51
|
Qu L, Zhang X, Wang J, Zhou H, Hou T, Wei L, Xu F, Liang X. Phenotypic assessment and ligand screening of ETA/ETB receptors with label-free dynamic mass redistribution assay. Naunyn Schmiedebergs Arch Pharmacol 2019; 393:937-950. [PMID: 31781785 DOI: 10.1007/s00210-019-01756-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/23/2019] [Indexed: 01/16/2023]
Abstract
Endothelin receptors, consisting of two subtypes, ETA and ETB, are expressed in various tissues and widely regulate cardiovascular systems. The two receptors show distinct biological characteristics and are involved in different downstream pathways. Hence, to evaluate the ETA and ETB receptors on the same platform is helpful to display their pharmacological features. In this study, we developed a label-free dynamic mass redistribution (DMR) assay to investigate the phenotypic features of the ETA and ETB receptors in native cell lines. Meanwhile, specific agonists and antagonists were investigated for their pharmacological parameters. Results indicated that the DMR response of endothelin 1 (ET-1, an endogenous ETA/ETB agonist) was cell line dependent on ETA receptors and this ligand generated a biphasic dose-response curve in SH-SY5Y as well as PC3 cell lines. ET-1 and IRL 1620 (an ETB agonist) showed different DMR responses in U251 cells. IC50 values of antagonists were consistent with the Ki values previously reported. Furthermore, a list of compounds was screened on the ETA and ETB receptor models established by the high-throughput DMR assays. This study demonstrated that the DMR assay had great potential in the phenotypic-based investigation and ligand screening of GPCRs.
Collapse
Affiliation(s)
- Lala Qu
- Key Lab of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiuli Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215006, China.
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, China.
| | - Jixia Wang
- Key Lab of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Han Zhou
- Key Lab of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Tao Hou
- Key Lab of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Lai Wei
- Key Lab of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Fangfang Xu
- Key Lab of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinmiao Liang
- Key Lab of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China.
| |
Collapse
|
52
|
Chen L, Li P, Lu X, Wang S, Zheng Z. Binary polymer brush patterns from facile initiator stickiness for cell culturing. Faraday Discuss 2019; 219:189-202. [PMID: 31317169 DOI: 10.1039/c9fd00013e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report a new initiator stickiness method to fabricate micropatterned binary polymer brush surfaces, which are ideal platforms for studying cell adhesion behavior. The atom transfer radical polymerization (ATRP) initiator, ω-mercaptoundecyl bromoisobutyrate (MUDBr), is found to adsorb on several hosting polymer brushes, including poly[oligo(ethylene glycol)methyl ether methacrylate] (POEGMA), poly(2-hydroxyethyl methacrylate) (PHEMA), and poly(glycidyl methacrylate) (PGMA) brushes. Based on the initiator stickiness, micropatterned initiator molecules are printed onto a layer of homogenous hosting polymer brushes via microcontact printing (μCP), and then, vertically, a patterned second layer of polymer brushes is grown from the initiator areas. With this simple, fast, and additive method, we demonstrate the fabrication of various binary polymer brushes, and show their applications for patterning cell microarrays and controlling cell orientation. This new approach to generating binary polymer brushes shows great potential for the manipulation of interfacial phenomena, facilitating a range of applications from semiconductors and lubrication to fundamental cell biology studies.
Collapse
Affiliation(s)
- Lina Chen
- Laboratory for Advanced Interfacial Materials and Devices, Institute of Textiles and Clothing, The Hong Kong Polytechnic University, Hong Kong SAR, P. R. China.
| | | | | | | | | |
Collapse
|
53
|
Banville FA, Moreau J, Chabot K, Cattoni A, Fröhlich U, Bryche JF, Collin S, Charette PG, Grandbois M, Canva M. Nanoplasmonics-enhanced label-free imaging of endothelial cell monolayer integrity. Biosens Bioelectron 2019; 141:111478. [PMID: 31280004 DOI: 10.1016/j.bios.2019.111478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 01/24/2023]
Abstract
Surface plasmon resonance imaging (SPRI) is a powerful label-free imaging modality for the analysis of morphological dynamics in cell monolayers. However, classical plasmonic imaging systems have relatively poor spatial resolution along one axis due to the plasmon mode attenuation distance (tens of μm, typically), which significantly limits their ability to resolve subcellular structures. We address this limitation by adding an array of nanostructures onto the metal sensing surface (25 nm thick, 200 nm width, 400 nm period grating) to couple localized plasmons with propagating plasmons, thereby reducing attenuation length and commensurately increasing spatial imaging resolution, without significant loss of sensitivity or image contrast. In this work, experimental results obtained with both conventional unstructured and nanostructured gold film SPRI sensor chips show a clear gain in spatial resolution achieved with surface nanostructuring. The work demonstrates the ability of the nanostructured SPRI chips to resolve fine morphological detail (intercellular gaps) in experiments monitoring changes in endothelial cell monolayer integrity following the activation of the cell surface protease-activated receptor 1 (PAR1) by thrombin. In particular, the nanostructured chips reveal the persistence of small intercellular gaps (<5 μm2) well after apparent recovery of cell monolayer integrity as determined by conventional unstructured surface based SPRI. This new high spatial resolution plasmonic imaging technique uses low-cost and reusable patterned substrates and is likely to find applications in cell biology and pharmacology by allowing label-free quantification of minute cell morphological activities associated with receptor dependent intracellular signaling activity.
Collapse
Affiliation(s)
- Frederic A Banville
- Laboratoire Nanotechnologies Nanosystèmes (LN2), CNRS UMI-3463, Université de Sherbrooke, Sherbrooke, J1K 0A5, Canada; Institut Interdisciplinaire d'Innovation Technologique (3IT), Université de Sherbrooke, Sherbrooke, J1K 0A5, Canada; Laboratoire Charles Fabry (LCF), Institut d'Optique Graduate School, Université Paris-Saclay, CNRS, Palaiseau, 91127, France
| | - Julien Moreau
- Laboratoire Charles Fabry (LCF), Institut d'Optique Graduate School, Université Paris-Saclay, CNRS, Palaiseau, 91127, France
| | - Kevin Chabot
- Laboratoire Nanotechnologies Nanosystèmes (LN2), CNRS UMI-3463, Université de Sherbrooke, Sherbrooke, J1K 0A5, Canada; Institut Interdisciplinaire d'Innovation Technologique (3IT), Université de Sherbrooke, Sherbrooke, J1K 0A5, Canada
| | - Andrea Cattoni
- Centre de Nanosciences et de Nanotechnologies (C2N), CNRS UMR-9001, Université Paris-Sud/Paris-Saclay, Palaiseau, 91120, France
| | - Ulrike Fröhlich
- Département de Pharmacologie et Physiologie, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Canada
| | - Jean-François Bryche
- Laboratoire Nanotechnologies Nanosystèmes (LN2), CNRS UMI-3463, Université de Sherbrooke, Sherbrooke, J1K 0A5, Canada; Institut Interdisciplinaire d'Innovation Technologique (3IT), Université de Sherbrooke, Sherbrooke, J1K 0A5, Canada
| | - Stéphane Collin
- Centre de Nanosciences et de Nanotechnologies (C2N), CNRS UMR-9001, Université Paris-Sud/Paris-Saclay, Palaiseau, 91120, France
| | - Paul G Charette
- Laboratoire Nanotechnologies Nanosystèmes (LN2), CNRS UMI-3463, Université de Sherbrooke, Sherbrooke, J1K 0A5, Canada; Institut Interdisciplinaire d'Innovation Technologique (3IT), Université de Sherbrooke, Sherbrooke, J1K 0A5, Canada
| | - Michel Grandbois
- Laboratoire Nanotechnologies Nanosystèmes (LN2), CNRS UMI-3463, Université de Sherbrooke, Sherbrooke, J1K 0A5, Canada; Département de Pharmacologie et Physiologie, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Canada
| | - Michael Canva
- Laboratoire Nanotechnologies Nanosystèmes (LN2), CNRS UMI-3463, Université de Sherbrooke, Sherbrooke, J1K 0A5, Canada; Institut Interdisciplinaire d'Innovation Technologique (3IT), Université de Sherbrooke, Sherbrooke, J1K 0A5, Canada; Laboratoire Charles Fabry (LCF), Institut d'Optique Graduate School, Université Paris-Saclay, CNRS, Palaiseau, 91127, France.
| |
Collapse
|
54
|
Silicon nitride sugar chips for detection of Ricinus communis proteins and Escherichia coli O157 Shiga toxins. Anal Biochem 2019; 580:42-48. [PMID: 31173726 DOI: 10.1016/j.ab.2019.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/21/2019] [Accepted: 06/03/2019] [Indexed: 11/21/2022]
Abstract
Lactosides having either an amino-triethylene glycol or an azido-triethylene glycol were designed and synthesized, and the two derivatives were immobilized onto silicon nitride (SiN) surfaces. When a click reaction was applied for the immobilization of the azido-sugar, a Ricinus communis lectin (RCA120) was detected with a higher response by reflectometric interference spectroscopy (RIfS). When an N-hydroxysuccinimide (NHS) method was applied for the sugar immobilization, the response was less than that of the click one. The response of bovine serum albumin (BSA) as the negative control was negligible, but the lactose-SiN chip prepared by the click method suppressed nonspecific binding more effectively than did the chip from the NHS method. Next, we examined an antibody-immobilized SiN chip prepared by the click reaction. The detection response was, however, lower than that of the lactose-SiN chip, meaning that the sugar-chip by the click reaction was superior to the antibody-chip. Finally, to detect Shiga toxins from Escherichia coli O157:H7, globotrisaccharide (Gb3) with an azido-triethylene glycol was synthesized and immobilized onto the SiN chip by the click reaction. The Gb3-SiN chips enabled us to detect the toxins at concentrations less than 100 ng/mL. RCA120, horse gram, gorse lectins and BSA showed no response to the Gb3-SiN chip, showing a high specificity for the toxin.
Collapse
|
55
|
Resonant Grating without a Planar Waveguide Layer as a Refractive Index Sensor. SENSORS 2019; 19:s19133003. [PMID: 31288404 PMCID: PMC6651248 DOI: 10.3390/s19133003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 06/28/2019] [Accepted: 07/05/2019] [Indexed: 12/13/2022]
Abstract
Dielectric grating-based sensors are usually based on the guided mode resonance (GMR) obtained using a thin planar waveguide layer (PWL) adjacent to a thin subwavelength grating layer. In this work, we present a detailed investigation of thick subwavelength dielectric grating structures that exhibit reflection resonances above a certain thickness without the need for the waveguide layer, showing great potential for applications in biosensing and tunable filtering. Analytic and numerical results are thoroughly discussed, as well as an experimental demonstration of the structure as a chemical sensor in the SWIR (short wave infrared) spectral range (1200-1800 nm). In comparison to the GMR structure with PWL, the thick grating structure has several unique properties: (i) It gives higher sensitivity when the spaces are filled, with the analyte peaking at certain space values due to an increase in the interaction volume between the analyte and the evanescent optical field between the grating lines; (ii) the TM (transverse magnetic) resonance, in certain cases, provides a better figure of merit; (iii) the sensitivity increases as the grating height increases; (iii) the prediction of the resonance locations based on the effective medium approximation does not give satisfactory results when the grating height is larger than a certain value, and the invalidity becomes more severe as the period increases; (iv) a sudden increase in the Q-factor of the resonance occurs at a specific height value accompanied by the high local field enhancement (~103) characteristic of a nano-antenna type pattern. Rigorous numerical simulations of the field distribution are presented to explain the different observed phenomena.
Collapse
|
56
|
Székács I, Tokarz P, Horvath R, Kovács K, Kubas A, Shimura M, Brasun J, Murzin V, Caliebe W, Szewczuk Z, Paluch A, Wojnárovits L, Tóth T, Pap JS, Szyrwiel Ł. In vitro SOD-like activity of mono- and di-copper complexes with a phosphonate substituted SALAN-type ligand. Chem Biol Interact 2019; 306:78-88. [DOI: 10.1016/j.cbi.2019.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/20/2019] [Accepted: 04/01/2019] [Indexed: 10/27/2022]
|
57
|
Hou HS, Lee KL, Wang CH, Hsieh TH, Sun JJ, Wei PK, Cheng JY. Simultaneous assessment of cell morphology and adhesion using aluminum nanoslit-based plasmonic biosensing chips. Sci Rep 2019; 9:7204. [PMID: 31076598 PMCID: PMC6510726 DOI: 10.1038/s41598-019-43442-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/24/2019] [Indexed: 11/13/2022] Open
Abstract
A variety of physiological and pathological processes rely on cell adhesion, which is most often tracked by changes in cellular morphology. We previously reported a novel gold nanoslit-based biosensor that is capable of real-time and label-free monitoring of cell morphological changes and cell viability. However, the preparation of gold biosensors is inefficient, complicated and costly. Recently, nanostructure-based aluminum (Al) sensors have been introduced for biosensing applications. The Al-based sensor has a longer decay length and is capable of analyzing large-sized mass such as cells. Here, we developed two types of double-layer Al nanoslit-based plasmonic biosensors, which were nanofabricated and used to evaluate the correlation between metastatic potency and adhesion of lung cancer and melanoma cell lines. Cell adhesion was determined by Fano resonance signals that were induced by binding of the cells to the nanoslit. The peak and dip of the Fano resonance spectrum respectively reflected long- and short-range cellular changes, allowing us to simultaneously detect and distinguish between focal adhesion and cell spreading. Also, the Al nanoslit-based biosensor chips were used to evaluate the inhibitory effects of drugs on cancer cell spreading. We are the first to report the use of double layer Al nanoslit-based biosensors for detection of cell behavior, and such devices may become powerful tools for anti-metastasis drug screening in the future.
Collapse
Affiliation(s)
- Hsien-San Hou
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Kuang-Li Lee
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Chen-Hung Wang
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Tung-Han Hsieh
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Juan-Jie Sun
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Pei-Kuen Wei
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan.,Institute of Biophotonics, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Ji-Yen Cheng
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan. .,Institute of Biophotonics, National Yang-Ming University, Taipei, 11221, Taiwan. .,Department of Mechanical and Mechatronic Engineering, National Taiwan Ocean University, Keelung, 20224, Taiwan. .,College of Engineering, Chang Gung University, Taoyuan, 33302, Taiwan.
| |
Collapse
|
58
|
Advantages and shortcomings of cell-based electrical impedance measurements as a GPCR drug discovery tool. Biosens Bioelectron 2019; 137:33-44. [PMID: 31077988 DOI: 10.1016/j.bios.2019.04.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/05/2019] [Accepted: 04/20/2019] [Indexed: 12/13/2022]
Abstract
G Protein-Coupled Receptors (GPCRs) transduce extracellular signals and activate intracellular pathways, usually through activating associated G proteins. Due to their involvement in many human diseases, they are recognized worldwide as valuable drug targets. Many experimental approaches help identify small molecules that target GPCRs, including in vitro cell-based reporter assays and binding studies. Most cell-based assays use one signaling pathway or reporter as an assay readout. Moreover, they often require cell labeling or the integration of reporter systems. Over the last decades, cell-based electrical impedance biosensors have been explored for drug discovery. This label-free method holds many advantages over other cellular assays in GPCR research. The technology requires no cell manipulation and offers real-time kinetic measurements of receptor-mediated cellular changes. Instead of measuring the activity of a single reporter, the impedance readout includes information on multiple signaling events. This is beneficial when screening for ligands targeting orphan GPCRs since the signaling cascade(s) of the majority of these receptors are unknown. Due to its sensitivity, the method also applies to cellular models more relevant to disease, including patient-derived cell cultures. Despite its advantages, remaining issues regarding data comparability and interpretability has limited implementation of cell-based electrical impedance (CEI) in drug discovery. Future optimization must include both full exploitation of CEI response data using various ways of analysis as well as further exploration of its potential to detect biased activities early on in drug discovery. Here, we review the contribution of CEI technology to GPCR research, discuss its comparative benefits, and provide recommendations.
Collapse
|
59
|
Prasad A, Choi J, Jia Z, Park S, Gartia MR. Nanohole array plasmonic biosensors: Emerging point-of-care applications. Biosens Bioelectron 2019; 130:185-203. [PMID: 30738247 PMCID: PMC6475599 DOI: 10.1016/j.bios.2019.01.037] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/03/2019] [Accepted: 01/18/2019] [Indexed: 01/18/2023]
Abstract
Point-of-care (POC) applications have expanded hugely in recent years and is likely to continue, with an aim to deliver cheap, portable, and reliable devices to meet the demands of healthcare industry. POC devices are designed, prototyped, and assembled using numerous strategies but the key essential features that biosensing devices require are: (1) sensitivity, (2) selectivity, (3) specificity, (4) repeatability, and (5) good limit of detection. Overall the fabrication and commercialization of the nanohole array (NHA) setup to the outside world still remains a challenge. Here, we review the various methods of NHA fabrication, the design criteria, the geometrical features, the effects of surface plasmon resonance (SPR) on sensing as well as current state-of-the-art of existing NHA sensors. This review also provides easy-to-understand examples of NHA-based POC biosensing applications, its current status, challenges, and future prospects.
Collapse
Affiliation(s)
- Alisha Prasad
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Junseo Choi
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA; NIH Center for BioModular Multiscale Systems for Precision Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Zheng Jia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA; NIH Center for BioModular Multiscale Systems for Precision Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Sunggook Park
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA; NIH Center for BioModular Multiscale Systems for Precision Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
60
|
Wang R, Wang J, Liu Y, Zhang X, Liang X. Resonant waveguide grating based assays for colloidal aggregate detection and promiscuity characterization in natural products. RSC Adv 2019; 9:38055-38064. [PMID: 35541809 PMCID: PMC9075791 DOI: 10.1039/c9ra06466d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 11/11/2019] [Indexed: 01/10/2023] Open
Abstract
Small molecules, including natural compounds, in aqueous buffer that self-associate into colloidal aggregates is the main cause of false results in the early stage of drug discovery. Here we reported resonant waveguide grating (RWG) based assays to identify natural compound aggregation and characterize its influence on membrane receptors in living cells. We first applied a cell-free aggregation assay to determine compound critical aggregation concentration (CAC) values. Then we characterized the aggregators' influence on membrane receptors using three types of dynamic mass redistribution (DMR) assays. Results showed that colloidal aggregates may cause false activity in DMR desensitization assays; some of the false activities can be implied by the large response in DMR agonism assays and can further be identified by DMR antagonism assays. Furthermore, the aggregation mechanism was confirmed by addition of 0.025% tween-80, with cell signals attenuated and potency decreased. Finally, these observations were used for aggregate examination and promiscuity investigation of a traditional herbal medicine, Rhodiola rosea, which ultimately led to the revealing of the true target and reduced the risk of a bioactivity tracking process at the very first stage. This study highlights that the RWG based assays can be used as practical tools to distinguish between real and false hits to provide reliable results in the early stage of drug discovery. Resonant waveguide grating based assays to eliminate colloidal aggregate induced false activity involving natural products.![]()
Collapse
Affiliation(s)
- Rong Wang
- Key Lab of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- China
| | - Jixia Wang
- Key Lab of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- China
| | - Yanfang Liu
- Key Lab of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- China
| | - Xiuli Zhang
- College of Pharmaceutical Science
- Soochow University
- Suzhou 215123
- China
| | - Xinmiao Liang
- Key Lab of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
- China
| |
Collapse
|
61
|
Krebs K, Pfeil EM, Simon K, Grundmann M, Häberlein F, Bautista-Aguilera OM, Gütschow M, Weaver CD, Fleischmann BK, Kostenis E. Label-Free Whole Cell Biosensing for High-Throughput Discovery of Activators and Inhibitors Targeting G Protein-Activated Inwardly Rectifying Potassium Channels. ACS OMEGA 2018; 3:14814-14823. [PMID: 30555990 PMCID: PMC6289404 DOI: 10.1021/acsomega.8b02254] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/22/2018] [Indexed: 06/09/2023]
Abstract
Dynamic mass redistribution (DMR) and cellular dielectric spectroscopy (CDS) are label-free biosensor technologies that capture real-time integrated cellular responses upon exposure to extra- and intracellular stimuli. They register signaling routes that are accompanied by cell shape changes and/or molecular movement of cells proximal to the biosensor to which they are attached. Here, we report the unexpected observation that robust DMR and CDS signatures are also elicited upon direct stimulation of G protein-activated inwardly rectifying potassium (GIRK) channels, which are involved in the regulation of excitability in the heart and brain. Using ML297, a small-molecule GIRK activator, along with channel blockers and cytoskeletal network inhibitors, we found that GIRK activation exerts its effects on cell shape by a mechanism which depends on actin but not the microtubule network. Because label-free real-time biosensing (i) quantitatively determines concentration dependency of GIRK activators, (ii) accurately assesses the impact of GIRK channel blockers, (iii) is high throughput-compatible, and (iv) visualizes previously unknown cellular consequences downstream of direct GIRK activation, we do not only provide a novel experimental strategy for identification of GIRK ligands but also an entirely new angle to probe GIRK (ligand) biology. We envision that DMR and CDS may add to the repertoire of technologies for systematic exploitation of ion channel function and, in turn, to the identification of novel GIRK ligands in order to treat cardiovascular and neurological disorders.
Collapse
Affiliation(s)
- Katrin
M. Krebs
- Molecular,
Cellular and Pharmacobiology Section, Institute for Pharmaceutical
Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
- Research
Training Group 1873, University of Bonn, Bonn, Germany
| | - Eva M. Pfeil
- Molecular,
Cellular and Pharmacobiology Section, Institute for 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 for Pharmaceutical
Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Manuel Grundmann
- Molecular,
Cellular and Pharmacobiology Section, Institute for Pharmaceutical
Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Felix Häberlein
- Molecular,
Cellular and Pharmacobiology Section, Institute for Pharmaceutical
Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | - Oscar M. Bautista-Aguilera
- Pharmaceutical
Chemistry I, Institute of Pharmacy, University
of Bonn, An der Immenburg
4, 53121 Bonn, Germany
| | - Michael Gütschow
- Pharmaceutical
Chemistry I, Institute of Pharmacy, University
of Bonn, An der Immenburg
4, 53121 Bonn, Germany
| | - C. David Weaver
- Vanderbilt
Institute of Chemical Biology, Department of Pharmacology and Department
of Chemistry, Vanderbilt University, Nashville, 37232 Tennessee, United States
| | - Bernd K. Fleischmann
- Institute
of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Sigmund-Freud-Straße 25, 53105 Bonn, Germany
| | - Evi Kostenis
- Molecular,
Cellular and Pharmacobiology Section, Institute for Pharmaceutical
Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| |
Collapse
|
62
|
Szekacs I, Farkas E, Gemes BL, Takacs E, Szekacs A, Horvath R. Integrin targeting of glyphosate and its cell adhesion modulation effects on osteoblastic MC3T3-E1 cells revealed by label-free optical biosensing. Sci Rep 2018; 8:17401. [PMID: 30479368 PMCID: PMC6258691 DOI: 10.1038/s41598-018-36081-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 10/26/2018] [Indexed: 12/14/2022] Open
Abstract
This study is a discovery of interesting and far reaching properties of the world leading herbicide active ingredient glyphosate. Here we demonstrate the cell adhesion-modifying characteristics of glyphosate affecting cellular interactions via Arg-Gly-Asp (RGD)-dependent integrins. This conclusion was supported by the observations that a glyphosate surface coating induced integrin-specific cell adhesion, while glyphosate in solution inhibited cell adhesion on an RGD-displaying surface. A sensitive, real-time, label-free, whole cell approach was used to monitor the cell adhesion kinetic processes with excellent data quality. The half maximal inhibitory concentration (IC50) for glyphosate was determined to be 0.47 ± 0.07% (20.6 mM) in serum-free conditions. A three-dimensional dissociation constant of 0.352 mM was calculated for the binding between RGD-specific integrins in intact MC3T3-E1 cells and soluble glyphosate by measuring its competition for RGD-motifs binding, while the affinity of those RGD-specific integrins to the RGD-motifs was 5.97 µM. The integrin-targeted affinity of glyphosate was proven using competitive binding assays to recombinant receptor αvβ3. The present study shows not only ligand-binding properties of glyphosate, but also illustrates its remarkable biomimetic power in the case of cell adhesion.
Collapse
Affiliation(s)
- Inna Szekacs
- Nanobiosensorics Momentum Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Hungarian Academy of Sciences, Konkoly-Thege M. út 29-33, H-1120, Budapest, Hungary
| | - Eniko Farkas
- Nanobiosensorics Momentum Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Hungarian Academy of Sciences, Konkoly-Thege M. út 29-33, H-1120, Budapest, Hungary
- Subdoctoral School of Molecular and Nanotechnologies, Chemical Engineering and Material Science Doctoral School, University of Pannonia, Egyetem u.10, H-8200, Veszprém, Hungary
| | - Borbala Leticia Gemes
- Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre, Herman Ottó u. 15, H-1022, Budapest, Hungary
| | - Eszter Takacs
- Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre, Herman Ottó u. 15, H-1022, Budapest, Hungary
| | - Andras Szekacs
- Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre, Herman Ottó u. 15, H-1022, Budapest, Hungary.
| | - Robert Horvath
- Nanobiosensorics Momentum Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Hungarian Academy of Sciences, Konkoly-Thege M. út 29-33, H-1120, Budapest, Hungary.
| |
Collapse
|
63
|
Piccinno MS, Petrachi T, Resca E, Strusi V, Bergamini V, Mulas GA, Mari G, Dominici M, Veronesi E. Label-free toxicology screening of primary human mesenchymal cells and iPS-derived neurons. PLoS One 2018; 13:e0201671. [PMID: 30180158 PMCID: PMC6122932 DOI: 10.1371/journal.pone.0201671] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/19/2018] [Indexed: 11/21/2022] Open
Abstract
The high-throughput, label-free Corning Epic assay has applications in drug discovery, pharmacogenomics, cell receptor signaling, cell migration, and viral titration. The utility of Epic technology for biocompatibility testing has not been well established. In manufacturing of medical devices, in vitro and in vivo biocompatibility assessments are mandatory, according to ISO 10993. The new medical device regulation MDR 745/2017 specifies that ex vivo assays that can closely recapitulate in vivo scenarios are needed to better evaluate biomedical devices. We propose herein that Epic technology—which enables detection of variations in cell mass distribution—is suitable for biocompatibility screening of compounds. In this study, we challenged primary human osteoblasts, endothelial cells, and neurons derived from induced pluripotent stem cells with specific concentrations of methyl methacrylate (MMA). Polymeric MMA has long been applied in cranioplasty, where it makes contact with multiple cell types. Application of Epic technology yielded real-time cytotoxicity profiles for all considered cell types. The results were compared with those from microscopic observation of the same culture plate used in the Epic analyses. The Epic assay should be further examined for its utility for cell biology, genomics, and proteomics companion assays. Our results suggest that Epic technology can be applied to biocompatibility evaluation of human cells in medical device development.
Collapse
Affiliation(s)
- Maria Serena Piccinno
- Science & Technology Park for Medicine (TPM), Mirandola, Italy
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
- * E-mail: (EV); (MSP)
| | - Tiziana Petrachi
- Science & Technology Park for Medicine (TPM), Mirandola, Italy
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Resca
- Science & Technology Park for Medicine (TPM), Mirandola, Italy
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | | | - Valentina Bergamini
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | | | - Giorgio Mari
- Science & Technology Park for Medicine (TPM), Mirandola, Italy
| | - Massimo Dominici
- Science & Technology Park for Medicine (TPM), Mirandola, Italy
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Elena Veronesi
- Science & Technology Park for Medicine (TPM), Mirandola, Italy
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
- * E-mail: (EV); (MSP)
| |
Collapse
|
64
|
Farkas E, Szekacs A, Kovacs B, Olah M, Horvath R, Szekacs I. Label-free optical biosensor for real-time monitoring the cytotoxicity of xenobiotics: A proof of principle study on glyphosate. JOURNAL OF HAZARDOUS MATERIALS 2018; 351:80-89. [PMID: 29518655 DOI: 10.1016/j.jhazmat.2018.02.045] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 02/08/2018] [Accepted: 02/23/2018] [Indexed: 12/17/2023]
Abstract
Rapid and inexpensive biosensor technologies allowing real-time analysis of biomolecular and cellular events have become the basis of next-generation cell-based screening techniques. Our work opens up novel opportunities in the application of the high-throughput label-free Epic BenchTop optical biosensor in cell toxicity studies. The Epic technology records integrated cellular responses about changes in cell morphology and dynamic mass redistribution of cellular contents at the 100-150 nm layer above the sensor surface. The aim of the present study was to apply this novel technology to identify the effect of the herbicide Roundup Classic, its co-formulant polyethoxylated tallow amine (POEA), and its active ingredient glyphosate, on MC3T3-E1 cells adhered on the biosensor surface. The half maximal inhibitory concentrations of Roundup Classic, POEA and glyphosate upon 1 h of exposure were found to be 0.024%, 0.021% and 0.163% in serum-containing medium and 0.028%, 0.019% and 0.538% in serum-free conditions, respectively (at concentrations equivalent to the diluted Roundup solution). These results showed a good correlation with parallel end-point assays, demonstrating the outstanding utility of the Epic technique in cytotoxicity screening, allowing not only high-throughput, real-time detection, but also reduced assay run time and cytotoxicity assessment at end-points far before cell death would occur.
Collapse
Affiliation(s)
- Eniko Farkas
- Nanobiosensorics Momentum Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Hungarian Academy of Sciences, Konkoly-Thege M. út 29-33, H-1120 Budapest, Hungary; Subdoctoral School of Molecular and Nanotechnologies, Chemical Engineering and Material Science Doctoral School, University of Pannonia, Egyetem u.10, H-8200 Veszprém, Hungary
| | - Andras Szekacs
- Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre, Herman Ottó u. 15, H-1022 Budapest, Hungary
| | - Boglarka Kovacs
- Nanobiosensorics Momentum Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Hungarian Academy of Sciences, Konkoly-Thege M. út 29-33, H-1120 Budapest, Hungary; Subdoctoral School of Molecular and Nanotechnologies, Chemical Engineering and Material Science Doctoral School, University of Pannonia, Egyetem u.10, H-8200 Veszprém, Hungary
| | - Marianna Olah
- Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre, Herman Ottó u. 15, H-1022 Budapest, Hungary; Doctoral School of Environmental Sciences, Szent István University, Páter K. u.1, H-2100 Gödöllő, Hungary
| | - Robert Horvath
- Nanobiosensorics Momentum Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Hungarian Academy of Sciences, Konkoly-Thege M. út 29-33, H-1120 Budapest, Hungary.
| | - Inna Szekacs
- Nanobiosensorics Momentum Group, Institute of Technical Physics and Materials Science, Centre for Energy Research, Hungarian Academy of Sciences, Konkoly-Thege M. út 29-33, H-1120 Budapest, Hungary.
| |
Collapse
|
65
|
Li Z, Zhu C, Guo Z, Wang B, Wu X, Fei Y. Highly Sensitive Label-Free Detection of Small Molecules with an Optofluidic Microbubble Resonator. MICROMACHINES 2018; 9:mi9060274. [PMID: 30424207 PMCID: PMC6187366 DOI: 10.3390/mi9060274] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 05/20/2018] [Accepted: 05/29/2018] [Indexed: 01/29/2023]
Abstract
The detection of small molecules has increasingly attracted the attention of researchers because of its important physiological function. In this manuscript, we propose a novel optical sensor which uses an optofluidic microbubble resonator (OFMBR) for the highly sensitive detection of small molecules. This paper demonstrates the binding of the small molecule biotin to surface-immobilized streptavidin with a detection limit reduced to 0.41 pM. Furthermore, binding specificity of four additional small molecules to surface-immobilized streptavidin is shown. A label-free OFMBR-based optical sensor has great potential in small molecule detection and drug screening because of its high sensitivity, low detection limit, and minimal sample consumption.
Collapse
Affiliation(s)
- Zihao Li
- Department of Optical Science and Engineering, Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Fudan University, Shanghai 200433, China.
| | - Chenggang Zhu
- Department of Optical Science and Engineering, Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Fudan University, Shanghai 200433, China.
| | - Zhihe Guo
- Department of Optical Science and Engineering, Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Fudan University, Shanghai 200433, China.
| | - Bowen Wang
- Department of Optical Science and Engineering, Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Fudan University, Shanghai 200433, China.
| | - Xiang Wu
- Department of Optical Science and Engineering, Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Fudan University, Shanghai 200433, China.
| | - Yiyan Fei
- Department of Optical Science and Engineering, Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Fudan University, Shanghai 200433, China.
| |
Collapse
|
66
|
Peter B, Ungai-Salanki R, Szabó B, Nagy AG, Szekacs I, Bősze S, Horvath R. High-Resolution Adhesion Kinetics of EGCG-Exposed Tumor Cells on Biomimetic Interfaces: Comparative Monitoring of Cell Viability Using Label-Free Biosensor and Classic End-Point Assays. ACS OMEGA 2018; 3:3882-3891. [PMID: 29732447 PMCID: PMC5928488 DOI: 10.1021/acsomega.7b01902] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/05/2018] [Indexed: 05/25/2023]
Abstract
A high-throughput label-free resonant waveguide grating biosensor, the Epic BenchTop, was utilized to in situ monitor the adhesion process of cancer cells on Arg-Gly-Asp tripeptide displaying biomimetic polymer surfaces. Using highly adherent human cervical adenocarcinoma (HeLa) cells as a model system, cell adhesion kinetic data with outstanding temporal resolution were obtained. We found that pre-exposing the cells to various concentrations of the main extract of green tea, the (-)-epigallocatechin gallate (EGCG), largely affected the temporal evolution of the adhesion process. For unexposed and low dosed cells, sigmoid shaped spreading kinetics was recorded. Higher dose of EGCG resulted in a complete absence of the sigmoidal character, and displayed adsorption-like kinetics. By using the first derivatives of the kinetic curves, a simple model was developed to quantify the sigmoidal character and the transition from sigmoidal to adsorption-like kinetics. The calculations showed that the transition happened at EGCG concentration of around 60 μg/mL. Using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide end-point assay, we concluded that EGCG is cytostatic but not cytotoxic. The effect of EGCG was also characterized by flow cytometry. We concluded that, using the introduced label-free methodology, the shape of the cell adhesion kinetic curves can be used to quantify in vitro cell viability in a fast, cost-effective, and highly sensitive manner.
Collapse
Affiliation(s)
- Beatrix Peter
- Doctoral
School of Molecular and Nanotechnologies, Faculty of Information Technology, University of Pannonia, Egyetem utca 10, H-8200 Veszprém, Hungary
- Nanobiosensorics
Group, Hungarian Academy of Sciences, Research Centre for Natural
Sciences, Institute for Technical Physics
and Materials Science, Konkoly-Thege M. út 29-33, H-1120 Budapest, Hungary
| | - Rita Ungai-Salanki
- Nanobiosensorics
Group, Hungarian Academy of Sciences, Research Centre for Natural
Sciences, Institute for Technical Physics
and Materials Science, Konkoly-Thege M. út 29-33, H-1120 Budapest, Hungary
- Department
of Biological Physics, Eötvös
Loránd University, Pázmány P. sétány 1/A, H-1117 Budapest, Hungary
- CellSorter
Company for Innovations, Erdőalja út 174, H-1037 Budapest, Hungary
| | - Bálint Szabó
- Nanobiosensorics
Group, Hungarian Academy of Sciences, Research Centre for Natural
Sciences, Institute for Technical Physics
and Materials Science, Konkoly-Thege M. út 29-33, H-1120 Budapest, Hungary
- Department
of Biological Physics, Eötvös
Loránd University, Pázmány P. sétány 1/A, H-1117 Budapest, Hungary
- CellSorter
Company for Innovations, Erdőalja út 174, H-1037 Budapest, Hungary
| | - Agoston G. Nagy
- Nanobiosensorics
Group, Hungarian Academy of Sciences, Research Centre for Natural
Sciences, Institute for Technical Physics
and Materials Science, Konkoly-Thege M. út 29-33, H-1120 Budapest, Hungary
| | - Inna Szekacs
- Nanobiosensorics
Group, Hungarian Academy of Sciences, Research Centre for Natural
Sciences, Institute for Technical Physics
and Materials Science, Konkoly-Thege M. út 29-33, H-1120 Budapest, Hungary
| | - Szilvia Bősze
- MTA-ELTE
Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös Loránd University, 112, P.O. Box 32, H-1518 Budapest, Hungary
| | - Robert Horvath
- Nanobiosensorics
Group, Hungarian Academy of Sciences, Research Centre for Natural
Sciences, Institute for Technical Physics
and Materials Science, Konkoly-Thege M. út 29-33, H-1120 Budapest, Hungary
| |
Collapse
|
67
|
|
68
|
Pulikkathodi AK, Sarangadharan I, Chen YH, Lee GY, Chyi JI, Lee GB, Wang YL. Dynamic monitoring of transmembrane potential changes: a study of ion channels using an electrical double layer-gated FET biosensor. LAB ON A CHIP 2018; 18:1047-1056. [PMID: 29488525 DOI: 10.1039/c7lc01305a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
In this research, we have designed, fabricated and characterized an electrical double layer (EDL)-gated AlGaN/GaN high electron mobility transistor (HEMT) biosensor array to study the transmembrane potential changes of cells. The sensor array platform is designed to detect and count circulating tumor cells (CTCs) of colorectal cancer (CRC) and investigate cellular bioelectric signals. Using the EDL FET biosensor platform, cellular responses can be studied in physiological salt concentrations, thereby eliminating complex automation. Upon investigation, we discovered that our sensor response follows the transmembrane potential changes of captured cells. Our whole cell sensor platform can be used to monitor the dynamic changes in the membrane potential of cells. The effects of continuously changing electrolyte ion concentrations and ion channel blocking using cadmium are investigated. This methodology has the potential to be used as an electrophysiological probe for studying ion channel gating and the interaction of biomolecules in cells. The sensor can also be a point-of-care diagnostic tool for rapid screening of diseases.
Collapse
Affiliation(s)
- Anil Kumar Pulikkathodi
- Institute of NanoEngineering and MicroSystems, National Tsing Hua University, Hsinchu, Taiwan 300, R.O.C.
| | | | | | | | | | | | | |
Collapse
|
69
|
Söllradl T, Banville FA, Fröhlich U, Canva M, Charette PG, Grandbois M. Label-free visualization and quantification of single cell signaling activity using metal-clad waveguide (MCWG)-based microscopy. Biosens Bioelectron 2018; 100:429-436. [DOI: 10.1016/j.bios.2017.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/01/2017] [Accepted: 09/01/2017] [Indexed: 01/01/2023]
|
70
|
Lack of beta-arrestin signaling in the absence of active G proteins. Nat Commun 2018; 9:341. [PMID: 29362459 PMCID: PMC5780443 DOI: 10.1038/s41467-017-02661-3] [Citation(s) in RCA: 284] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
G protein-independent, arrestin-dependent signaling is a paradigm that broadens the signaling scope of G protein-coupled receptors (GPCRs) beyond G proteins for numerous biological processes. However, arrestin signaling in the collective absence of functional G proteins has never been demonstrated. Here we achieve a state of “zero functional G” at the cellular level using HEK293 cells depleted by CRISPR/Cas9 technology of the Gs/q/12 families of Gα proteins, along with pertussis toxin-mediated inactivation of Gi/o. Together with HEK293 cells lacking β-arrestins (“zero arrestin”), we systematically dissect G protein- from arrestin-driven signaling outcomes for a broad set of GPCRs. We use biochemical, biophysical, label-free whole-cell biosensing and ERK phosphorylation to identify four salient features for all receptors at “zero functional G”: arrestin recruitment and internalization, but—unexpectedly—complete failure to activate ERK and whole-cell responses. These findings change our understanding of how GPCRs function and in particular of how they activate ERK1/2. Arrestins terminate signaling from GPCRs, but several lines of evidence suggest that they are also able to transduce signals independently of G proteins. Here, the authors systematically ablate G proteins in cell lines, and show that arrestins are unable to act as genuine signal initiators.
Collapse
|
71
|
Chen JY, Penn LS, Xi J. Quartz crystal microbalance: Sensing cell-substrate adhesion and beyond. Biosens Bioelectron 2018; 99:593-602. [DOI: 10.1016/j.bios.2017.08.032] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/03/2017] [Accepted: 08/12/2017] [Indexed: 10/19/2022]
|
72
|
Söllradl T, Chabot K, Fröhlich U, Canva M, Charette PG, Grandbois M. Monitoring individual cell-signaling activity using combined metal-clad waveguide and surface-enhanced fluorescence imaging. Analyst 2018; 143:5559-5567. [DOI: 10.1039/c8an00911b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Validation of a combined metal-clad waveguide and surface enhanced fluorescence imaging platform for live cell imaging.
Collapse
Affiliation(s)
- Thomas Söllradl
- Laboratoire Nanotechnologies Nanosystèmes (LN2) – CNRS UMI-3463
- Université de Sherbrooke
- Canada
- Institut Interdisciplinaire d'Innovation Technologique (3IT)
- Université de Sherbrooke
| | - Kevin Chabot
- Laboratoire Nanotechnologies Nanosystèmes (LN2) – CNRS UMI-3463
- Université de Sherbrooke
- Canada
- Institut Interdisciplinaire d'Innovation Technologique (3IT)
- Université de Sherbrooke
| | - Ulrike Fröhlich
- Département de Pharmacologie et Physiologie
- Université de Sherbrooke
- Canada
| | - Michael Canva
- Laboratoire Nanotechnologies Nanosystèmes (LN2) – CNRS UMI-3463
- Université de Sherbrooke
- Canada
- Institut Interdisciplinaire d'Innovation Technologique (3IT)
- Université de Sherbrooke
| | - Paul G. Charette
- Laboratoire Nanotechnologies Nanosystèmes (LN2) – CNRS UMI-3463
- Université de Sherbrooke
- Canada
- Institut Interdisciplinaire d'Innovation Technologique (3IT)
- Université de Sherbrooke
| | - Michel Grandbois
- Laboratoire Nanotechnologies Nanosystèmes (LN2) – CNRS UMI-3463
- Université de Sherbrooke
- Canada
- Département de Pharmacologie et Physiologie
- Université de Sherbrooke
| |
Collapse
|
73
|
Affiliation(s)
- Sonja M. Weiz
- Institute for Integrative Nanosciences (IIN); IFW Dresden; Helmholtzstraße 20 01069 Dresden Germany
| | - Mariana Medina-Sánchez
- Institute for Integrative Nanosciences (IIN); IFW Dresden; Helmholtzstraße 20 01069 Dresden Germany
| | - Oliver G. Schmidt
- Institute for Integrative Nanosciences (IIN); IFW Dresden; Helmholtzstraße 20 01069 Dresden Germany
- Material Systems for Nanoelectronics; Chemnitz University of Technology; Reichenhainer Straße 70 09107 Chemnitz Germany
| |
Collapse
|
74
|
Grundmann M, Kostenis E. Holistic Methods for the Analysis of cNMP Effects. Handb Exp Pharmacol 2017; 238:339-357. [PMID: 26721676 DOI: 10.1007/164_2015_42] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Cyclic nucleotide monophosphates (cNMPs) typify the archetype second messenger in living cells and serve as molecular switches with broad functionality. cAMP and cGMP are the best-described cNMPs; however, there is a growing body of evidence indicating that also cCMP and cUMP play a substantial role in signal transduction. Despite research efforts, to date, relatively little is known about the biology of these noncanonical cNMPs, which is due, at least in part, to methodological issues in the past entailing setbacks of the entire field. Only recently, with the use of state-of-the-art techniques, it was possible to revive noncanonical cNMP research. While high-sensitive detection methods disclosed relevant levels of cCMP and cUMP in mammalian cells, knowledge about the biological effectors and their physiological interplay is still incomplete. Holistic biophysical readouts capture cell responses label-free and in an unbiased fashion with the advantage to detect concealed aspects of cell signaling that are arduous to access via traditional biochemical assay approaches. In this chapter, we introduce the dynamic mass redistribution (DMR) technology to explore cell signaling beyond established receptor-controlled mechanisms. Both common and distinctive features in the signaling structure of cCMP and cUMP were identified. Moreover, the integrated response of whole live cells revealed a hitherto undisclosed additional effector of the noncanonical cNMPs. Future studies will show how holistic methods will become integrated into the methodological arsenal of contemporary cNMP research.
Collapse
Affiliation(s)
- Manuel Grundmann
- Molecular-, Cellular- and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany.
| | - Evi Kostenis
- Molecular-, Cellular- and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| |
Collapse
|
75
|
Lirtsman V, Golosovsky M, Davidov D. Surface plasmon excitation using a Fourier-transform infrared spectrometer: Live cell and bacteria sensing. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2017; 88:103105. [PMID: 29092505 DOI: 10.1063/1.4997388] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
We report an accessory for beam collimation to be used as a plug-in for a conventional Fourier-Transform Infrared (FTIR) spectrometer. The beam collimator makes use of the built-in focusing mirror of the FTIR spectrometer which focuses the infrared beam onto the pinhole mounted in the place usually reserved for the sample. The beam is collimated by a small parabolic mirror and is redirected to the sample by a pair of plane mirrors. The reflected beam is conveyed by another pair of plane mirrors to the built-in detector of the FTIR spectrometer. This accessory is most useful for the surface plasmon excitation. We demonstrate how it can be employed for label-free and real-time sensing of dynamic processes in bacterial and live cell layers. In particular, by measuring the intensity of the CO2 absorption peak one can assess the cell layer metabolism, while by measuring the position of the surface plasmon resonance one assesses the cell layer morphology.
Collapse
Affiliation(s)
- Vladislav Lirtsman
- The Racah Institute of Physics, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
| | - Michael Golosovsky
- The Racah Institute of Physics, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
| | - Dan Davidov
- The Racah Institute of Physics, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
| |
Collapse
|
76
|
Bosma R, Witt G, Vaas LAI, Josimovic I, Gribbon P, Vischer HF, Gul S, Leurs R. The Target Residence Time of Antihistamines Determines Their Antagonism of the G Protein-Coupled Histamine H1 Receptor. Front Pharmacol 2017; 8:667. [PMID: 29033838 PMCID: PMC5627017 DOI: 10.3389/fphar.2017.00667] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/07/2017] [Indexed: 11/13/2022] Open
Abstract
The pharmacodynamics of drug-candidates is often optimized by metrics that describe target binding (Kd or Ki value) or target modulation (IC50). However, these metrics are determined at equilibrium conditions, and consequently information regarding the onset and offset of target engagement and modulation is lost. Drug-target residence time is a measure for the lifetime of the drug-target complex, which has recently been receiving considerable interest, as target residence time is shown to have prognostic value for the in vivo efficacy of several drugs. In this study, we have investigated the relation between the increased residence time of antihistamines at the histamine H1 receptor (H1R) and the duration of effective target-inhibition by these antagonists. Hela cells, endogenously expressing low levels of the H1R, were incubated with a series of antihistamines and dissociation was initiated by washing away the unbound antihistamines. Using a calcium-sensitive fluorescent dye and a label free, dynamic mass redistribution based assay, functional recovery of the H1R responsiveness was measured by stimulating the cells with histamine over time, and the recovery was quantified as the receptor recovery time. Using these assays, we determined that the receptor recovery time for a set of antihistamines differed more than 40-fold and was highly correlated to their H1R residence times, as determined with competitive radioligand binding experiments to the H1R in a cell homogenate. Thus, the receptor recovery time is proposed as a cell-based and physiologically relevant metric for the lead optimization of G protein-coupled receptor antagonists, like the H1R antagonists. Both, label-free or real-time, classical signaling assays allow an efficient and physiologically relevant determination of kinetic properties of drug molecules.
Collapse
Affiliation(s)
- Reggie Bosma
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Lea A I Vaas
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Ivana Josimovic
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Philip Gribbon
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Henry F Vischer
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening PortHamburg, Germany
| | - Rob Leurs
- Amsterdam Institute for Molecules, Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University AmsterdamAmsterdam, Netherlands
| |
Collapse
|
77
|
Antibiotic-induced release of small extracellular vesicles (exosomes) with surface-associated DNA. Sci Rep 2017; 7:8202. [PMID: 28811610 PMCID: PMC5557920 DOI: 10.1038/s41598-017-08392-1] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 07/10/2017] [Indexed: 02/08/2023] Open
Abstract
Recently, biological roles of extracellular vesicles (which include among others exosomes, microvesicles and apoptotic bodies) have attracted substantial attention in various fields of biomedicine. Here we investigated the impact of sustained exposure of cells to the fluoroquinolone antibiotic ciprofloxacin on the released extracellular vesicles. Ciprofloxacin is widely used in humans against bacterial infections as well as in cell cultures against Mycoplasma contamination. However, ciprofloxacin is an inducer of oxidative stress and mitochondrial dysfunction of mammalian cells. Unexpectedly, here we found that ciprofloxacin induced the release of both DNA (mitochondrial and chromosomal sequences) and DNA-binding proteins on the exofacial surfaces of small extracellular vesicles referred to in this paper as exosomes. Furthermore, a label-free optical biosensor analysis revealed DNA-dependent binding of exosomes to fibronectin. DNA release on the surface of exosomes was not affected any further by cellular activation or apoptosis induction. Our results reveal for the first time that prolonged low-dose ciprofloxacin exposure leads to the release of DNA associated with the external surface of exosomes.
Collapse
|
78
|
Kaminski T, Geschwindner S. Perspectives on optical biosensor utility in small-molecule screening. Expert Opin Drug Discov 2017; 12:1083-1086. [PMID: 28777014 DOI: 10.1080/17460441.2017.1364727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Tim Kaminski
- a Discovery Sciences, Innovative Medicines and Early Development Biotech Unit , AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Stefan Geschwindner
- a Discovery Sciences, Innovative Medicines and Early Development Biotech Unit , AstraZeneca R&D Gothenburg , Mölndal , Sweden
| |
Collapse
|
79
|
Christensen HB, Gloriam DE, Pedersen DS, Cowland JB, Borregaard N, Bräuner-Osborne H. Applying label-free dynamic mass redistribution assay for studying endogenous FPR1 receptor signalling in human neutrophils. J Pharmacol Toxicol Methods 2017; 88:72-78. [PMID: 28716665 DOI: 10.1016/j.vascn.2017.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/02/2017] [Accepted: 07/13/2017] [Indexed: 11/18/2022]
Abstract
INTRODUCTION The label-free dynamic mass redistribution-based assay (DMR) is a powerful method for studying signalling pathways of G protein-coupled receptors (GPCRs). Herein we present the label-free DMR assay as a robust readout for pharmacological characterization of formyl peptide receptors (FPRs) in human neutrophils. METHODS Neutrophils were isolated from fresh human blood and their responses to FPR1 and FPR2 agonists, i.e. compound 43, fMLF and WKYMVm were measured in a label-free DMR assay using Epic Benchtop System from Corning®. Obtained DMR traces were used to calculate agonist potencies. RESULTS The potencies (pEC50) of fMLF, WKYMVm and compound 43, determined on human neutrophils using the label-free DMR assay were 8.63, 7.76 and 5.92, respectively. The DMR response to fMLF, but not WKYMVm and compound 43 could be blocked by the FPR1-specific antagonist cyclosporin H. DISCUSSION We conclude that the DMR assay can be used, and complements more traditional methods, to study the signalling and pharmacology of endogenous FPR receptors in human neutrophils.
Collapse
Affiliation(s)
- Hanna B Christensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| | - David E Gloriam
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| | - Daniel Sejer Pedersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| | - Jack B Cowland
- Granulocyte Research Laboratory, Department of Hematology, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Niels Borregaard
- Granulocyte Research Laboratory, Department of Hematology, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
80
|
Leonard H, Halachmi S, Ben-Dov N, Nativ O, Segal E. Unraveling Antimicrobial Susceptibility of Bacterial Networks on Micropillar Architectures Using Intrinsic Phase-Shift Spectroscopy. ACS NANO 2017; 11:6167-6177. [PMID: 28485961 DOI: 10.1021/acsnano.7b02217] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
With global antimicrobial resistance becoming increasingly detrimental to society, improving current clinical antimicrobial susceptibility testing (AST) is crucial to allow physicians to initiate appropriate antibiotic treatment as early as possible, reducing not only mortality rates but also the emergence of resistant pathogens. In this work, we tackle the main bottlenecks in clinical AST by designing biofunctionalized silicon micropillar arrays to provide both a preferable solid-liquid interface for bacteria networking and a simultaneous transducing element that monitors the response of bacteria when exposed to chosen antibiotics in real time. We harness the intrinsic ability of the micropillar architectures to relay optical phase-shift reflectometric interference spectroscopic measurements (referred to as PRISM) and employ it as a platform for culture-free, label-free phenotypic AST. The responses of E. coli to various concentrations of five clinically relevant antibiotics are optically tracked by PRISM, allowing for the minimum inhibitory concentration (MIC) values to be determined and compared to both standard broth microdilution testing and clinic-based automated AST system readouts. Capture of bacteria within these microtopologies, followed by incubation of the cells with the appropriate antibiotic solution, yields rapid determinations of antibiotic susceptibility. This platform not only provides accurate MIC determinations in a rapid manner (total assay time of 2-3 h versus 8 h with automated AST systems) but can also be employed as an advantageous method to differentiate bacteriostatic and bactericidal antibiotics.
Collapse
Affiliation(s)
- Heidi Leonard
- Department of Biotechnology and Food Engineering, ‡Department of Urology, Bnai Zion Medical Center, Faculty of Medicine, and §The Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Sarel Halachmi
- Department of Biotechnology and Food Engineering, ‡Department of Urology, Bnai Zion Medical Center, Faculty of Medicine, and §The Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Nadav Ben-Dov
- Department of Biotechnology and Food Engineering, ‡Department of Urology, Bnai Zion Medical Center, Faculty of Medicine, and §The Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Ofer Nativ
- Department of Biotechnology and Food Engineering, ‡Department of Urology, Bnai Zion Medical Center, Faculty of Medicine, and §The Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| | - Ester Segal
- Department of Biotechnology and Food Engineering, ‡Department of Urology, Bnai Zion Medical Center, Faculty of Medicine, and §The Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology , Haifa 3200003, Israel
| |
Collapse
|
81
|
Grundmann M. Label-Free Dynamic Mass Redistribution and Bio-Impedance Methods for Drug Discovery. ACTA ACUST UNITED AC 2017. [PMID: 28640952 DOI: 10.1002/cpph.24] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Label-free biosensors are increasingly employed in drug discovery. Cell-based biosensors provide valuable insights into the biological consequences of exposing cells and tissues to chemical agents and the underlying molecular mechanisms associated with these effects. Optical biosensors based on the detection of dynamic mass redistribution (DMR) and impedance biosensors using cellular dielectric spectroscopy (CDS) capture changes of the cytoskeleton of living cells in real time. Because signal transduction correlates with changes in cell morphology, DMR and CDS biosensors are exquisitely suited for recording integrated cell responses in an unbiased, yet pathway-specific manner without the use of labels that may interfere with cell function. Described in this unit are several experimental approaches utilizing optical label-free system capturing dynamic mass redistribution (DMR) in living cells (Epic System) and an impedance-based CDS technology (CellKey). In addition, potential pitfalls associated with these assays and alternative approaches for overcoming such technical challenges are discussed. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Manuel Grundmann
- Section Cellular, Molecular and Pharmacobiology, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| |
Collapse
|
82
|
Bates KE, Lu H. Optics-Integrated Microfluidic Platforms for Biomolecular Analyses. Biophys J 2017; 110:1684-1697. [PMID: 27119629 DOI: 10.1016/j.bpj.2016.03.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 02/19/2016] [Accepted: 03/08/2016] [Indexed: 02/06/2023] Open
Abstract
Compared with conventional optical methods, optics implemented on microfluidic chips provide small, and often much cheaper ways to interrogate biological systems from the level of single molecules up to small model organisms. The optical probing of single molecules has been used to investigate the mechanical properties of individual biological molecules; however, multiplexing of these measurements through microfluidics and nanofluidics confers many analytical advantages. Optics-integrated microfluidic systems can significantly simplify sample processing and allow a more user-friendly experience; alignments of on-chip optical components are predetermined during fabrication and many purely optical techniques are passively controlled. Furthermore, sample loss from complicated preparation and fluid transfer steps can be virtually eliminated, a particularly important attribute for biological molecules at very low concentrations. Excellent fluid handling and high surface area/volume ratios also contribute to faster detection times for low abundance molecules in small sample volumes. Although integration of optical systems with classical microfluidic analysis techniques has been limited, microfluidics offers a ready platform for interrogation of biophysical properties. By exploiting the ease with which fluids and particles can be precisely and dynamically controlled in microfluidic devices, optical sensors capable of unique imaging modes, single molecule manipulation, and detection of minute changes in concentration of an analyte are possible.
Collapse
Affiliation(s)
- Kathleen E Bates
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, Atlanta, Georgia; School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Hang Lu
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, Atlanta, Georgia; School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia.
| |
Collapse
|
83
|
Rawal S, Yang YP, Cote R, Agarwal A. Identification and Quantitation of Circulating Tumor Cells. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2017; 10:321-343. [PMID: 28301753 DOI: 10.1146/annurev-anchem-061516-045405] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Circulating tumor cells (CTCs) are shed from the primary tumor into the circulatory system and act as seeds that initiate cancer metastasis to distant sites. CTC enumeration has been shown to have a significant prognostic value as a surrogate marker in various cancers. The widespread clinical utility of CTC tests, however, is still limited due to the inherent rarity and heterogeneity of CTCs, which necessitate robust techniques for their efficient enrichment and detection. Significant recent advances have resulted in technologies with the ability to improve yield and purity of CTC enrichment as well as detection sensitivity. Current efforts are largely focused on the translation and standardization of assays to fully realize the clinical utility of CTCs. In this review, we aim to provide a comprehensive overview of CTC enrichment and detection techniques with an emphasis on novel approaches for rapid quantification of CTCs.
Collapse
Affiliation(s)
- Siddarth Rawal
- Department of Pathology, DJTMF Biomedical Nanotechnology Institute, University of Miami, Coral Gables, Florida 33146
| | - Yu-Ping Yang
- Department of Pathology, DJTMF Biomedical Nanotechnology Institute, University of Miami, Coral Gables, Florida 33146
- Department of Biochemistry and Molecular Biology, University of Miami, Coral Gables, Florida 33146
| | - Richard Cote
- Department of Pathology, DJTMF Biomedical Nanotechnology Institute, University of Miami, Coral Gables, Florida 33146
- Department of Biochemistry and Molecular Biology, University of Miami, Coral Gables, Florida 33146
| | - Ashutosh Agarwal
- Department of Pathology, DJTMF Biomedical Nanotechnology Institute, University of Miami, Coral Gables, Florida 33146
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida 33146;
| |
Collapse
|
84
|
Hong HJ, Koom WS, Koh WG. Cell Microarray Technologies for High-Throughput Cell-Based Biosensors. SENSORS (BASEL, SWITZERLAND) 2017; 17:E1293. [PMID: 28587242 PMCID: PMC5492771 DOI: 10.3390/s17061293] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/24/2017] [Accepted: 05/31/2017] [Indexed: 12/27/2022]
Abstract
Due to the recent demand for high-throughput cellular assays, a lot of efforts have been made on miniaturization of cell-based biosensors by preparing cell microarrays. Various microfabrication technologies have been used to generate cell microarrays, where cells of different phenotypes are immobilized either on a flat substrate (positional array) or on particles (solution or suspension array) to achieve multiplexed and high-throughput cell-based biosensing. After introducing the fabrication methods for preparation of the positional and suspension cell microarrays, this review discusses the applications of the cell microarray including toxicology, drug discovery and detection of toxic agents.
Collapse
Affiliation(s)
- Hye Jin Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Korea.
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Korea.
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Korea.
| |
Collapse
|
85
|
Hu HH, Deng H, Ling S, Sun H, Kenakin T, Liang X, Fang Y. Chemical genomic analysis of GPR35 signaling. Integr Biol (Camb) 2017; 9:451-463. [PMID: 28425521 DOI: 10.1039/c7ib00005g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
GPR35, a family A orphan G protein-coupled receptor, has been implicated in inflammatory, neurological, and cardiovascular diseases. However, not much is known about the signaling and functions of GPR35. We performed a label-free kinome short hairpin RNA screen and identified a putative signaling network of GPR35 in HT-29 cells, some of which was validated using gene expression, biochemical and cellular assays. The results showed that GPR35 induced hypoxia-inducible factor 1α, and was involved in synaptic transmission, sensory perception, the immune system, and morphogenetic processes. Collectively, our data suggest that GPR35 may play an important role in response to hypoxic stress and be a potential target for the treatment of inflammatory, cardiovascular, and neurological disorders.
Collapse
Affiliation(s)
- Heidi Haibei Hu
- Biochemical Technologies, Corning R&D Corporation, Corning Incorporated, Corning, NY 14831, USA.
| | | | | | | | | | | | | |
Collapse
|
86
|
Harris DA, Park JM, Lee KS, Xu C, Stella N, Hague C. Label-Free Dynamic Mass Redistribution Reveals Low-Density, Prosurvival α1B-Adrenergic Receptors in Human SW480 Colon Carcinoma Cells. J Pharmacol Exp Ther 2017; 361:219-228. [PMID: 28196836 PMCID: PMC5399639 DOI: 10.1124/jpet.116.237255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 02/10/2017] [Indexed: 12/11/2022] Open
Abstract
Small molecules that target the adrenergic family of G protein-coupled receptors (GPCRs) show promising therapeutic efficacy for the treatment of various cancers. In this study, we report that human colon cancer cell line SW480 expresses low-density functional α1B-adrenergic receptors (ARs) as revealed by label-free dynamic mass redistribution (DMR) signaling technology and confirmed by quantitative reverse-transcriptase polymerase chain reaction analysis. Remarkably, although endogenous α1B-ARs are not detectable via either [3H]-prazosin-binding analysis or phosphoinositol hydrolysis assays, their activation leads to robust DMR and enhanced cell viability. We provide pharmacological evidence that stimulation of α1B-ARs enhances SW480 cell viability without affecting proliferation, whereas stimulating β-ARs diminishes both viability and proliferation of SW480 cells. Our study illustrates the power of label-free DMR technology for identifying and characterizing low-density GPCRs in cells and suggests that drugs targeting both α1B- and β-ARs may represent valuable small-molecule therapeutics for the treatment of colon cancer.
Collapse
Affiliation(s)
- Dorathy-Ann Harris
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| | - Ji-Min Park
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| | - Kyung-Soon Lee
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| | - Cong Xu
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| | - Nephi Stella
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| | - Chris Hague
- Departments of Pharmacology (D.-A.H., J.-M.P., K.-S.L., C.X., N.S., C.H.) and Psychiatry and Behavioral Sciences (C.X., N.S.), University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
87
|
Peter B, Farkas E, Forgacs E, Saftics A, Kovacs B, Kurunczi S, Szekacs I, Csampai A, Bosze S, Horvath R. Green tea polyphenol tailors cell adhesivity of RGD displaying surfaces: multicomponent models monitored optically. Sci Rep 2017; 7:42220. [PMID: 28186133 PMCID: PMC5301484 DOI: 10.1038/srep42220] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/03/2017] [Indexed: 01/17/2023] Open
Abstract
The interaction of the anti-adhesive coating, poly(L-lysine)-graft-poly(ethylene glycol) (PLL-g-PEG) and its Arg-Gly-Asp (RGD) functionalized form, PLL-g-PEG-RGD, with the green tea polyphenol, epigallocatechin-gallate (EGCg) was in situ monitored. After, the kinetics of cellular adhesion on the EGCg exposed coatings were recorded in real-time. The employed plate-based waveguide biosensor is applicable to monitor small molecule binding and sensitive to sub-nanometer scale changes in cell membrane position and cell mass distribution; while detecting the signals of thousands of adhering cells. The combination of this remarkable sensitivity and throughput opens up new avenues in testing complicated models of cell-surface interactions. The systematic studies revealed that, despite the reported excellent antifouling properties of the coatings, EGCg strongly interacted with them, and affected their cell adhesivity in a concentration dependent manner. Moreover, the differences between the effects of the fresh and oxidized EGCg solutions were first demonstrated. Using a semiempirical quantumchemical method we showed that EGCg binds to the PEG chains of PLL-g-PEG-RGD and effectively blocks the RGD sites by hydrogen bonds. The calculations supported the experimental finding that the binding is stronger for the oxidative products. Our work lead to a new model of polyphenol action on cell adhesion ligand accessibility and matrix rigidity.
Collapse
Affiliation(s)
- Beatrix Peter
- Doctoral School of Molecular and Nanotechnologies, Faculty of Information Technology, University of Pannonia, H-8200 Egyetem u. 10, Veszprém, Hungary
- Nanobiosensorics Group, Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute for Technical Physics and Materials Science, Konkoly-Thege u, 29-33, H-1120 Budapest, Hungary
| | - Eniko Farkas
- Nanobiosensorics Group, Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute for Technical Physics and Materials Science, Konkoly-Thege u, 29-33, H-1120 Budapest, Hungary
- Chemical Engineering and Material Science Doctoral School, University of Pannonia, H-8200 Egyetem u, 10, Veszprém, Hungary
| | - Eniko Forgacs
- Nanobiosensorics Group, Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute for Technical Physics and Materials Science, Konkoly-Thege u, 29-33, H-1120 Budapest, Hungary
| | - Andras Saftics
- Nanobiosensorics Group, Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute for Technical Physics and Materials Science, Konkoly-Thege u, 29-33, H-1120 Budapest, Hungary
- George Olah Doctoral School, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, Budapest 1111, Hungary
| | - Boglarka Kovacs
- Nanobiosensorics Group, Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute for Technical Physics and Materials Science, Konkoly-Thege u, 29-33, H-1120 Budapest, Hungary
- Chemical Engineering and Material Science Doctoral School, University of Pannonia, H-8200 Egyetem u, 10, Veszprém, Hungary
| | - Sandor Kurunczi
- Nanobiosensorics Group, Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute for Technical Physics and Materials Science, Konkoly-Thege u, 29-33, H-1120 Budapest, Hungary
| | - Inna Szekacs
- Nanobiosensorics Group, Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute for Technical Physics and Materials Science, Konkoly-Thege u, 29-33, H-1120 Budapest, Hungary
| | - Antal Csampai
- Institute of Chemistry, Eötvös Loránd University, Budapest 112, POB 32, H-1518, Hungary
| | - Szilvia Bosze
- MTA-ELTE Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös Loránd University, Budapest 112, POB 32, H-1518, Hungary
| | - Robert Horvath
- Nanobiosensorics Group, Hungarian Academy of Sciences, Research Centre for Natural Sciences, Institute for Technical Physics and Materials Science, Konkoly-Thege u, 29-33, H-1120 Budapest, Hungary
| |
Collapse
|
88
|
Tang W, Huang B, Wang J, An L, Zhong H, Yang H, Li P, Chen J. A label-free screening approach targeted protease-activated receptor 1 based on dynamic mass redistribution in living cells. RSC Adv 2017. [DOI: 10.1039/c7ra07927c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Protease-activated receptor 1 (PAR-1) antagonists strongly inhibit thrombin-induced platelet aggregation and are proved to be effective as anti-thrombotic drugs.
Collapse
Affiliation(s)
- Weiwei Tang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Bixia Huang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Jiancheng Wang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Lin An
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | | | - Hua Yang
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Ping Li
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Jun Chen
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- P. R. China
| |
Collapse
|
89
|
Febles NK, Chandrasekaran S, Fang Y. Resonant Waveguide Grating Imager for Single Cell Monitoring of the Invasion of 3D Speheroid Cancer Cells Through Matrigel. Methods Mol Biol 2017; 1571:143-160. [PMID: 28281255 DOI: 10.1007/978-1-4939-6848-0_10] [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: 08/29/2023]
Abstract
The invasion of cancer cells through their surrounding extracellular matrices is the first critical step to metastasis, a devastating event to cancer patients. However, in vitro cancer cell invasion is mostly studied using two-dimensional (2D) models. Three-dimensional (3D) multicellular spheroids may offer an advantageous cell model for cancer research and oncology drug discovery. This chapter describes a label-free, real-time, and single-cell approach to quantify the invasion of 3D spheroid colon cancer cells through Matrigel using a spatially resolved resonant waveguide grating imager.
Collapse
Affiliation(s)
- Nicole K Febles
- Biochemical Technologies, Corning Research and Development Corporation, Corning Incorporated, Sullivan Park, Corning, NY, 14831, USA
- NanoScience Technology Center, Department of Mechanical, Materials and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Siddarth Chandrasekaran
- Biochemical Technologies, Corning Research and Development Corporation, Corning Incorporated, Sullivan Park, Corning, NY, 14831, USA
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Ye Fang
- Biochemical Technologies, Corning Research and Development Corporation, Corning Incorporated, Sullivan Park, Corning, NY, 14831, USA.
| |
Collapse
|
90
|
Suutari T, Silen T, S En Karaman D, Saari H, Desai D, Kerkelä E, Laitinen S, Hanzlikova M, Rosenholm JM, Yliperttula M, Viitala T. Real-Time Label-Free Monitoring of Nanoparticle Cell Uptake. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:6289-6300. [PMID: 27690329 DOI: 10.1002/smll.201601815] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/28/2016] [Indexed: 05/10/2023]
Abstract
The surface plasmon resonance technique in combination with whole cell sensing is used for the first time for real-time label-free monitoring of nanoparticle cell uptake. The uptake kinetics of several types of nanoparticles relevant to drug delivery applications into HeLa cells is determined. The cell uptake of the nanoparticles is confirmed by confocal microscopy. The cell uptake of silica nanoparticles and polyethylenimine-plasmid DNA polyplexes is studied as a function of temperature, and the uptake energies are determined by Arrhenius plots. The phase transition temperature of the HeLa cell membrane is detected when monitoring cell uptake of silica nanoparticles at different temperatures. The HeLa cell uptake of the mesoporous silica nanoparticles is energy-independent at temperatures slightly higher than the phase transition temperature of the HeLa cell membrane, while the uptake of polyethylenimine-DNA polyplexes is energy-dependent and linear as a function of temperature with an activation energy of Ea = 62 ± 7 kJ mol-1 = 15 ± 2 kcal mol-1 . The HeLa cell uptake of red blood cell derived extracellular vesicles is also studied as a function of the extracellular vesicle concentration. The results show a concentration dependent behavior reaching a saturation level of the extracellular vesicle uptake by HeLa cells.
Collapse
Affiliation(s)
- Teemu Suutari
- Centre for Drug Research at the Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Tiina Silen
- Centre for Drug Research at the Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Didem S En Karaman
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, BioCity 3rd floor, Artillerigatan 6A, 20520, Åbo, Finland
- Centre for Functional Materials, Laboratory for Physical Chemistry, Faculty of Science and Engineering, Åbo Akademi University, Porthansgatan 3-5, 20500, Åbo, Finland
| | - Heikki Saari
- Centre for Drug Research at the Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Diti Desai
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, BioCity 3rd floor, Artillerigatan 6A, 20520, Åbo, Finland
| | - Erja Kerkelä
- Finnish Red Cross Blood Service, Kivihaantie 7, 00310, Helsinki, Finland
| | - Saara Laitinen
- Finnish Red Cross Blood Service, Kivihaantie 7, 00310, Helsinki, Finland
| | - Martina Hanzlikova
- Centre for Drug Research at the Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, BioCity 3rd floor, Artillerigatan 6A, 20520, Åbo, Finland
| | - Marjo Yliperttula
- Centre for Drug Research at the Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via F. Marzolo 5, Padova, Italy
| | - Tapani Viitala
- Centre for Drug Research at the Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| |
Collapse
|
91
|
Kovacs B, Patko D, Szekacs I, Orgovan N, Kurunczi S, Sulyok A, Khanh NQ, Toth B, Vonderviszt F, Horvath R. Flagellin based biomimetic coatings: From cell-repellent surfaces to highly adhesive coatings. Acta Biomater 2016; 42:66-76. [PMID: 27381523 DOI: 10.1016/j.actbio.2016.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 06/28/2016] [Accepted: 07/01/2016] [Indexed: 10/21/2022]
Abstract
UNLABELLED Biomimetic coatings with cell-adhesion-regulating functionalities are intensively researched today. For example, cell-based biosensing for drug development, biomedical implants, and tissue engineering require that the surface adhesion of living cells is well controlled. Recently, we have shown that the bacterial flagellar protein, flagellin, adsorbs through its terminal segments to hydrophobic surfaces, forming an oriented monolayer and exposing its variable D3 domain to the solution. Here, we hypothesized that this nanostructured layer is highly cell-repellent since it mimics the surface of the flagellar filaments. Moreover, we proposed flagellin as a carrier molecule to display the cell-adhesive RGD (Arg-Gly-Asp) peptide sequence and induce cell adhesion on the coated surface. The D3 domain of flagellin was replaced with one or more RGD motifs linked by various oligopeptides modulating flexibility and accessibility of the inserted segment. The obtained flagellin variants were applied to create surface coatings inducing cell adhesion and spreading to different levels, while wild-type flagellin was shown to form a surface layer with strong anti-adhesive properties. As reference surfaces synthetic polymers were applied which have anti-adhesive (PLL-g-PEG poly(l-lysine)-graft-poly(ethylene glycol)) or adhesion inducing properties (RGD-functionalized PLL-g-PEG). Quantitative adhesion data was obtained by employing optical biochips and microscopy. Cell-adhesion-regulating coatings can be simply formed on hydrophobic surfaces by using the developed flagellin-based constructs. The developed novel RGD-displaying flagellin variants can be easily obtained by bacterial production and can serve as alternatives to create cell-adhesion-regulating biomimetic coatings. STATEMENT OF SIGNIFICANCE In the present work, we show for the first time that.
Collapse
|
92
|
Adhesion kinetics of human primary monocytes, dendritic cells, and macrophages: Dynamic cell adhesion measurements with a label-free optical biosensor and their comparison with end-point assays. Biointerphases 2016; 11:031001. [DOI: 10.1116/1.4954789] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
93
|
Kountz TS, Lee KS, Aggarwal-Howarth S, Curran E, Park JM, Harris DA, Stewart A, Hendrickson J, Camp ND, Wolf-Yadlin A, Wang EH, Scott JD, Hague C. Endogenous N-terminal Domain Cleavage Modulates α1D-Adrenergic Receptor Pharmacodynamics. J Biol Chem 2016; 291:18210-21. [PMID: 27382054 DOI: 10.1074/jbc.m116.729517] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Indexed: 01/11/2023] Open
Abstract
The α1D-adrenergic receptor (ADRA1D) is a key regulator of cardiovascular, prostate, and central nervous system functions. This clinically relevant G protein-coupled receptor has proven difficult to study, as it must form an obligate modular homodimer containing the PDZ proteins scribble and syntrophin or become retained in the endoplasmic reticulum as non-functional protein. We previously determined that targeted removal of the N-terminal (NT) 79 amino acids facilitates ADRA1D plasma membrane expression and agonist-stimulated functional responses. However, whether such an event occurs in physiological contexts was unknown. Herein, we report the ADRA1D is subjected to innate NT processing in cultured human cells. SNAP near-infrared imaging and tandem-affinity purification revealed the ADRA1D is expressed as both full-length and NT truncated forms in multiple human cell lines. Serial truncation mapping identified the cleavage site as Leu(90)/Val(91) in the 95-amino acid ADRA1D NT domain, suggesting human cells express a Δ1-91 ADRA1D species. Tandem-affinity purification MS/MS and co-immunoprecipitation analysis indicate NT processing of ADRA1D is not required to form scribble-syntrophin macromolecular complexes. Yet, label-free dynamic mass redistribution signaling assays demonstrate that Δ1-91 ADRA1D agonist responses were greater than WT ADRA1D. Mutagenesis of the cleavage site nullified the processing event, resulting in ADRA1D agonist responses less than the WT receptor. Thus, we propose that processing of the ADRA1D NT domain is a physiological mechanism employed by cells to generate a functional ADRA1D isoform with optimal pharmacodynamic properties.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Nathan D Camp
- Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195
| | - Alejandro Wolf-Yadlin
- Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195
| | | | - John D Scott
- the Departments of Pharmacology and From the Howard Hughes Medical Institute and
| | | |
Collapse
|
94
|
Feng S, Jiang JH, Rashid AA, John S. Biosensor architecture for enhanced disease diagnostics: lab-in-a-photonic-crystal. OPTICS EXPRESS 2016; 24:12166-12191. [PMID: 27410136 DOI: 10.1364/oe.24.012166] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A conceptual lab-in-a-photonic-crystal biosensor is demonstrated that can multiplex four or more distinct disease-markers and distinguish their presence and combinations simultaneously with unique spectral fingerprints. This biosensor consists of a photonic-band-gap, multi-mode waveguide coupled to surface modes on either side, encased in a glass slide with microfluidic channels. The spectral fingerprints consist of multiple peaks in optical transmission vs. frequency that respond sensitively and uniquely in both frequency shift and nonmonotonic change of peak transmittance levels to various analyte bindings. This special property enables complete, logical determination of twelve different combinations of four distinct disease-markers through one scan of the transmission spectrum. The results reveal unique phenomena such as switching between the strong-coupling and weak-coupling combinations of surface states by analyte binding at different locations along the central waveguide. The unconventional transmission spectra are explained using a Landauer-Büttiker, multiple-scattering, transmission theory that reproduces the main features of the exact finite-difference-time-domain simulation.
Collapse
|
95
|
Cunningham B, Zhang M, Zhuo Y, Kwon L, Race C. Recent Advances in Biosensing With Photonic Crystal Surfaces: A Review. IEEE SENSORS JOURNAL 2016; 16:3349-3366. [PMID: 27642265 PMCID: PMC5021450 DOI: 10.1109/jsen.2015.2429738] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Photonic crystal surfaces that are designed to function as wavelength-selective optical resonators have become a widely adopted platform for label-free biosensing, and for enhancement of the output of photon-emitting tags used throughout life science research and in vitro diagnostics. While some applications, such as analysis of drug-protein interactions, require extremely high resolution and the ability to accurately correct for measurement artifacts, others require sensitivity that is high enough for detection of disease biomarkers in serum with concentrations less than 1 pg/ml. As the analysis of cells becomes increasingly important for studying the behavior of stem cells, cancer cells, and biofilms under a variety of conditions, approaches that enable high resolution imaging of live cells without cytotoxic stains or photobleachable fluorescent dyes are providing new tools to biologists who seek to observe individual cells over extended time periods. This paper will review several recent advances in photonic crystal biosensor detection instrumentation and device structures that are being applied towards direct detection of small molecules in the context of high throughput drug screening, photonic crystal fluorescence enhancement as utilized for high sensitivity multiplexed cancer biomarker detection, and label-free high resolution imaging of cells and individual nanoparticles as a new tool for life science research and single-molecule diagnostics.
Collapse
Affiliation(s)
- B.T. Cunningham
- Dept. of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign
| | - M. Zhang
- Dept. of Physics, University of Illinois at Urbana-Champaign
| | - Y. Zhuo
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign
| | - L. Kwon
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign
| | - C. Race
- Dept. of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign
| |
Collapse
|
96
|
Intensity interrogation near cutoff resonance for label-free cellular profiling. Sci Rep 2016; 6:24685. [PMID: 27086879 PMCID: PMC4834563 DOI: 10.1038/srep24685] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/04/2016] [Indexed: 11/08/2022] Open
Abstract
We report a method enabling intensity-based readout for label-free cellular assays, and realize a reader device with the same footprint as a microtiter plate. For unambiguous resonance intensity measurements in resonance waveguide grating (RWG) sensors, we propose to apply resonances near the substrate cutoff wavelength. This method was validated in bulk refractive index, surface bilayer and G protein-coupled receptor (GPCR) experiments. The significantly reduced size of the reader device opens new opportunities for easy integration into incubators or liquid handling systems.
Collapse
|
97
|
ZnO Nanostructure Templates as a Cost-Efficient Mass-Producible Route for the Development of Cellular Networks. MATERIALS 2016; 9:ma9040256. [PMID: 28773382 PMCID: PMC5502920 DOI: 10.3390/ma9040256] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 03/18/2016] [Accepted: 03/18/2016] [Indexed: 01/18/2023]
Abstract
The development of artificial surfaces which can regulate or trigger specific functions of living cells, and which are capable of inducing in vivo-like cell behaviors under in vitro conditions has been a long-sought goal over the past twenty years. In this work, an alternative, facile and cost-efficient method for mass-producible cellular templates is presented. The proposed methodology consists of a cost-efficient, two-step, all-wet technique capable of producing ZnO-based nanostructures on predefined patterns on a variety of substrates. ZnO—apart from the fact that it is a biocompatible material—was chosen because of its multifunctional nature which has rendered it a versatile material employed in a wide range of applications. Si, Si3N4, emulated microelectrode arrays and conventional glass cover slips were patterned at the micrometer scale and the patterns were filled with ZnO nanostructures. Using HeLa cells, we demonstrated that the fabricated nanotopographical features could promote guided cellular adhesion on the pre-defined micron-scale patterns only through nanomechanical cues without the need for further surface activation or modification. The basic steps of the micro/nanofabrication are presented and the results from the cell adhesion experiments are discussed, showing the potential of the suggested methodology for creating low-cost templates for engineered cellular networks.
Collapse
|
98
|
Integrating biophysics with HTS-driven drug discovery projects. Drug Discov Today 2016; 21:491-8. [PMID: 26826422 DOI: 10.1016/j.drudis.2016.01.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 01/04/2016] [Accepted: 01/20/2016] [Indexed: 12/13/2022]
Abstract
Over the past decade biophysics has become an established discipline in HTS hit triaging, owing to its high fidelity in detecting protein-ligand interactions. Many pharma companies are using biophysical techniques to filter HTS output for false positives, as will be discussed in this review. Moreover, I will demonstrate how the earlier application of biophysics, already at the HTS assay development stage, is potentially even more impactful. Two key areas here are early mode-of-action studies and ensuring that the HTS assay and subsequent cascade are fit for purpose. Top-level results from 20 in-house projects are shown to underpin the impact of these studies.
Collapse
|
99
|
Lowry TW, Hariri H, Prommapan P, Kusi-Appiah A, Vafai N, Bienkiewicz EA, Van Winkle DH, Stagg SM, Lenhert S. Quantification of Protein-Induced Membrane Remodeling Kinetics In Vitro with Lipid Multilayer Gratings. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:506-15. [PMID: 26649649 PMCID: PMC4843995 DOI: 10.1002/smll.201502398] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/28/2015] [Indexed: 05/08/2023]
Abstract
The dynamic self-organization of lipids in biological systems is a highly regulated process that enables the compartmentalization of living systems at micro- and nanoscopic scales. Consequently, quantitative methods for assaying the kinetics of supramolecular remodeling such as vesicle formation from planar lipid bilayers or multilayers are needed to understand cellular self-organization. Here, a new nanotechnology-based method for quantitative measurements of lipid-protein interactions is presented and its suitability for quantifying the membrane binding, inflation, and budding activity of the membrane-remodeling protein Sar1 is demonstrated. Lipid multilayer gratings are printed onto surfaces using nanointaglio and exposed to Sar1, resulting in the inflation of lipid multilayers into unilamellar structures, which can be observed in a label-free manner by monitoring the diffracted light. Local variations in lipid multilayer volume on the surface is used to vary substrate availability in a microarray format. A quantitative model is developed that allows quantification of binding affinity (K D ) and kinetics (kon and koff ). Importantly, this assay is uniquely capable of quantifying membrane remodeling. Upon Sar1-induced inflation of single bilayers from surface supported multilayers, the semicylindrical grating lines are observed to remodel into semispherical buds when a critical radius of curvature is reached.
Collapse
Affiliation(s)
- Troy W. Lowry
- Department of Physics, Florida State University, Tallahassee, FL 32306-4350, USA
| | - Hanaa Hariri
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306-4380, USA
| | - Plengchart Prommapan
- Department of Physics, Florida State University, Tallahassee, FL 32306-4350, USA
| | - Aubrey Kusi-Appiah
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4370, USA
| | - Nicholas Vafai
- Department of Biological Science, Florida State University, Tallahassee, FL 32306-4370, USA
| | - Ewa A. Bienkiewicz
- Department of Biomedical Science, College of Medicine, Florida State University, Tallahassee, FL 32306-4300, USA
| | - David H. Van Winkle
- Department of Physics, Florida State University, Tallahassee, FL 32306-4350, USA
| | - Scott M. Stagg
- Institute of Molecular Biophysics and Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306-4380, USA
| | - Steven Lenhert
- Department of Biological Science and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306-4370, USA
| |
Collapse
|
100
|
Camp ND, Lee KS, Cherry A, Wacker-Mhyre JL, Kountz TS, Park JM, Harris DA, Estrada M, Stewart A, Stella N, Wolf-Yadlin A, Hague C. Dynamic mass redistribution reveals diverging importance of PDZ-ligands for G protein-coupled receptor pharmacodynamics. Pharmacol Res 2016; 105:13-21. [PMID: 26773201 DOI: 10.1016/j.phrs.2016.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/28/2015] [Accepted: 01/01/2016] [Indexed: 02/08/2023]
Abstract
G protein-coupled receptors (GPCRs) are essential membrane proteins that facilitate cell-to-cell communication and co-ordinate physiological processes. At least 30 human GPCRs contain a Type I PSD-95/DLG/Zo-1 (PDZ) ligand in their distal C-terminal domain; this four amino acid motif of X-[S/T]-X-[φ] sequence facilitates interactions with PDZ domain-containing proteins. Because PDZ protein interactions have profound effects on GPCR ligand pharmacology, cellular localization, signal-transduction effector coupling and duration of activity, we analyzed the importance of Type I PDZ ligands for the function of 23 full-length and PDZ-ligand truncated (ΔPDZ) human GPCRs in cultured human cells. SNAP-epitope tag polyacrylamide gel electrophoresis revealed most Type I PDZ GPCRs exist as both monomers and multimers; removal of the PDZ ligand played minimal role in multimer formation. Additionally, SNAP-cell surface staining indicated removal of the PDZ ligand had minimal effects on plasma membrane localization for most GPCRs examined. Label-free dynamic mass redistribution functional responses, however, revealed diverging effects of the PDZ ligand. While no clear trend was observed across all GPCRs tested or even within receptor families, a subset of GPCRs displayed diminished agonist efficacy in the absence of a PDZ ligand (i.e. HT2RB, ADRB1), whereas others demonstrated enhanced agonist efficacies (i.e. LPAR2, SSTR5). These results demonstrate the utility of label-free functional assays to tease apart the contributions of conserved protein interaction domains for GPCR signal-transduction coupling in cultured cells.
Collapse
Affiliation(s)
- Nathan D Camp
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Kyung-Soon Lee
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Allison Cherry
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Jennifer L Wacker-Mhyre
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Timothy S Kountz
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Ji-Min Park
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Dorathy-Ann Harris
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Marianne Estrada
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Aaron Stewart
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Nephi Stella
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Alejandro Wolf-Yadlin
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Chris Hague
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|