 Zurich, Switzerland
| | | | - Florian Wilhelm
- InterAx Biotech, PARK innovAARE, 5234 Villigen, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Andreas Frutiger
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, University and ETH Zurich, 8092 Zurich, Switzerland
| | - Yves Blickenstorfer
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, University and ETH Zurich, 8092 Zurich, Switzerland
| | - Christof Fattinger
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Maria Waldhoer
- InterAx Biotech, PARK innovAARE, 5234 Villigen, Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, University and ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
46
|
Gauglitz G. Critical assessment of relevant methods in the field of biosensors with direct optical detection based on fibers and waveguides using plasmonic, resonance, and interference effects. Anal Bioanal Chem 2020; 412:3317-3349. [PMID: 32313998 PMCID: PMC7214504 DOI: 10.1007/s00216-020-02581-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 12/16/2022]
Abstract
Direct optical detection has proven to be a highly interesting tool in biomolecular interaction analysis to be used in drug discovery, ligand/receptor interactions, environmental analysis, clinical diagnostics, screening of large data volumes in immunology, cancer therapy, or personalized medicine. In this review, the fundamental optical principles and applications are reviewed. Devices are based on concepts such as refractometry, evanescent field, waveguides modes, reflectometry, resonance and/or interference. They are realized in ring resonators; prism couplers; surface plasmon resonance; resonant mirror; Bragg grating; grating couplers; photonic crystals, Mach-Zehnder, Young, Hartman interferometers; backscattering; ellipsometry; or reflectance interferometry. The physical theories of various optical principles have already been reviewed in detail elsewhere and are therefore only cited. This review provides an overall survey on the application of these methods in direct optical biosensing. The "historical" development of the main principles is given to understand the various, and sometimes only slightly modified variations published as "new" methods or the use of a new acronym and commercialization by different companies. Improvement of optics is only one way to increase the quality of biosensors. Additional essential aspects are the surface modification of transducers, immobilization strategies, selection of recognition elements, the influence of non-specific interaction, selectivity, and sensitivity. Furthermore, papers use for reporting minimal amounts of detectable analyte terms such as value of mass, moles, grams, or mol/L which are difficult to compare. Both these essential aspects (i.e., biochemistry and the presentation of LOD values) can be discussed only in brief (but references are provided) in order to prevent the paper from becoming too long. The review will concentrate on a comparison of the optical methods, their application, and the resulting bioanalytical quality.
Collapse
Affiliation(s)
- Günter Gauglitz
- Institute of Physical and Theoretical Chemistry, Eberhard Karls Universität, Auf der Morgenstelle 18, 72076, Tübingen, Germany.
| |
Collapse
|
47
|
Debreczeni ML, Szekacs I, Kovacs B, Saftics A, Kurunczi S, Gál P, Dobó J, Cervenak L, Horvath R. Human primary endothelial label-free biochip assay reveals unpredicted functions of plasma serine proteases. Sci Rep 2020; 10:3303. [PMID: 32094469 PMCID: PMC7039951 DOI: 10.1038/s41598-020-60158-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/07/2020] [Indexed: 11/09/2022] Open
Abstract
Tissue-on-a-chip technologies are more and more important in the investigation of cellular function and in the development of novel drugs by allowing the direct screening of substances on human cells. Constituting the inner lining of vessel walls, endothelial cells are the key players in various physiological processes, moreover, they are the first to be exposed to most drugs currently used. However, to date, there is still no appropriate technology for the label-free, real-time and high-throughput monitoring of endothelial function. To this end, we developed an optical biosensor-based endothelial label-free biochip (EnLaB) assay that meets all the above requirements. Using our EnLaB platform, we screened a set of plasma serine proteases as possible endothelial cell activators, and first identified the endothelial cell activating function of three important serine proteases - namely kallikrein, C1r and mannan-binding lectin-associated serine-protease 2 (MASP-2) - and verified these results in well-established functional assays. EnLaB proved to be an effective tool for revealing novel cellular mechanisms as well as for the high-throughput screening of various compounds on endothelial cells.
Collapse
Affiliation(s)
| | - Inna Szekacs
- Nanobiosensorics Momentum Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege M. út 29-33, H-1120, Budapest, Hungary
| | - Boglarka Kovacs
- Nanobiosensorics Momentum Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege M. út 29-33, H-1120, Budapest, Hungary
| | - Andras Saftics
- Nanobiosensorics Momentum Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege M. út 29-33, H-1120, Budapest, Hungary
| | - Sándor Kurunczi
- Nanobiosensorics Momentum Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Konkoly-Thege M. út 29-33, H-1120, Budapest, Hungary
| | - Péter Gál
- Institute of Enzymology, Research Centre for Natural Sciences, H-1113, Budapest, Hungary
| | - József Dobó
- Institute of Enzymology, Research Centre for Natural Sciences, H-1113, Budapest, Hungary
| | - László Cervenak
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary.
| | - Robert Horvath
- 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
48
|
Systematic characterization of AT1 receptor antagonists with label-free dynamic mass redistribution assays. J Pharmacol Toxicol Methods 2020; 102:106682. [PMID: 32087363 DOI: 10.1016/j.vascn.2020.106682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/23/2019] [Accepted: 02/07/2020] [Indexed: 11/23/2022]
Abstract
INTRODUCTION In the drug discovery field, the binding affinities and binding kinetics of drug candidates are very important. Angiotensin II type 1 (AT1) receptor antagonists, e.g., candesartan, telmisartan, irbesartan, losartan and valsartan, show high affinities and long-lasting bindings to the receptor, making them preferred medications for hypertension treatment. However, the molecular binding properties of AT1 receptor antagonists are controversial. METHODS In this work, we established a profile to study the phenotypic properties of AT1 receptor antagonists with label-free dynamic mass redistribution (DMR) assays in native human cells. With noninvasive features, DMR assay were conducted in multiple formats. Eleven antagonists were systematically evaluated with angiotensin II as an agonist probe in the Hep G2 cell line, which endogenously expresses the AT1 receptor. RESULTS The IC50 values to the AT1 receptor of individual antagonist varied with different incubation times. The antagonists showed competitive behavior with angiotensin II. Schild analysis was used to analyze the competitive behavior of the antagonist. All of the antagonist showed long-lasting possession of the AT1 receptor, except telmisartan. The systematic evaluation of the antagonists implied that 11 antagonists showed high binding affinity but distinct binding modes to AT1 receptor. DISCUSSION This study demonstrated that the DMR assay has great potential for determining the pharmacological parameters of ligands. This work may serve as guidance for other receptor and ligand assays.
Collapse
|
49
|
Sztilkovics M, Gerecsei T, Peter B, Saftics A, Kurunczi S, Szekacs I, Szabo B, Horvath R. Single-cell adhesion force kinetics of cell populations from combined label-free optical biosensor and robotic fluidic force microscopy. Sci Rep 2020; 10:61. [PMID: 31919421 PMCID: PMC6952389 DOI: 10.1038/s41598-019-56898-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/18/2019] [Indexed: 01/03/2023] Open
Abstract
Single-cell adhesion force plays a crucial role in biological sciences, however its in-depth investigation is hindered by the extremely low throughput and the lack of temporal resolution of present techniques. While atomic force microcopy (AFM) based methods are capable of directly measuring the detachment force values between individual cells and a substrate, their throughput is limited to few cells per day, and cannot provide the kinetic evaluation of the adhesion force over the timescale of several hours. In this study a high spatial and temporal resolution resonant waveguide grating based label-free optical biosensor was combined with robotic fluidic force microscopy to monitor the adhesion of living cancer cells. In contrast to traditional fluidic force microscopy methods with a manipulation range in the order of 300-400 micrometers, the robotic device employed here can address single cells over mm-cm scale areas. This feature significantly increased measurement throughput, and opened the way to combine the technology with the employed microplate-based, large area biosensor. After calibrating the biosensor signals with the direct force measuring technology on 30 individual cells, the kinetic evaluation of the adhesion force and energy of large cell populations was performed for the first time. We concluded that the distribution of the single-cell adhesion force and energy can be fitted by log-normal functions as cells are spreading on the surface and revealed the dynamic changes in these distributions. The present methodology opens the way for the quantitative assessment of the kinetics of single-cell adhesion force and energy with an unprecedented throughput and time resolution, in a completely non-invasive manner.
Collapse
Affiliation(s)
- Milan Sztilkovics
- Nanobiosensorics Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary
| | - Tamas Gerecsei
- Nanobiosensorics Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary
- Department of Biological Physics, Eötvös University, Budapest, Hungary
| | - Beatrix Peter
- Nanobiosensorics Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary
| | - Andras Saftics
- Nanobiosensorics Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary
| | - Sandor Kurunczi
- Nanobiosensorics Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary
| | - Inna Szekacs
- Nanobiosensorics Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary
| | - Balint Szabo
- Department of Biological Physics, Eötvös University, Budapest, Hungary
| | - Robert Horvath
- Nanobiosensorics Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Budapest, Hungary.
| |
Collapse
|
50
|
Rappaz B, Jourdain P, Banfi D, Kuttler F, Marquet P, Turcatti G. Image-Based Marker-Free Screening of GABA A Agonists, Antagonists, and Modulators. SLAS DISCOVERY 2019; 25:458-470. [PMID: 31779505 PMCID: PMC7243081 DOI: 10.1177/2472555219887142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The ionotropic GABAA receptors represent the main target for different groups of widely used drugs having hypnotic and anxiolytic effects. So far, most approaches used to assess GABA activity involve invasive low -throughput electrophysiological techniques or rely on fluorescent dyes, preventing the ability to conduct noninvasive and thus nonperturbing screens. To address this limitation, we have developed an automated marker-free cell imaging method, based on digital holographic microscopy (DHM). This technology allows the automatically screening of compounds in multiple plates without having to label the cells or use special plates. This methodological approach was first validated by screening the GABAA receptor expressed in HEK cells using a selection of active compounds in agonist, antagonist, and modulator modes. Then, in a second blind screen of a library of 3041 compounds (mostly composed of natural products), 5 compounds having a specific agonist action on the GABAA receptor were identified. The hits validated from this unbiased screen were the natural products muscimol, neurosteroid alphaxalone, and three compounds belonging to the avermectin family, all known for having an agonistic effect on the GABAA receptor. The results obtained were exempt from false negatives (structurally similar unassigned hits), and false-positive hits were detected and discarded without the need for performing electrophysiological measurements. The outcome of the screen demonstrates the applicability of our screening by imaging method for the discovery of new chemical structures, particularly regarding chemicals interacting with the ionotropic GABAA receptor and more generally with any ligand-gated ion channels and transporters.
Collapse
Affiliation(s)
- Benjamin Rappaz
- Biomolecular Screening Facility (BSF), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Pascal Jourdain
- Joint International Research Unit in Child Psychiatry, Département de Psychiatrie CHUV, University of Lausanne, Switzerland.,Université Laval, Québec, QC, Canada
| | - Damiano Banfi
- Biomolecular Screening Facility (BSF), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Fabien Kuttler
- Biomolecular Screening Facility (BSF), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Pierre Marquet
- Joint International Research Unit in Child Psychiatry, Département de Psychiatrie CHUV, University of Lausanne, Switzerland.,Université Laval, Québec, QC, Canada.,Centre de Recherche CERVO, Québec, QC, Canada
| | - Gerardo Turcatti
- Biomolecular Screening Facility (BSF), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|