1
|
Kolay J, Zhang P, Zhou X, Wan Z, Chieng A, Wang S. Ligand Binding-Induced Cellular Membrane Deformation is Correlated with the Changes in Membrane Stiffness. J Phys Chem B 2023; 127:9943-9953. [PMID: 37963180 PMCID: PMC10763494 DOI: 10.1021/acs.jpcb.3c06282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Study interaction between ligands and protein receptors is a key step for biomarker research and drug discovery. In situ measurement of cell surface membrane protein binding on whole cells eliminates the cost and pitfalls associated with membrane protein purification. Ligand binding to membrane protein was recently found to induce nanometer-scale cell membrane deformations, which can be monitored with real-time optical imaging to quantify ligand/protein binding kinetics. However, the insight into this phenomenon has still not been fully understood. We hypothesize that ligand binding can change membrane stiffness, which induces membrane deformation. To investigate this, cell height and membrane stiffness changes upon ligand binding are measured using atomic force microscopy (AFM). Wheat germ agglutinin (WGA) is used as a model ligand that binds to the cell surface glycoprotein. The changes in cell membrane stiffness and cell height upon ligand bindings are determined for three different cell lines (human A431, HeLa, and rat RBL-2H3) on two different substrates. AFM results show that cells become stiffer with increased height after WGA modification for all cases studied. The increase in cell membrane stiffness is further confirmed by plasmonic scattering microscopy, which shows an increased cell spring constant upon WGA binding. Therefore, this study provides direct experimental evidence that the membrane stiffness changes are directly correlated with ligand binding-induced cell membrane deformation.
Collapse
Affiliation(s)
- Jayeeta Kolay
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, Arizona 85287, USA
| | - Pengfei Zhang
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, Arizona 85287, USA
| | - Xinyu Zhou
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, Arizona 85287, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, USA
| | - Zijian Wan
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, Arizona 85287, USA
- School of Electrical, Energy and Computer Engineering, Arizona State University, Tempe, Arizona 85287, USA
| | - Andy Chieng
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, Arizona 85287, USA
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, USA
| | - Shaopeng Wang
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, Arizona 85287, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, USA
| |
Collapse
|
2
|
Putt KS, Du Y, Fu H, Zhang ZY. High-throughput screening strategies for space-based radiation countermeasure discovery. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:88-104. [PMID: 36336374 DOI: 10.1016/j.lssr.2022.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/13/2022] [Accepted: 07/19/2022] [Indexed: 06/16/2023]
Abstract
As humanity begins to venture further into space, approaches to better protect astronauts from the hazards found in space need to be developed. One particular hazard of concern is the complex radiation that is ever present in deep space. Currently, it is unlikely enough spacecraft shielding could be launched that would provide adequate protection to astronauts during long-duration missions such as a journey to Mars and back. In an effort to identify other means of protection, prophylactic radioprotective drugs have been proposed as a potential means to reduce the biological damage caused by this radiation. Unfortunately, few radioprotectors have been approved by the FDA for usage and for those that have been developed, they protect normal cells/tissues from acute, high levels of radiation exposure such as that from oncology radiation treatments. To date, essentially no radioprotectors have been developed that specifically counteract the effects of chronic low-dose rate space radiation. This review highlights how high-throughput screening (HTS) methodologies could be implemented to identify such a radioprotective agent. Several potential target, pathway, and phenotypic assays are discussed along with potential challenges towards screening for radioprotectors. Utilizing HTS strategies such as the ones proposed here have the potential to identify new chemical scaffolds that can be developed into efficacious radioprotectors that are specifically designed to protect astronauts during deep space journeys. The overarching goal of this review is to elicit broader interest in applying drug discovery techniques, specifically HTS towards the identification of radiation countermeasures designed to be efficacious towards the biological insults likely to be encountered by astronauts on long duration voyages.
Collapse
Affiliation(s)
- Karson S Putt
- Institute for Drug Discovery, Purdue University, West Lafayette IN 47907 USA
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Zhong-Yin Zhang
- Institute for Drug Discovery, Purdue University, West Lafayette IN 47907 USA; Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette IN 47907 USA.
| |
Collapse
|
3
|
Yao B, Yang Y, Yu N, Tao N, Wang D, Wang S, Zhang F. Label-Free Quantification of Molecular Interaction in Live Red Blood Cells by Tracking Nanometer Scale Membrane Fluctuations. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201623. [PMID: 35717672 PMCID: PMC9283308 DOI: 10.1002/smll.202201623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/17/2022] [Indexed: 06/15/2023]
Abstract
Molecular interactions in live cells play an important role in both cellular functions and drug discovery. Current methods for measuring binding kinetics involve extracting the membrane protein and labeling, while the in situ quantification of molecular interaction with surface plasmon resonance (SPR) imaging mainly worked with fixed cells due to the micro-motion related noises of live cells. Here, an optical imaging method is presented to measure the molecular interaction with live red blood cells by tracking the nanometer membrane fluctuations. The membrane fluctuation dynamics are measured by tracking the membrane displacement during glycoprotein interaction. The data are analyzed with a thermodynamic model to determine the elastic properties of the cell observing reduced membrane fluctuations under fixatives, indicating cell fixations affect membrane mechanical properties. The binding kinetics of glycoprotein to several lectins are obtained by tracking the membrane fluctuation amplitude changes on single live cells. The binding kinetics and strength of different lectins are quite different, indicating the glycoproteins expression heterogeneity in single cells. It is anticipated that the method will contribute to the understanding of mechanisms of cell interaction and communication, and have potential applications in the mechanical assessment of cancer or other diseases at the single-cell level, and screening of membrane protein targeting drugs.
Collapse
Affiliation(s)
- Bo Yao
- Department of Chemistry, Zhejiang University, Hangzhou, 310058, PR China
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Yunze Yang
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Nanxi Yu
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Nongjian Tao
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Di Wang
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- Intelligent Perception Research Institute, Zhejiang Laboratory, Hangzhou 311100, PR China
| | - Shaopeng Wang
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Fenni Zhang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, PR China
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
4
|
Danel A, Gondek E, Kucharek M, Szlachcic P, Gut A. 1 H-Pyrazolo[3,4- b]quinolines: Synthesis and Properties over 100 Years of Research. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092775. [PMID: 35566124 PMCID: PMC9099536 DOI: 10.3390/molecules27092775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/13/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022]
Abstract
This paper summarises a little over 100 years of research on the synthesis and the photophysical and biological properties of 1H-pyrazolo[3,4-b]quinolines that was published in the years 1911–2021. The main methods of synthesis are described, which include Friedländer condensation, synthesis from anthranilic acid derivatives, multicomponent synthesis and others. The use of this class of compounds as potential fluorescent sensors and biologically active compounds is shown. This review intends to summarize the abovementioned aspects of 1H-pyrazolo[3,4-b]quinoline chemistry. Some of the results that are presented in this publication come from the laboratories of the authors of this review.
Collapse
Affiliation(s)
- Andrzej Danel
- Faculty of Materials Engineering and Physics, Cracow University of Technology, Podchorążych Str. 1, 30-084 Krakow, Poland;
- Correspondence:
| | - Ewa Gondek
- Faculty of Materials Engineering and Physics, Cracow University of Technology, Podchorążych Str. 1, 30-084 Krakow, Poland;
| | - Mateusz Kucharek
- Faculty of Food Technology, University of Agriculture in Krakow, Balicka Str. 122, 30-149 Krakow, Poland; (M.K.); (P.S.)
| | - Paweł Szlachcic
- Faculty of Food Technology, University of Agriculture in Krakow, Balicka Str. 122, 30-149 Krakow, Poland; (M.K.); (P.S.)
| | - Arkadiusz Gut
- Faculty of Chemistry, Jagiellonian University, Gronostajowa Str. 2, 30-387 Krakow, Poland;
| |
Collapse
|
5
|
Nickelsen A, Jose J. Label-free flow cytometry-based enzyme inhibitor identification. Anal Chim Acta 2021; 1179:338826. [PMID: 34535248 DOI: 10.1016/j.aca.2021.338826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/22/2021] [Accepted: 07/02/2021] [Indexed: 11/24/2022]
Abstract
Fluorescence-based methods for the identification of enzyme inhibitors are widespread, but usually require protein or ligand labelling. In this study, we present a label-free displacement assay that takes advantage of the intrinsic fluorescence of a tight binding ligand avoiding any labeling. Autodisplay-based accessibility of the target enzyme on the cell surface of Escherichia coli enabled the quantification of fluorescent ligand binding by flow cytometry. Human protein kinase CK2 was used as proof-of-concept enzyme and its ATP competitive inhibitor (E)-1,3-dichloro-6-[(4-methoxyphenylimino)methyl]dibenzo[b,d]furan-2,7-diol (compound 5) was shown to exhibit intrinsic fluorescence (λmax(ex) = 370 nm, λmax(em) = 585 nm). Binding of compound 5 to CK2 displaying cells was quantified via flow cytometry with linearly increasing relative fluorescence up to a concentration of 1.25 μM. The addition of the non-fluorescent CK2 inhibitor 4,5,6,7-tetrabromobenzotriazole (TBB) competed for compound 5 binding with a half maximal fluorescence reduction at 15.6 μM TBB. This new and simple binding assay provides a valuable tool for the screening of high affinity enzyme inhibitors, overcoming the limitations of fluorescent ligand labelling.
Collapse
Affiliation(s)
- Anna Nickelsen
- Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Joachim Jose
- Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany.
| |
Collapse
|
6
|
Scholle MD, McLaughlin D, Gurard-Levin ZA. High-Throughput Affinity Selection Mass Spectrometry Using SAMDI-MS to Identify Small-Molecule Binders of the Human Rhinovirus 3C Protease. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2021; 26:974-983. [PMID: 34151629 DOI: 10.1177/24725552211023211] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Affinity selection mass spectrometry (ASMS) has emerged as a powerful high-throughput screening tool used in drug discovery to identify novel ligands against therapeutic targets. This report describes the first high-throughput screen using a novel self-assembled monolayer desorption ionization (SAMDI)-ASMS methodology to reveal ligands for the human rhinovirus 3C (HRV3C) protease. The approach combines self-assembled monolayers of alkanethiolates on gold with matrix-assisted laser desorption ionization time-of-flight (MALDI TOF) mass spectrometry (MS), a technique termed SAMDI-ASMS. The primary screen of more than 100,000 compounds in pools of 8 compounds per well was completed in less than 8 h, and informs on the binding potential and selectivity of each compound. Initial hits were confirmed in follow-up SAMDI-ASMS experiments in single-concentration and dose-response curves. The ligands identified by SAMDI-ASMS were further validated using differential scanning fluorimetry (DSF) and in functional protease assays against HRV3C and the related SARS-CoV-2 3CLpro enzyme. SAMDI-ASMS offers key benefits for drug discovery over traditional ASMS approaches, including the high-throughput workflow and readout, minimizing compound misbehavior by using smaller compound pools, and up to a 50-fold reduction in reagent consumption. The flexibility of this novel technology opens avenues for high-throughput ASMS assays of any target, thereby accelerating drug discovery for diverse diseases.
Collapse
|
7
|
Enomoto T, Nakako T, Goda M, Wada E, Kitamura A, Fujii Y, Ikeda K. A novel phosphodiesterase 1 inhibitor reverses L-dopa-induced dyskinesia, but not motivation deficits, in monkeys. Pharmacol Biochem Behav 2021; 205:173183. [PMID: 33774006 DOI: 10.1016/j.pbb.2021.173183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 11/28/2022]
Abstract
The enzyme phosphodiesterase 1 (PDE1) is highly expressed in the striatum and cortex. However, its role in corticostriatal function has not been fully investigated. The present study was aimed at evaluating the therapeutic potential of PDE1 inhibitors in treating motivation deficits and 3,4-dihydroxy-L-phenylalanine (L-dopa)-induced dyskinesia, which are pathological conditions of the corticostriatal system. We used a novel PDE1 inhibitor 3-ethyl-2-{[trans-4-(methoxymethyl)cyclohexyl]oxy}-7-(tetrahydro-2H-pyran-4-yl)-imidazo[5,1-f][1,2,4]triazin-4(3H)-one (DSR-143136), which was identified in our drug discovery program. Motivation in monkeys was measured using a progressive ratio task. L-Dopa-induced dyskinesia and disability scores were measured in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated monkeys. DSR-143136 had a high selectivity for PDE1 over other PDE families and 67 other biologic targets. A dopamine D1 receptor antagonist SCH-39166 at 0.01, 0.03 and 0.1 mg/kg potently decreased motivation in monkeys. However, DSR-143136 at 0.3 and 3 mg/kg did not affect motivation deficits induced by low-dose SCH-39166 (0.01 mg/kg). On the other hand, DSR-143136 at 3 mg/kg potently decreased L-dopa-induced dyskinesia in the Parkinsonian monkey model. Importantly, this antidyskinesic efficacy was NOT accompanied by detrimental effects on motor function. Further, this compound decreased on-time with marked or severe dyskinesia, without affecting on-time itself. These findings suggest that PDE1 inhibitor could be a therapeutic candidate for treating L-dopa-induced dyskinesia in Parkinson's disease, but not for motivation deficits.
Collapse
Affiliation(s)
- Takeshi Enomoto
- Drug Research Division, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan.
| | - Tomokazu Nakako
- Drug Research Division, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Masao Goda
- Drug Research Division, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Erika Wada
- Drug Research Division, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Atsushi Kitamura
- Drug Research Division, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Yuki Fujii
- Drug Research Division, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Kazuhito Ikeda
- Drug Research Division, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| |
Collapse
|
8
|
Buñay J, Fouache A, Trousson A, de Joussineau C, Bouchareb E, Zhu Z, Kocer A, Morel L, Baron S, Lobaccaro JMA. Screening for liver X receptor modulators: Where are we and for what use? Br J Pharmacol 2020; 178:3277-3293. [PMID: 33080050 DOI: 10.1111/bph.15286] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/14/2020] [Accepted: 10/05/2020] [Indexed: 12/11/2022] Open
Abstract
Liver X receptors (LXRs) are members of the nuclear receptor superfamily that are canonically activated by oxidized derivatives of cholesterol. Since the mid-90s, numerous groups have identified LXRs as endocrine receptors that are involved in the regulation of various physiological functions. As a result, when their expression is genetically modified in mice, phenotypic analyses reveal endocrine disorders ranging from infertility to diabetes and obesity, nervous system pathologies such Alzheimer's or Parkinson's disease, immunological disturbances, inflammatory response, and enhancement of tumour development. Based on such findings, it appears that LXRs could constitute good pharmacological targets to prevent and/or to treat these diseases. This review discusses the various aspects of LXR drug discovery, from the tools available for the screening of potential LXR modulators to the current situational analysis of the drugs in development. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Julio Buñay
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Allan Fouache
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Amalia Trousson
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Cyrille de Joussineau
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Erwan Bouchareb
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Zhekun Zhu
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Ayhan Kocer
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Laurent Morel
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Silvere Baron
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| | - Jean-Marc A Lobaccaro
- Université Clermont Auvergne, GReD, CNRS, INSERM, and Centre de Recherche en Nutrition Humaine d'Auvergne Clermont-Ferrand, Clermont-Ferrand, France
| |
Collapse
|
9
|
Simon RP, Winter M, Kleiner C, Wehrle L, Karnath M, Ries R, Zeeb M, Schnapp G, Fiegen D, Häbe TT, Runge F, Bretschneider T, Luippold AH, Bischoff D, Reindl W, Büttner FH. MALDI-TOF-Based Affinity Selection Mass Spectrometry for Automated Screening of Protein-Ligand Interactions at High Throughput. SLAS DISCOVERY 2020; 26:44-57. [PMID: 33073664 DOI: 10.1177/2472555220959266] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Demonstration of in vitro target engagement for small-molecule ligands by measuring binding to a molecular target is an established approach in early drug discovery and a pivotal step in high-throughput screening (HTS)-based compound triaging. We describe the setup, evaluation, and application of a ligand binding assay platform combining automated affinity selection (AS)-based sample preparation and label-free matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) analysis. The platform enables mass spectrometry (MS)-based HTS for small-molecule target interactions from single-compound incubation mixtures and is embedded into a regular assay automation environment. Efficient separation of target-ligand complexes is achieved by in-plate size exclusion chromatography (SEC), and small-molecule ligands are subsequently identified by MALDI-TOF analysis. In contrast to alternative HTS-capable binding assay formats, MALDI-TOF AS-MS is capable of identifying orthosteric and allosteric ligands, as shown for the model system protein tyrosine phosphatase 1B (PTP1B), irrespective of protein function. Furthermore, determining relative binding affinities (RBAs) enabled ligand ranking in accordance with functional inhibition and reference data for PTP1B and a number of diverse protein targets. Finally, we present a validation screen of more than 23,000 compounds within 24 h, demonstrating the general applicability of the platform for the HTS-compatible assessment of protein-ligand interactions.
Collapse
Affiliation(s)
- Roman P Simon
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Martin Winter
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Carola Kleiner
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Lucie Wehrle
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Michael Karnath
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Robert Ries
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Markus Zeeb
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Gisela Schnapp
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dennis Fiegen
- Bioprocess Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Tim T Häbe
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Frank Runge
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Tom Bretschneider
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Andreas H Luippold
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Daniel Bischoff
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Wolfgang Reindl
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Frank H Büttner
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
10
|
Belov AM, Kozole J, Bean MF, Machutta CA, Zhang G, Gao EN, Ghislain L, Datwani SS, Leveridge M, Annan RS. Acoustic Mist Ionization-Mass Spectrometry: A Comparison to Conventional High-Throughput Screening and Compound Profiling Platforms. Anal Chem 2020; 92:13847-13854. [DOI: 10.1021/acs.analchem.0c02508] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
| | | | | | | | | | | | - Luke Ghislain
- Beckman Coulter Life Sciences, 170 Rose Orchard Way, San Jose, California 95134, United States
| | - Sammy S. Datwani
- Beckman Coulter Life Sciences, 170 Rose Orchard Way, San Jose, California 95134, United States
| | | | | |
Collapse
|
11
|
Blay V, Tolani B, Ho SP, Arkin MR. High-Throughput Screening: today's biochemical and cell-based approaches. Drug Discov Today 2020; 25:1807-1821. [PMID: 32801051 DOI: 10.1016/j.drudis.2020.07.024] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/01/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022]
Abstract
High-throughput screening (HTS) provides starting chemical matter in the adventure of developing a new drug. In this review, we survey several HTS methods used today for hit identification, organized in two main flavors: biochemical and cell-based assays. Biochemical assays discussed include fluorescence polarization and anisotropy, FRET, TR-FRET, and fluorescence lifetime analysis. Binding-based methods are also surveyed, including NMR, SPR, mass spectrometry, and DSF. On the other hand, cell-based assays discussed include viability, reporter gene, second messenger, and high-throughput microscopy assays. We devote some emphasis to high-content screening, which is becoming very popular. An advisable stage after hit discovery using phenotypic screens is target deconvolution, and we provide an overview of current chemical proteomics, in silico, and chemical genetics tools. Emphasis is made on recent CRISPR/dCas-based screens. Lastly, we illustrate some of the considerations that inform the choice of HTS methods and point to some areas with potential interest for future research.
Collapse
Affiliation(s)
- Vincent Blay
- Division of Biomaterials and Bioengineering, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Bhairavi Tolani
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Sunita P Ho
- Division of Biomaterials and Bioengineering, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Michelle R Arkin
- Department of Pharmaceutical Chemistry and the Small Molecule Discovery Center, University of California, San Francisco, CA, USA.
| |
Collapse
|
12
|
Targeting Glycosylation: A New Road for Cancer Drug Discovery. Trends Cancer 2020; 6:757-766. [PMID: 32381431 DOI: 10.1016/j.trecan.2020.04.002] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/04/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022]
Abstract
Cancer is a deadly disease that encompasses numerous cellular modifications. Among them, alterations in glycosylation are a proven reliable hallmark of cancer, with most biomarkers used in the clinic detecting cancer-associated glycans. Despite their clear potential as therapy targets, glycans have been overlooked in drug discovery strategies. The complexity associated with the glycosylation process, and lack of specific methodologies to study it, have long hampered progress. However, recent advances in new methodologies, such as glycoengineering of cells and high-throughput screening (HTS), have opened new avenues of discovery. We envision that glycan-based targeting has the potential to start a new era of cancer therapy. In this article, we discuss the promise of cancer-associated glycosylation for the discovery of effective cancer drugs.
Collapse
|
13
|
Miniaturized technologies for high-throughput drug screening enzymatic assays and diagnostics – A review. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.115862] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
14
|
Enomoto T, Tatara A, Goda M, Nishizato Y, Nishigori K, Kitamura A, Kamada M, Taga S, Hashimoto T, Ikeda K, Fujii Y. A Novel Phosphodiesterase 1 Inhibitor DSR-141562 Exhibits Efficacies in Animal Models for Positive, Negative, and Cognitive Symptoms Associated with Schizophrenia. J Pharmacol Exp Ther 2019; 371:692-702. [PMID: 31578257 DOI: 10.1124/jpet.119.260869] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/18/2019] [Indexed: 01/09/2023] Open
Abstract
In our drug discovery program, we identified a novel orally available and brain-penetrant phosphodiesterase (PDE) 1 inhibitor, 3-methyl-7-(tetrahydro-2H-pyran-4-yl)-2-{[trans-4-(trifluoromethyl)cyclohexyl]-methoxy}imidazo[5,1-f][1,2,4]triazin-4(3H)-one (DSR-141562). In the present study, we characterized the preclinical profile of DSR-141562. This compound has preferential selectivity for predominantly brain-expressed PDE1B over other PDE1 family members, and high selectivity for the PDE1 family over other PDE families and 65 other tested biologic targets. Oral administration of DSR-141562 at 10 mg/kg slightly elevated the cGMP concentration, and it potently enhanced the increase of cGMP induced by a dopamine D1 receptor agonist in mouse brains. The cGMP level in monkey cerebrospinal fluid was also elevated after treatment with DSR-141562 at 30 and 100 mg/kg and could be used as a translational biomarker. Since PDE1B is believed to regulate dopaminergic and glutamatergic signal transduction, we evaluated the effects of this compound using schizophrenia-related behavioral assays. DSR-141562 at 3-30 mg/kg potently inhibited methamphetamine-induced locomotor hyperactivity in rats, while it had only minimal effects on the spontaneous locomotor activity. Furthermore, DSR-141562 at 1-100 mg/kg did not induce any signs of catalepsy in rats. DSR-141562 at 0.3-3 mg/kg reversed social interaction and novel object recognition deficits induced by repeated treatment with an N-methyl-D-aspartate receptor antagonist, phencyclidine, in mice and rats, respectively. In common marmosets, DSR-141562 at 3 and 30 mg/kg improved the performance in object retrieval with detour tasks. These results suggest that DSR-141562 is a therapeutic candidate for positive, negative, and cognitive symptoms in schizophrenia. SIGNIFICANCE STATEMENT: This is the first paper showing that a phosphodiesterase 1 inhibitor is efficacious in animal models for positive and negative symptoms associated with schizophrenia. Furthermore, we demonstrated that this compound improved cognitive function in the common marmoset, a nonhuman primate.
Collapse
Affiliation(s)
- Takeshi Enomoto
- Drug Research Division, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Ayaka Tatara
- Drug Research Division, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Masao Goda
- Drug Research Division, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Yohei Nishizato
- Drug Research Division, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Kantaro Nishigori
- Drug Research Division, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Atsushi Kitamura
- Drug Research Division, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Mami Kamada
- Drug Research Division, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Shiori Taga
- Drug Research Division, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Takashi Hashimoto
- Drug Research Division, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Kazuhito Ikeda
- Drug Research Division, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| | - Yuki Fujii
- Drug Research Division, Sumitomo Dainippon Pharma, Co., Ltd., Osaka, Japan
| |
Collapse
|
15
|
Ayukawa K, Suzuki C, Ogasawara H, Kinoshita T, Furuno M, Suzuki G. Development of a High-Throughput Screening-Compatible Assay for Discovery of GPR3 Inverse Agonists Using a cAMP Biosensor. SLAS DISCOVERY 2019; 25:287-298. [PMID: 31516076 DOI: 10.1177/2472555219875101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
While G-protein-coupled receptors (GPCRs) represent the largest class of cell surface proteins, there are ≥100 orphan GPCRs whose endogenous ligands are unknown. Accordingly, these could prove to be potential therapeutic targets for the pharmaceutical intervention of various diseases. Constitutively active orphan GPCRs are activated without ligands; thus, inverse agonists may be very useful pharmacological tools for inhibiting constitutive activity. However, in general, inverse agonist screening is considered more difficult to perform with high quality than antagonist screening, particularly due to the narrow assay window. We developed a high-throughput screening (HTS)-compatible assay to identify inverse agonists of GPR3. GPR3 is expressed in the central nervous system (CNS) and is known to be related to Alzheimer's disease and other CNS diseases. The GPR3 inducible cell line was established using T-REx 293 cells that stably expressed the tetracycline repressor protein, and the cAMP biosensor, GloSensor, was stably co-expressed. After optimization of the induction level of GPR3 and assay conditions, the GloSensor assay showed an approximately 20-fold signal-to-background ratio and high sensitivity. Using the HTS method, we successfully screened a library of hundreds of thousands of compounds for the inhibition of constitutive activity with good quality and excellent reproducibility. Finally, 35 compounds were identified as GPR3 selective inverse agonists. This inverse agonist screening approach using GloSensor in combination with the inducible expression of orphan GPCR indicates universal applicability to the search for inverse agonists of constitutively active orphan GPCRs.
Collapse
Affiliation(s)
- Kumiko Ayukawa
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| | - Chie Suzuki
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| | - Hiroyuki Ogasawara
- Pharmaceutical Frontier Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Tomomi Kinoshita
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| | - Masahiro Furuno
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| | - Gentaroh Suzuki
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| |
Collapse
|
16
|
Zhang J, Hu Y, Musharrafieh R, Yin H, Wang J. Focusing on the Influenza Virus Polymerase Complex: Recent Progress in Drug Discovery and Assay Development. Curr Med Chem 2019; 26:2243-2263. [PMID: 29984646 DOI: 10.2174/0929867325666180706112940] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 03/27/2018] [Accepted: 05/06/2018] [Indexed: 12/17/2022]
Abstract
Influenza viruses are severe human pathogens that pose persistent threat to public health. Each year more people die of influenza virus infection than that of breast cancer. Due to the limited efficacy associated with current influenza vaccines, as well as emerging drug resistance from small molecule antiviral drugs, there is a clear need to develop new antivirals with novel mechanisms of action. The influenza virus polymerase complex has become a promising target for the development of the next-generation of antivirals for several reasons. Firstly, the influenza virus polymerase, which forms a heterotrimeric complex that consists of PA, PB1, and PB2 subunits, is highly conserved. Secondly, both individual polymerase subunit (PA, PB1, and PB2) and inter-subunit interactions (PA-PB1, PB1- PB2) represent promising drug targets. Lastly, growing insight into the structure and function of the polymerase complex has spearheaded the structure-guided design of new polymerase inhibitors. In this review, we highlight recent progress in drug discovery and assay development targeting the influenza virus polymerase complex and discuss their therapeutic potentials.
Collapse
Affiliation(s)
- Jiantao Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Rami Musharrafieh
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Hang Yin
- Department of Chemistry and Biochemistry, BioFrontiers Institute, University of Colorado, Boulder, Colorado 80309, United States
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States.,BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
17
|
Janczak CM, Calderon IAC, Noviana E, Hadvani P, Lee JR, Aspinwall CA. Hybrid Nanoparticle Platform for Nanoscale Scintillation Proximity Assay. ACS APPLIED NANO MATERIALS 2019; 2:1259-1266. [PMID: 34316544 PMCID: PMC8313019 DOI: 10.1021/acsanm.8b02136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
β-particle emitting radionuclides, such as 3H, 14C, 32P, 33P, and 35S, are important molecular labels due to their small size and the prevalence of these atoms in biomolecules but are challenging to selectively detect and quantify within aqueous biological samples and systems. Here, we present a core-shell nanoparticle-based scintillation proximity assay platform (nanoSPA) for the separation-free, selective detection of radiolabeled analytes. nanoSPA is prepared by incorporating scintillant fluorophores into polystyrene core particles and encapsulating the scintillant-doped cores within functionalized silica shells. The functionalized surface enables covalent attachment of specific binding moieties such as small molecules, proteins, or DNA that can be used for analyte-specific detection. nanoSPA was demonstrated for detection of 3H-labeled analytes, the most difficult biologically relevant β-emitter to measure due to the low energy β-particle emission, using three model assays that represent covalent and non-covalent binding systems that necessitate selectivity over competing 3H-labeled species. In each model, nmol quantities of target were detected directly in aqueous solution without separation from unbound 3H-labeled analyte. The nanoSPA platform facilitated measurement of 3H-labeled analytes directly in bulk aqueous samples without surfactants or other agents used to aid particle dispersal. Selectivity for bound 3H-analytes over unbound 3H analytes was enhanced up to 30-fold when the labeled species was covalently bound to nanoSPA, and 4- and 8-fold for two non-covalent binding assays using nanoSPA. The small size and enhanced selectivity of nanoSPA should enable new applications compared to the commonly used microSPA platform, including the potential for separation-free, analyte-specific cellular or intracellular detection.
Collapse
Affiliation(s)
- Colleen M. Janczak
- Department of Chemistry and Biochemistry, University of Arizona, Tucson 85721-00041, United States
| | - Isen A. C. Calderon
- Department of Chemistry and Biochemistry, University of Arizona, Tucson 85721-00041, United States
| | - Eka Noviana
- Department of Chemistry and Biochemistry, University of Arizona, Tucson 85721-00041, United States
| | - Priyanka Hadvani
- Department of Chemistry and Biochemistry, University of Arizona, Tucson 85721-00041, United States
| | - Joo Ryung Lee
- Department of Chemistry and Biochemistry, University of Arizona, Tucson 85721-00041, United States
| | - Craig A. Aspinwall
- Department of Chemistry and Biochemistry, University of Arizona, Tucson 85721-00041, United States
- BIO5 Institute, University of Arizona, Tucson 85721-00041, United States
- Department of Biomedical Engineering, University of Arizona, Tucson 85721-00041, United States
| |
Collapse
|
18
|
Muraki Y, Naito T, Tohyama K, Shibata S, Kuniyeda K, Nio Y, Hazama M, Matsuo T. Improvement of pulmonary arterial hypertension, inflammatory response, and epithelium injury by dual activation of cAMP/cGMP pathway in a rat model of monocrotaline-induced pulmonary hypertension. Biosci Biotechnol Biochem 2019; 83:1000-1010. [PMID: 30835622 DOI: 10.1080/09168451.2019.1584520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pulmonary hypertension (PH) is a life-threatening lung disease. PH with concomitant lung diseases, e.g., idiopathic pulmonary fibrosis, is associated with poor prognosis. Development of novel therapeutic vasodilators for treatment of these patients is a key imperative. We evaluated the efficacy of dual activation of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) using an active, small-molecule phosphodiesterase (PDE4)/PDE5 dual inhibitor (Compound A). Compound A increased both cAMP and cGMP levels in WI-38 lung fibroblasts and suppressed the expressions of type-1 collagen α1 chain and fibronectin. Additionally, compound A reduced right ventricular weight/left ventricular weight+septal weight ratio, brain natriuretic peptide expression levels in right ventricle, C─C motif chemokine ligand 2 expression levels in lung, and plasma surfactant protein D. Our data indicate that dual activation of cAMP/cGMP pathways may be a novel treatment strategy for PH.
Collapse
Affiliation(s)
- Yo Muraki
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Takako Naito
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Kimio Tohyama
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Sachio Shibata
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Kanako Kuniyeda
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Yasunori Nio
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Masatoshi Hazama
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Takanori Matsuo
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| |
Collapse
|
19
|
Otvos RA, Still KBM, Somsen GW, Smit AB, Kool J. Drug Discovery on Natural Products: From Ion Channels to nAChRs, from Nature to Libraries, from Analytics to Assays. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2019; 24:362-385. [PMID: 30682257 PMCID: PMC6484542 DOI: 10.1177/2472555218822098] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/16/2018] [Accepted: 12/07/2018] [Indexed: 12/21/2022]
Abstract
Natural extracts are complex mixtures that may be rich in useful bioactive compounds and therefore are attractive sources for new leads in drug discovery. This review describes drug discovery from natural products and in explaining this process puts the focus on ion-channel drug discovery. In particular, the identification of bioactives from natural products targeting nicotinic acetylcholine receptors (nAChRs) and serotonin type 3 receptors (5-HT3Rs) is discussed. The review is divided into three parts: "Targets," "Sources," and "Approaches." The "Targets" part will discuss the importance of ion-channel drug targets in general, and the α7-nAChR and 5-HT3Rs in particular. The "Sources" part will discuss the relevance for drug discovery of finding bioactive compounds from various natural sources such as venoms and plant extracts. The "Approaches" part will give an overview of classical and new analytical approaches that are used for the identification of new bioactive compounds with the focus on targeting ion channels. In addition, a selected overview is given of traditional venom-based drug discovery approaches and of diverse hyphenated analytical systems used for screening complex bioactive mixtures including venoms.
Collapse
Affiliation(s)
- Reka A. Otvos
- The Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of BioAnalytical Chemistry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Kristina B. M. Still
- The Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of BioAnalytical Chemistry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Govert W. Somsen
- The Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of BioAnalytical Chemistry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jeroen Kool
- The Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of BioAnalytical Chemistry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Chai SC, Lin W, Li Y, Chen T. Drug discovery technologies to identify and characterize modulators of the pregnane X receptor and the constitutive androstane receptor. Drug Discov Today 2019; 24:906-915. [PMID: 30731240 PMCID: PMC6421094 DOI: 10.1016/j.drudis.2019.01.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/27/2018] [Accepted: 01/30/2019] [Indexed: 11/24/2022]
Abstract
The pregnane X receptor (PXR) and the constitutive androstane receptor (CAR) are ligand-activated nuclear receptors (NRs) that are notorious for their role in drug metabolism, causing unintended drug-drug interactions and decreasing drug efficacy. They control the xenobiotic detoxification system by regulating the expression of an array of drug-metabolizing enzymes and transporters that excrete exogenous chemicals and maintain homeostasis of endogenous metabolites. Much effort has been invested in recognizing potential drugs for clinical use that can activate PXR and CAR to enhance the expression of their target genes, and in identifying PXR and CAR inhibitors that can be used as co-therapeutics to prevent adverse effects. Here, we present current technologies and assays used in the quest to characterize PXR and CAR modulators, which range from biochemical to cell-based and animal models.
Collapse
Affiliation(s)
- Sergio C Chai
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Wenwei Lin
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Yongtao Li
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| |
Collapse
|
21
|
Vasylieva N, Kitamura S, Dong J, Barnych B, Hvorecny KL, Madden DR, Gee SJ, Wolan DW, Morisseau C, Hammock BD. Nanobody-based binding assay for the discovery of potent inhibitors of CFTR inhibitory factor (Cif). Anal Chim Acta 2019; 1057:106-113. [PMID: 30832908 DOI: 10.1016/j.aca.2018.12.060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/21/2018] [Accepted: 12/26/2018] [Indexed: 12/23/2022]
Abstract
Lead identification and optimization are essential steps in the development of a new drug. It requires cost-effective, selective and sensitive chemical tools. Here, we report a novel method using nanobodies that allows the efficient screening for potent ligands. The method is illustrated with the cystic fibrosis transmembrane conductance regulator inhibitory factor (Cif), a virulence factor secreted by the opportunistic pathogen Pseudomonas aeruginosa. 18 nanobodies selective to Cif were isolated by bio-panning from nanobody-phage library constructed from immunized llama. 8 out of 18 nanobodies were identified as potent inhibitors of Cif enzymatic activity with IC50s in the range of 0.3-6.4 μM. A nanobody VHH219 showed high affinity (KD = 0.08 nM) to Cif and the highest inhibitory potency, IC50 = 0.3 μM. A displacement sandwich ELISA (dsELISA) with VHH219 was then developed for classification of synthetic small molecule inhibitors according their inhibitory potency. The developed assay allowed identification of new inhibitor with highest potency reported so far (0.16 ± 0.02 μM). The results from dsELISA assay correlates strongly with a conventional fluorogenic assay (R = 0.9998) in predicting the inhibitory potency of the tested compounds. However, the novel dsELISA is an order of magnitude more sensitive and allows the identification and ranking of potent inhibitors missed by the classic fluorogenic assay method. These data were supported with Octet biolayer interferometry measurements. The novel method described herein relies solely on the binding properties of the specific neutralizing nanobody, and thus is applicable to any pharmacological target for which such a nanobody can be found, independent of any requirement for catalytic activity.
Collapse
Affiliation(s)
- Natalia Vasylieva
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, CA, 95616, United States
| | - Seiya Kitamura
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, CA, 95616, United States; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jiexian Dong
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, CA, 95616, United States
| | - Bogdan Barnych
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, CA, 95616, United States
| | - Kelli L Hvorecny
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Dean R Madden
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Shirley J Gee
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, CA, 95616, United States
| | - Dennis W Wolan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Christophe Morisseau
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, CA, 95616, United States.
| | - Bruce D Hammock
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, CA, 95616, United States
| |
Collapse
|
22
|
Colley CS, England E, Linley JE, Wilkinson TCI. Screening Strategies for the Discovery of Ion Channel Monoclonal Antibodies. ACTA ACUST UNITED AC 2018; 82:e44. [DOI: 10.1002/cpph.44] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Caroline S. Colley
- Antibody Discovery and Protein Engineering, MedImmune; Cambridge United Kingdom
| | - Elizabeth England
- Antibody Discovery and Protein Engineering, MedImmune; Cambridge United Kingdom
| | - John E. Linley
- Neuroscience, IMED Biotech Unit, AstraZeneca; Cambridge United Kingdom
| | | |
Collapse
|
23
|
Zhang F, Jing W, Hunt A, Yu H, Yang Y, Wang S, Chen HY, Tao N. Label-Free Quantification of Small-Molecule Binding to Membrane Proteins on Single Cells by Tracking Nanometer-Scale Cellular Membrane Deformation. ACS NANO 2018; 12:2056-2064. [PMID: 29397682 PMCID: PMC5851003 DOI: 10.1021/acsnano.8b00235] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Measuring molecular binding to membrane proteins is critical for understanding cellular functions, validating biomarkers, and screening drugs. Despite the importance, developing such a capability has been a difficult challenge, especially for small-molecule binding to membrane proteins in their native cellular environment. Here we show that the binding of both large and small molecules to membrane proteins can be quantified on single cells by trapping single cells with a microfluidic device and detecting binding-induced cellular membrane deformation on the nanometer scale with label-free optical imaging. We develop a thermodynamic model to describe the binding-induced membrane deformation, validate the model by examining the dependence of membrane deformation on cell stiffness, membrane protein expression level, and binding affinity, and study four major types of membrane proteins, including glycoproteins, ion channels, G-protein coupled receptors, and tyrosine kinase receptors. The single-cell detection capability reveals the importance of local membrane environment on molecular binding and variability in the binding kinetics of different cell lines and heterogeneity of different cells within the same cell line.
Collapse
Affiliation(s)
- Fenni Zhang
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- School of Electrical Computer and Energy Engineering, Arizona State University, Tempe, Arizona 85287, USA
| | - Wenwen Jing
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Ashley Hunt
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Hui Yu
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Yunze Yang
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- School of Electrical Computer and Energy Engineering, Arizona State University, Tempe, Arizona 85287, USA
| | - Shaopeng Wang
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Hong-Yuan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
| | - Nongjian Tao
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
- School of Electrical Computer and Energy Engineering, Arizona State University, Tempe, Arizona 85287, USA
| |
Collapse
|
24
|
Atzrodt J, Derdau V, Kerr WJ, Reid M. Deuterium- und tritiummarkierte Verbindungen: Anwendungen in den modernen Biowissenschaften. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201704146] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jens Atzrodt
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry; Industriepark Höchst, G876 65926 Frankfurt Deutschland
| | - Volker Derdau
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry; Industriepark Höchst, G876 65926 Frankfurt Deutschland
| | - William J. Kerr
- Department of Pure and Applied Chemistry, WestCHEM; University of Strathclyde; 295 Cathedral Street Glasgow Scotland G1 1XL Großbritannien
| | - Marc Reid
- Department of Pure and Applied Chemistry, WestCHEM; University of Strathclyde; 295 Cathedral Street Glasgow Scotland G1 1XL Großbritannien
| |
Collapse
|
25
|
Atzrodt J, Derdau V, Kerr WJ, Reid M. Deuterium- and Tritium-Labelled Compounds: Applications in the Life Sciences. Angew Chem Int Ed Engl 2018; 57:1758-1784. [PMID: 28815899 DOI: 10.1002/anie.201704146] [Citation(s) in RCA: 421] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/27/2017] [Indexed: 12/19/2022]
Abstract
Hydrogen isotopes are unique tools for identifying and understanding biological and chemical processes. Hydrogen isotope labelling allows for the traceless and direct incorporation of an additional mass or radioactive tag into an organic molecule with almost no changes in its chemical structure, physical properties, or biological activity. Using deuterium-labelled isotopologues to study the unique mass-spectrometric patterns generated from mixtures of biologically relevant molecules drastically simplifies analysis. Such methods are now providing unprecedented levels of insight in a wide and continuously growing range of applications in the life sciences and beyond. Tritium (3 H), in particular, has seen an increase in utilization, especially in pharmaceutical drug discovery. The efforts and costs associated with the synthesis of labelled compounds are more than compensated for by the enhanced molecular sensitivity during analysis and the high reliability of the data obtained. In this Review, advances in the application of hydrogen isotopes in the life sciences are described.
Collapse
Affiliation(s)
- Jens Atzrodt
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry, Industriepark Höchst, G876, 65926, Frankfurt, Germany
| | - Volker Derdau
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry, Industriepark Höchst, G876, 65926, Frankfurt, Germany
| | - William J Kerr
- Department of Pure and Applied Chemistry, WestCHEM, University of Strathclyde, 295 Cathedral Street, Glasgow, Scotland, G1 1XL, UK
| | - Marc Reid
- Department of Pure and Applied Chemistry, WestCHEM, University of Strathclyde, 295 Cathedral Street, Glasgow, Scotland, G1 1XL, UK
| |
Collapse
|
26
|
Chen H, Wang F, Moore T, Qi B, Sulejmanovic D, Hwu SJ, Mefford OT, Alexis F, Anker JN. Bright X-ray and up-conversion nanophosphors annealed using encapsulated sintering agents for bioimaging applications. J Mater Chem B 2017; 5:5412-5424. [PMID: 29497532 PMCID: PMC5826634 DOI: 10.1039/c7tb01289f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nanophosphors are promising contrast agents for deep tissue optical imaging applications because they can be excited by X-ray and near infrared light that penetrates deeply through tissue and generates almost no autofluorescence background in the tissue. For these bioimaging applications, the nanophosophors should ideally be small, monodispersed and brightly luminescent. However, most methods used to improve luminescence yield by annealing the particles to reduce crystal and surface defects (e.g. using flux or sintering agents) also cause particle fusion or require multiple component core-shell structures. Here, we report a novel method to prepare bright, uniformly sized X-ray nanophosphors (Gd2O2S:Eu or Tb) and upconversion nanophosphors (Y2O2S: Yb/Er, or Yb/Tm) with large crystal domain size without causing aggregation. A core-shell nanoparticle is formed, with NaF only in the core. We observe that increasing the NaF sintering agent concentration up to 7.6 mol% increases both crystal domain size and luminescence intensity (up to 40% of commercial microphosphors) without affecting the physical particticle diameter. Above 7.6 mol%, particle fusion is observed. The annealing is insensitive to the cation (Na+ or K+) but varies strongly with anion, with F->Cl->CO32->Br->I-. The luminescence depends strongly on crystal domain size. The data agree reasonably well with a simple domain surface quenching model, although the size-dependence suggests additional quenching mechanisms within small domains. The prepared bright nanophosphors were subsequently functionalized with PEG-folic acid to target MCF-7 breast cancer cells which overexpress folic acid receptors. Both X-ray and upconversion nanophosphors provided low background and bright luminescence which was imaged through 1 cm chicken breast tissue at a low dose of nanophosphors 200 µL (0.1 mg/mL). We anticipate these highly monodispersed and bright X-ray and upconversion nanophosphors will have significant potential for tumor targeted imaging.
Collapse
Affiliation(s)
- Hongyu Chen
- Department of Chemistry, Department of BioEngineering, Center for Optical Materials Science and Engineering Technologies (COMSET), and Institute of Environmental Toxicology (CU-ENTOX); Clemson University, Clemson, SC, 29634, USA. Tel:+1-864-656-1726.
| | - Fenglin Wang
- Department of Chemistry, Department of BioEngineering, Center for Optical Materials Science and Engineering Technologies (COMSET), and Institute of Environmental Toxicology (CU-ENTOX); Clemson University, Clemson, SC, 29634, USA. Tel:+1-864-656-1726.
| | - Thomas Moore
- Department of Bioengineering, Clemson University, Clemson, SC, 29634, USA
| | - Bin Qi
- Department of Materials Science and Engineering and Center for Optical Materials Science and Engineering Technologies (COMSET), Clemson University, Clemson, SC, 29634, USA
| | - Dino Sulejmanovic
- Department of Chemistry, Clemson University, Clemson, SC, 29634, USA
| | - Shiou-Jyh Hwu
- Department of Chemistry, Clemson University, Clemson, SC, 29634, USA
| | - O Thompson Mefford
- Department of Materials Science and Engineering and Center for Optical Materials Science and Engineering Technologies (COMSET), Clemson University, Clemson, SC, 29634, USA
| | - Frank Alexis
- Department of Bioengineering, Clemson University, Clemson, SC, 29634, USA
| | - Jeffrey N Anker
- Department of Chemistry, Department of BioEngineering, Center for Optical Materials Science and Engineering Technologies (COMSET), and Institute of Environmental Toxicology (CU-ENTOX); Clemson University, Clemson, SC, 29634, USA. Tel:+1-864-656-1726.
| |
Collapse
|
27
|
Liu W, Gómez-Durán CFA, Smith BD. Fluorescent Neuraminidase Assay Based on Supramolecular Dye Capture After Enzymatic Cleavage. J Am Chem Soc 2017; 139:6390-6395. [PMID: 28426220 DOI: 10.1021/jacs.7b01628] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A conceptually new type of enzymatic cleavage assay is reported that utilizes in situ supramolecular capture of the fluorescent product. A squaraine-derived substrate with large blocking groups at each end of its structure cannot be threaded by a tetralactam macrocycle until the blocking groups are removed by enzyme cleavage. A prototype design responds to viral neuraminidase, an indicator of influenza infection, and also measures susceptibility of the sample to neuraminidase inhibitor drugs. The substrate structure incorporates three key features: (a) a bis(4-amino-3-hydroxyphenyl)squaraine core with bright deep-red fluorescence and excellent photostability, (b) an N-methyl group at each end of the squaraine core that ensures fast macrocycle threading kinetics, and (c) sialic acid blocking groups that prevent macrocycle threading until they are removed by viral neuraminidase. The enzyme assay can be conducted in aqueous solution where dramatic colorimetric and fluorescence changes are easily observed by the naked eye. Alternatively, affinity capture beads coated with macrocycle can be used to immobilize the liberated squaraine and enable a range of heterogeneous analysis options. With further optimization, this new type of neuraminidase assay may be useful in a point of care clinic to rapidly diagnose influenza infection and also determine which of the approved antiviral inhibitor drugs is likely to be the most effective treatment for an individual patient. The assay design is generalizable and can be readily modified to monitor virtually any type of enzyme-catalyzed cleavage reaction.
Collapse
Affiliation(s)
- Wenqi Liu
- Department of Chemistry and Biochemistry, University of Notre Dame , 236 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - César F A Gómez-Durán
- Department of Chemistry and Biochemistry, University of Notre Dame , 236 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Bradley D Smith
- Department of Chemistry and Biochemistry, University of Notre Dame , 236 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| |
Collapse
|
28
|
Gul S. Epigenetic assays for chemical biology and drug discovery. Clin Epigenetics 2017; 9:41. [PMID: 28439316 PMCID: PMC5399855 DOI: 10.1186/s13148-017-0342-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 04/12/2017] [Indexed: 12/27/2022] Open
Abstract
The implication of epigenetic abnormalities in many diseases and the approval of a number of compounds that modulate specific epigenetic targets in a therapeutically relevant manner in cancer specifically confirms that some of these targets are druggable by small molecules. Furthermore, a number of compounds are currently in clinical trials for other diseases including cardiovascular, neurological and metabolic disorders. Despite these advances, the approved treatments for cancer only extend progression-free survival for a relatively short time and being associated with significant side effects. The current clinical trials involving the next generation of epigenetic drugs may address the disadvantages of the currently approved epigenetic drugs. The identification of chemical starting points of many drugs often makes use of screening in vitro assays against libraries of synthetic or natural products. These assays can be biochemical (using purified protein) or cell-based (using for example, genetically modified, cancer cell lines or primary cells) and performed in microtiter plates, thus enabling a large number of samples to be tested. A considerable number of such assays are available to monitor epigenetic target activity, and this review provides an overview of drug discovery and chemical biology and describes assays that monitor activities of histone deacetylase, lysine-specific demethylase, histone methyltransferase, histone acetyltransferase and bromodomain. It is of critical importance that an appropriate assay is developed and comprehensively validated for a given drug target prior to screening in order to improve the probability of the compound progressing in the drug discovery value chain.
Collapse
Affiliation(s)
- Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology - ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| |
Collapse
|
29
|
Bernetti M, Cavalli A, Mollica L. Protein-ligand (un)binding kinetics as a new paradigm for drug discovery at the crossroad between experiments and modelling. MEDCHEMCOMM 2017; 8:534-550. [PMID: 30108770 PMCID: PMC6072069 DOI: 10.1039/c6md00581k] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/25/2017] [Indexed: 12/14/2022]
Abstract
In the last three decades, protein and nucleic acid structure determination and comprehension of the mechanisms, leading to their physiological and pathological functions, have become a cornerstone of biomedical sciences. A deep understanding of the principles governing the fates of cells and tissue at the molecular level has been gained over the years, offering a solid basis for the rational design of drugs aimed at the pharmacological treatment of numerous diseases. Historically, affinity indicators (i.e. Kd and IC50/EC50) have been assumed to be valid indicators of the in vivo efficacy of a drug. However, recent studies pointed out that the kinetics of the drug-receptor binding process could be as important or even more important than affinity in determining the drug efficacy. This eventually led to a growing interest in the characterisation and prediction of the rate constants of protein-ligand association and dissociation. For instance, a drug with a longer residence time can kinetically select a given receptor over another, even if the affinity for both receptors is comparable, thus increasing its therapeutic index. Therefore, understanding the molecular features underlying binding and unbinding processes is of central interest towards the rational control of drug binding kinetics. In this review, we report the theoretical framework behind protein-ligand association and highlight the latest advances in the experimental and computational approaches exploited to investigate the binding kinetics.
Collapse
Affiliation(s)
- M Bernetti
- Department of Pharmacy and Biotechnology , University of Bologna , via Belmeloro 6 , 40126 Bologna , Italy
- CompuNet , Istituto Italiano di Tecnologia , via Morego 30 , 16163 Genova , Italy .
| | - A Cavalli
- Department of Pharmacy and Biotechnology , University of Bologna , via Belmeloro 6 , 40126 Bologna , Italy
- CompuNet , Istituto Italiano di Tecnologia , via Morego 30 , 16163 Genova , Italy .
| | - L Mollica
- CompuNet , Istituto Italiano di Tecnologia , via Morego 30 , 16163 Genova , Italy .
| |
Collapse
|
30
|
Dynamics of ligand binding to GPCR: Residence time of melanocortins and its modulation. Pharmacol Res 2016; 113:747-753. [DOI: 10.1016/j.phrs.2016.05.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/22/2016] [Accepted: 05/27/2016] [Indexed: 01/06/2023]
|
31
|
Development of a Novel High-Density [3H]Hypoxanthine Scintillation Proximity Assay To Assess Plasmodium falciparum Growth. Antimicrob Agents Chemother 2016; 60:5949-56. [PMID: 27458216 DOI: 10.1128/aac.00433-16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/09/2016] [Indexed: 12/30/2022] Open
Abstract
The discovery and development of new antimalarial drugs are becoming imperative because of the spread of resistance to current clinical treatments. The lack of robustly validated antimalarial targets and the difficulties with the building in of whole-cell activity in screening hits are hampering target-based approaches. However, phenotypic screens of structurally diverse molecule libraries are offering new opportunities for the identification of novel antimalarials. Several methodologies can be used to determine the whole-cell in vitro potencies of antimalarial hits. The [(3)H]hypoxanthine incorporation assay is considered the "gold standard" assay for measurement of the activity of antimalarial compounds against intraerythrocytic forms of Plasmodium falciparum However, the method has important limitations, as the assay is not amenable for high-throughput screening since it remains associated with the 96-well plate format. We have overcome this drawback by adapting the [(3)H]hypoxanthine incorporation method to a 384-well high-density format by coupling a homogeneous scintillation proximity assay (SPA) and thus eliminating the limiting filtration step. This SPA has been validated using a diverse set of 1,000 molecules, including both a representative set from the Tres Cantos Antimalarial Set (TCAMS) of compounds and molecules inactive against whole cells. The results were compared with those from the P. falciparum lactate dehydrogenase whole-cell assay, another method that is well established as a surrogate for parasite growth and is amenable for high-throughput screening. The results obtained demonstrate that the SPA-based [(3)H]hypoxanthine incorporation assay is a suitable design that is adaptable to high-throughput antimalarial drug screening and that maintains the features, robustness, and reliability of the standard filtration hypoxanthine incorporation method.
Collapse
|
32
|
Meleza C, Thomasson B, Ramachandran C, O'Neill JW, Michelsen K, Lo MC. Development of a scintillation proximity binding assay for high-throughput screening of hematopoietic prostaglandin D2 synthase. Anal Biochem 2016; 511:17-23. [PMID: 27485270 DOI: 10.1016/j.ab.2016.07.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/25/2016] [Accepted: 07/28/2016] [Indexed: 01/22/2023]
Abstract
Prostaglandin D2 synthase (PGDS) catalyzes the isomerization of prostaglandin H2 (PGH2) to prostaglandin D2 (PGD2). PGD2 produced by hematopoietic prostaglandin D2 synthase (H-PGDS) in mast cells and Th2 cells is proposed to be a mediator of allergic and inflammatory responses. Consequently, inhibitors of H-PGDS represent potential therapeutic agents for the treatment of inflammatory diseases such as asthma. Due to the instability of the PGDS substrate PGH2, an in-vitro enzymatic assay is not feasible for large-scale screening of H-PGDS inhibitors. Herein, we report the development of a competition binding assay amenable to high-throughput screening (HTS) in a scintillation proximity assay (SPA) format. This assay was used to screen an in-house compound library of approximately 280,000 compounds for novel H-PGDS inhibitors. The hit rate of the H-PGDS primary screen was found to be 4%. This high hit rate suggests that the active site of H-PGDS can accommodate a large diversity of chemical scaffolds. For hit prioritization, these initial hits were rescreened at a lower concentration in SPA and tested in the LAD2 cell assay. 116 compounds were active in both assays with IC50s ranging from 6 to 807 nM in SPA and 82 nM to 10 μM in the LAD2 cell assay.
Collapse
Affiliation(s)
- Cesar Meleza
- Discovery Technologies, Amgen Inc., South San Francisco, CA 94080, USA
| | | | | | | | - Klaus Michelsen
- Discovery Attribute Sciences, Amgen Inc., Cambridge, MA 02141, USA
| | - Mei-Chu Lo
- Discovery Technologies, Amgen Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
33
|
Functional Stability of the Human Kappa Opioid Receptor Reconstituted in Nanodiscs Revealed by a Time-Resolved Scintillation Proximity Assay. PLoS One 2016; 11:e0150658. [PMID: 27035823 PMCID: PMC4817975 DOI: 10.1371/journal.pone.0150658] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 02/16/2016] [Indexed: 11/19/2022] Open
Abstract
Long-term functional stability of isolated membrane proteins is crucial for many in vitro applications used to elucidate molecular mechanisms, and used for drug screening platforms in modern pharmaceutical industry. Compared to soluble proteins, the understanding at the molecular level of membrane proteins remains a challenge. This is partly due to the difficulty to isolate and simultaneously maintain their structural and functional stability, because of their hydrophobic nature. Here we show, how scintillation proximity assay can be used to analyze time-resolved high-affinity ligand binding to membrane proteins solubilized in various environments. The assay was used to establish conditions that preserved the biological function of isolated human kappa opioid receptor. In detergent solution the receptor lost high-affinity ligand binding to a radiolabelled ligand within minutes at room temperature. After reconstitution in Nanodiscs made of phospholipid bilayer the half-life of high-affinity ligand binding to the majority of receptors increased 70-fold compared to detergent solubilized receptors—a level of stability that is appropriate for further downstream applications. Time-resolved scintillation proximity assay has the potential to screen numerous conditions in parallel to obtain high levels of stable and active membrane proteins, which are intrinsically unstable in detergent solution, and with minimum material consumption.
Collapse
|
34
|
Abstract
Radioligand binding assays provide sensitive and quantitative information about guanine nucleotide protein G protein-coupled receptor (GPCR) expression and affinity for a wide variety of ligands, making them essential for drug structure-activity studies and basic GPCR research. Three basic radioligand binding protocols, saturation, indirect (competition, displacement, or modulation), and kinetic binding assays, are used to assess GPCR expression (Bmax), equilibrium dissociation constants for radioligands (Kd) and nonradioactive ligands (Ki), association and dissociation rates, and to distinguish competitive and allosteric mechanisms of GPCR-ligand interactions. Nonspecific radioligand binding may be mitigated by appropriate choices of reaction conditions. Radioligand depletion (bound radioactivity >10% of total radioligand), which compromises accuracy of Kd and Ki measurements, can be limited by adjusting receptor concentration and appropriate radioligand choice. Accurate Kd and Ki values in saturation and indirect binding assays depend on binding equilibrium. Equilibration time for high-affinity ligands, with slow dissociation rates, may require much extended incubation times or increased incubation temperature.
Collapse
Affiliation(s)
- Colleen A Flanagan
- School of Physiology and Medical Research Council Receptor Biology Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Wits Parktown, Johannesburg, South Africa.
| |
Collapse
|
35
|
Kuhne S, Nøhr AC, Marek A, Elbert T, Klein AB, Bräuner-Osborne H, Wellendorph P, Pedersen DS. Radiosynthesis and characterisation of a potent and selective GPR139 agonist radioligand. RSC Adv 2016. [DOI: 10.1039/c5ra21326f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Compound 1 is a selective and potent agonist of the G protein-coupled receptor GPR139 (EC50 = 39 nM).
Collapse
Affiliation(s)
- Sebastiaan Kuhne
- Department of Drug Design and Pharmacology
- Faculty of Health and Medical Sciences
- University of Copenhagen
- Denmark
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS)
| | - Anne Cathrine Nøhr
- Department of Drug Design and Pharmacology
- Faculty of Health and Medical Sciences
- University of Copenhagen
- Denmark
| | - Aleš Marek
- Institute of Organic Chemistry and Biochemistry
- Academy of Sciences of the Czech Republic
- Prague 6
- Czech Republic
| | - Tomáš Elbert
- Institute of Organic Chemistry and Biochemistry
- Academy of Sciences of the Czech Republic
- Prague 6
- Czech Republic
| | - Anders Bue Klein
- Department of Drug Design and Pharmacology
- Faculty of Health and Medical Sciences
- University of Copenhagen
- Denmark
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology
- Faculty of Health and Medical Sciences
- University of Copenhagen
- Denmark
| | - Petrine Wellendorph
- Department of Drug Design and Pharmacology
- Faculty of Health and Medical Sciences
- University of Copenhagen
- Denmark
| | - Daniel Sejer Pedersen
- Department of Drug Design and Pharmacology
- Faculty of Health and Medical Sciences
- University of Copenhagen
- Denmark
| |
Collapse
|
36
|
Xia L, de Vries H, IJzerman AP, Heitman LH. Scintillation proximity assay (SPA) as a new approach to determine a ligand's kinetic profile. A case in point for the adenosine A1 receptor. Purinergic Signal 2015; 12:115-26. [PMID: 26647040 PMCID: PMC4749533 DOI: 10.1007/s11302-015-9485-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/19/2015] [Indexed: 01/11/2023] Open
Abstract
Scintillation proximity assay (SPA) is a radio-isotopic technology format used to measure a wide range of biological interactions, including drug-target binding affinity studies. The assay is homogeneous in nature, as it relies on a “mix and measure” format. It does not involve a filtration step to separate bound from free ligand as is the case in a traditional receptor-binding assay. For G protein-coupled receptors (GPCRs), it has been shown that optimal binding kinetics, next to a high affinity of a ligand, can result in more desirable pharmacological profiles. However, traditional techniques to assess kinetic parameters tend to be cumbersome and laborious. We thus aimed to evaluate whether SPA can be an alternative platform for real-time receptor-binding kinetic measurements on GPCRs. To do so, we first validated the SPA technology for equilibrium binding studies on a prototypic class A GPCR, the human adenosine A1 receptor (hA1R). Differently to classic kinetic studies, the SPA technology allowed us to study binding kinetic processes almost real time, which is impossible in the filtration assay. To demonstrate the reliability of this technology for kinetic purposes, we performed the so-called competition association experiments. The association and dissociation rate constants (kon and koff) of unlabeled hA1R ligands were reliably and quickly determined and agreed very well with the same parameters from a traditional filtration assay performed simultaneously. In conclusion, SPA is a very promising technique to determine the kinetic profile of the drug-target interaction. Its robustness and potential for high-throughput may render this technology a preferred choice for further kinetic studies.
Collapse
Affiliation(s)
- Lizi Xia
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Henk de Vries
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Ad P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands.
| | - Laura H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| |
Collapse
|
37
|
Carraher C, Dalziel J, Jordan MD, Christie DL, Newcomb RD, Kralicek AV. Towards an understanding of the structural basis for insect olfaction by odorant receptors. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2015; 66:31-41. [PMID: 26416146 DOI: 10.1016/j.ibmb.2015.09.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 09/16/2015] [Accepted: 09/16/2015] [Indexed: 06/05/2023]
Abstract
Insects have co-opted a unique family of seven transmembrane proteins for odour sensing. Odorant receptors are believed to have evolved from gustatory receptors somewhere at the base of the Hexapoda and have expanded substantially to become the dominant class of odour recognition elements within the Insecta. These odorant receptors comprise an obligate co-receptor, Orco, and one of a family of highly divergent odorant "tuning" receptors. The two subunits are thought to come together at some as-yet unknown stoichiometry to form a functional complex that is capable of both ionotropic and metabotropic signalling. While there are still no 3D structures for these proteins, site-directed mutagenesis, resonance energy transfer, and structural modelling efforts, all mainly on Drosophila odorant receptors, are beginning to inform hypotheses of their structures and how such complexes function in odour detection. Some of the loops, especially the second extracellular loop that has been suggested to form a lid over the binding pocket, and the extracellular regions of some transmembrane helices, especially the third and to a less extent the sixth and seventh, have been implicated in ligand recognition in tuning receptors. The possible interaction between Orco and tuning receptor subunits through the final intracellular loop and the adjacent transmembrane helices is thought to be important for transducing ligand binding into receptor activation. Potential phosphorylation sites and a calmodulin binding site in the second intracellular loop of Orco are also thought to be involved in regulating channel gating. A number of new methods have recently been developed to express and purify insect odorant receptor subunits in recombinant expression systems. These approaches are enabling high throughput screening of receptors for agonists and antagonists in cell-based formats, as well as producing protein for the application of biophysical methods to resolve the 3D structure of the subunits and their complexes.
Collapse
Affiliation(s)
- Colm Carraher
- The New Zealand Institute for Plant & Food Research Limited, Private Bag 92169, Auckland 1142, New Zealand
| | - Julie Dalziel
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Private Bag 11008, Palmerston North 4442, New Zealand
| | - Melissa D Jordan
- The New Zealand Institute for Plant & Food Research Limited, Private Bag 92169, Auckland 1142, New Zealand
| | - David L Christie
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Richard D Newcomb
- The New Zealand Institute for Plant & Food Research Limited, Private Bag 92169, Auckland 1142, New Zealand; School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Andrew V Kralicek
- The New Zealand Institute for Plant & Food Research Limited, Private Bag 92169, Auckland 1142, New Zealand.
| |
Collapse
|
38
|
Zhang H, Wu Q, Berezin MY. Fluorescence anisotropy (polarization): from drug screening to precision medicine. Expert Opin Drug Discov 2015; 10:1145-61. [PMID: 26289575 DOI: 10.1517/17460441.2015.1075001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Fluorescence anisotropy (FA) is one of the major established methods accepted by industry and regulatory agencies for understanding the mechanisms of drug action and selecting drug candidates utilizing a high-throughput format. AREAS COVERED This review covers the basics of FA and complementary methods, such as fluorescence lifetime anisotropy and their roles in the drug discovery process. The authors highlight the factors affecting FA readouts, fluorophore selection and instrumentation. Furthermore, the authors describe the recent development of a successful, commercially valuable FA assay for long QT syndrome drug toxicity to illustrate the role that FA can play in the early stages of drug discovery. EXPERT OPINION Despite the success in drug discovery, the FA-based technique experiences competitive pressure from other homogeneous assays. That being said, FA is an established yet rapidly developing technique, recognized by academic institutions, the pharmaceutical industry and regulatory agencies across the globe. The technical problems encountered in working with small molecules in homogeneous assays are largely solved, and new challenges come from more complex biological molecules and nanoparticles. With that, FA will remain one of the major work-horse techniques leading to precision (personalized) medicine.
Collapse
Affiliation(s)
- Hairong Zhang
- a 1 Washington University School of Medicine, Department of Radiology , St. Louis 63110, USA
| | - Qian Wu
- a 1 Washington University School of Medicine, Department of Radiology , St. Louis 63110, USA
| | - Mikhail Y Berezin
- a 1 Washington University School of Medicine, Department of Radiology , St. Louis 63110, USA.,b 2 Washington University School of Medicine, Institute of Materials Science and Engineering, Department of Radiology , 510 S. Kingshighway, Barnard Bldg, 6th floor, 6604A, St. Louis, MO, USA +1 314 747 0701 ; +1 314 747 5191 ;
| |
Collapse
|
39
|
Blanc E, Wagner P, Plaisier F, Schmitt M, Durroux T, Bourguignon JJ, Partiseti M, Dupuis E, Bihel F. Design and validation of a homogeneous time-resolved fluorescence cell-based assay targeting the ligand-gated ion channel 5-HT3A. Anal Biochem 2015; 484:105-12. [PMID: 25998104 DOI: 10.1016/j.ab.2015.03.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 03/16/2015] [Accepted: 03/24/2015] [Indexed: 11/29/2022]
Abstract
Ligand-gated ion channels (LGICs) are considered as attractive protein targets in the search for new therapeutic agents. Nowadays, this strategy involves the capability to screen large chemical libraries. We present a new Tag-lite ligand binding assay targeting LGICs on living cells. This technology combines the use of suicide enzyme tags fused to channels of interest with homogeneous time-resolved fluorescence (HTRF) as the detection readout. Using the 5-HT3 receptor as system model, we showed that the pharmacology of the HALO-5HT3 receptor was identical to that of the native receptor. After validation of the assay by using 5-HT3 agonists and antagonists of reference, a pilot screen enabled us to identify azelastine, a well-known histamine H1 antagonist, as a potent 5-HT3 antagonist. This interesting result was confirmed with electrophysiological experiments. The method described here is easy to implement and could be applicable for other LGICs, opening new ways for the screening of chemical libraries.
Collapse
Affiliation(s)
- Emilie Blanc
- Cisbio Bioassays, 30200 Codolet, France; Institut de Génomique Fonctionnelle, Département de Pharmacologie Moléculaire, CNRS UMR 5203, INSERM U661, Université Montpellier I et II, 34094 Montpellier Cedex 5, France
| | - Patrick Wagner
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200 CNRS/Université de Strasbourg, 67401 Illkirch, France
| | - Fabrice Plaisier
- Sanofi R&D, Research Center of Vitry/Alfortville, 94403 Vitry-sur-Seine, France
| | - Martine Schmitt
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200 CNRS/Université de Strasbourg, 67401 Illkirch, France
| | - Thierry Durroux
- Institut de Génomique Fonctionnelle, Département de Pharmacologie Moléculaire, CNRS UMR 5203, INSERM U661, Université Montpellier I et II, 34094 Montpellier Cedex 5, France
| | - Jean-Jacques Bourguignon
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200 CNRS/Université de Strasbourg, 67401 Illkirch, France
| | - Michel Partiseti
- Sanofi R&D, Research Center of Vitry/Alfortville, 94403 Vitry-sur-Seine, France
| | | | - Frederic Bihel
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200 CNRS/Université de Strasbourg, 67401 Illkirch, France.
| |
Collapse
|
40
|
Kopra K, Härmä H. Quenching resonance energy transfer (QRET): a single-label technique for inhibitor screening and interaction studies. N Biotechnol 2015; 32:575-80. [PMID: 25721971 DOI: 10.1016/j.nbt.2015.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 02/10/2015] [Accepted: 02/13/2015] [Indexed: 12/01/2022]
Abstract
The increased number of therapeutic targets has led to a growing need for screening methods enabling possible inhibitor compound selection. Information for new therapeutic targets has been found mostly from sequencing of the human genome but this knowledge cannot be directly converted into clinically relevant drug molecules. After target identification, the multistep drug development process takes many years and hundreds of millions of dollars are spent without certainty of the outcome. The first and the most critical step in the drug development process is hit selection. The optimal high throughput screening method should provide the highest possible number of true positive hits for further studies and lead discovery. The result should be achieved with low material consumption in a rapid and automated process. Radioactive label based methods are sensitive, but due to the problems arising from the radioactivity, luminescence-based methods have become increasingly popular in screening. In this review, the time-resolved luminescence based quenching resonance energy transfer (QRET) technique is discussed for primary screening.
Collapse
Affiliation(s)
- Kari Kopra
- Institute of Biomedicine, Department of Cell Biology and Anatomy, University of Turku, Kiinamyllynkatu 10, 3rd Floor, FI-20520 Turku, Finland.
| | - Harri Härmä
- Institute of Biomedicine, Department of Cell Biology and Anatomy, University of Turku, Kiinamyllynkatu 10, 3rd Floor, FI-20520 Turku, Finland
| |
Collapse
|
41
|
Peng G, Wang B, Meng X, Liu B, Luo R. Different dispersion polymerization strategies influence the quality of fluorescent poly (St-co-GMA) microspheres. J Appl Polym Sci 2015. [DOI: 10.1002/app.41927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Gang Peng
- Department of Chemistry and Material Science; Hengyang Normal University; Hengyang 421008 China
- Key Laboratory of Functional Organometallic Materials of Hunan Province College; Hengyang Normal University; Hengyang 421008 China
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences; Chengdu Sichuan 610041 China
| | - Bin Wang
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences; Chengdu Sichuan 610041 China
| | - Xu Meng
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences; Chengdu Sichuan 610041 China
| | - Bailing Liu
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences; Chengdu Sichuan 610041 China
| | - Rong Luo
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences; Chengdu Sichuan 610041 China
| |
Collapse
|
42
|
Yanamandra M, Mitra S, Giri A. Development and application of PI3K assays for novel drug discovery. Expert Opin Drug Discov 2014; 10:171-86. [DOI: 10.1517/17460441.2015.997205] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Mahesh Yanamandra
- 1Scientist, GVK Biosciences Private Ltd, Biology, Campus MLR 1, Survey Nos. 125 (part) and 126, IDA Mallapur, Hyderabad, Telangana, 500076, India
- 2Jawaharlal Nehru Technological University, Institute of Science and Technology, Centre for Biotechnology, Kukatpally, Hyderabad, Telangana, 500085, India
| | - Sayan Mitra
- 3GVK Biosciences Private Ltd, Biology, Campus MLR 1, Survey Nos. 125 (part) and 126, IDA Mallapur, Hyderabad, Telangana, 500076, India
| | - Archana Giri
- 4Jawaharlal Nehru Technological University, Institute of Science and Technology, Centre for Biotechnology, Kukatpally, Hyderabad, Telangana, 500085, India
| |
Collapse
|
43
|
Peng G, Meng X, Wang B, Liu B, Chen H. The Surface Characteristics of Chitosan Modified PSt-GMA Microspheres Influenced the Interactions and Properties of Immobilized Pepsin. JOURNAL OF MACROMOLECULAR SCIENCE PART A-PURE AND APPLIED CHEMISTRY 2014. [DOI: 10.1080/10601325.2014.976745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
44
|
Lee SH, Rhee HW, van Noort D, Lee HJ, Park HH, Shin IS, Hong JI, Park TH. Microfluidic bead-based sensing platform for monitoring kinase activity. Biosens Bioelectron 2014; 57:1-9. [DOI: 10.1016/j.bios.2014.01.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/18/2014] [Accepted: 01/20/2014] [Indexed: 10/25/2022]
|
45
|
Kashem MA, Wa C, Wolak JP, Grafos NS, Ryan KR, Sanville-Ross ML, Fogarty KE, Rybina IV, Shoultz A, Molinaro T, Desai SN, Rajan A, Huber JD, Nelson RM. A High-Throughput Scintillation Proximity Assay for Sphingosine-1-Phosphate Lyase. Assay Drug Dev Technol 2014; 12:293-302. [DOI: 10.1089/adt.2014.575] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Mohammed A. Kashem
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Chunling Wa
- Department of Biotherapeutics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - John P. Wolak
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Nicholas S. Grafos
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Kelli R. Ryan
- Department of Immunology and Inflammation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Mary L. Sanville-Ross
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Kylie E. Fogarty
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Irina V. Rybina
- Department of Biotherapeutics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Alycia Shoultz
- Department of Biotherapeutics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Teresa Molinaro
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Sudha N. Desai
- Department of Immunology and Inflammation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Anusha Rajan
- Department of Immunology and Inflammation, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - John D. Huber
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Richard M. Nelson
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| |
Collapse
|
46
|
Considerations for the design and reporting of enzyme assays in high-throughput screening applications. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.pisc.2013.12.001] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
47
|
Lieberman OJ, Orr MW, Wang Y, Lee VT. High-throughput screening using the differential radial capillary action of ligand assay identifies ebselen as an inhibitor of diguanylate cyclases. ACS Chem Biol 2014; 9:183-92. [PMID: 24134695 DOI: 10.1021/cb400485k] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The rise of bacterial resistance to traditional antibiotics has motivated recent efforts to identify new drug candidates that target virulence factors or their regulatory pathways. One such antivirulence target is the cyclic-di-GMP (cdiGMP) signaling pathway, which regulates biofilm formation, motility, and pathogenesis. Pseudomonas aeruginosa is an important opportunistic pathogen that utilizes cdiGMP-regulated polysaccharides, including alginate and pellicle polysaccharide (PEL), to mediate virulence and antibiotic resistance. CdiGMP activates PEL and alginate biosynthesis by binding to specific receptors including PelD and Alg44. Mutations that abrogate cdiGMP binding to these receptors prevent polysaccharide production. Identification of small molecules that can inhibit cdiGMP binding to the allosteric sites on these proteins could mimic binding defective mutants and potentially reduce biofilm formation or alginate secretion. Here, we report the development of a rapid and quantitative high-throughput screen for inhibitors of protein-cdiGMP interactions based on the differential radial capillary action of ligand assay (DRaCALA). Using this approach, we identified ebselen as an inhibitor of cdiGMP binding to receptors containing an RxxD domain including PelD and diguanylate cyclases (DGC). Ebselen reduces diguanylate cyclase activity by covalently modifying cysteine residues. Ebselen oxide, the selenone analogue of ebselen, also inhibits cdiGMP binding through the same covalent mechanism. Ebselen and ebselen oxide inhibit cdiGMP regulation of biofilm formation and flagella-mediated motility in P. aeruginosa through inhibition of diguanylate cyclases. The identification of ebselen provides a proof-of-principle that a DRaCALA high-throughput screening approach can be used to identify bioactive agents that reverse regulation of cdiGMP signaling by targeting cdiGMP-binding domains.
Collapse
Affiliation(s)
- Ori J. Lieberman
- Department
of Cell Biology and Molecular Genetics, University of Maryland College Park, 3114 Bioscience Research Building, College
Park, Maryland 20742, United States
- Maryland Pathogen Research Institute, Bioscience Research Building, College Park, Maryland 20742, United States
| | - Mona W. Orr
- Department
of Cell Biology and Molecular Genetics, University of Maryland College Park, 3114 Bioscience Research Building, College
Park, Maryland 20742, United States
- Maryland Pathogen Research Institute, Bioscience Research Building, College Park, Maryland 20742, United States
| | - Yan Wang
- Proteomics
Core Facility, College of Computer, Mathematical and Natural Science, University of Maryland College Park, 0111 Biology Psychology Building College Park, Maryland 20742, United States
| | - Vincent T. Lee
- Department
of Cell Biology and Molecular Genetics, University of Maryland College Park, 3114 Bioscience Research Building, College
Park, Maryland 20742, United States
- Maryland Pathogen Research Institute, Bioscience Research Building, College Park, Maryland 20742, United States
| |
Collapse
|
48
|
Present and future approaches to screening of G-protein-coupled receptors. Future Med Chem 2013; 5:523-38. [PMID: 23573971 DOI: 10.4155/fmc.13.9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
As G-protein-coupled receptors (GPCRs) mediate a multitude of cellular signal transduction events, affecting more or less all human disease areas, it is, therefore, no surprise that they comprise the largest family of current drug targets. Screening of compounds interacting with GPCRs has developed during the past decade from receptor binding assays, to various functional determination of coupling to G-proteins, and, more recently, G-protein-independent signal transduction events. Additional opportunities have been presented in drug discovery through novel pharmacological properties obtained for receptor dimers and by identification of ligands for orphan GPCRs. Furthermore, high-throughput formats and automation has substantially facilitated and accelerated the screening process providing powerful tools in improving modern drug discovery.
Collapse
|
49
|
Li S, Gu XJ, Hao Q, Fan H, Li L, Zhou S, Zhao K, Chan HM, Wang YK. A liquid chromatography/mass spectrometry-based generic detection method for biochemical assay and hit discovery of histone methyltransferases. Anal Biochem 2013; 443:214-21. [PMID: 24018340 DOI: 10.1016/j.ab.2013.08.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/30/2013] [Indexed: 11/19/2022]
Abstract
Epigenetic modifications of the genome, such as DNA methylation and posttranslational modifications of histone proteins, contribute to gene regulation. Growing evidence suggests that histone methyltransferases are associated with the development of various human diseases, including cancer, and are promising drug targets. High-quality generic assays will facilitate drug discovery efforts in this area. In this article, we present a liquid chromatography/mass spectrometry (LC/MS)-based S-adenosyl homocysteine (SAH) detection assay for histone methyltransferases (HMTs) and its applications in HMT drug discovery, including analyzing the activity of newly produced enzymes, developing and optimizing assays, performing focused compound library screens and orthogonal assays for hit confirmations, selectivity profiling against a panel of HMTs, and studying mode of action of select hits. This LC/MS-based generic assay has become a critical platform for our methyltransferase drug discovery efforts.
Collapse
Affiliation(s)
- Shu Li
- Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Beyleveld G, White KM, Ayllon J, Shaw ML. New-generation screening assays for the detection of anti-influenza compounds targeting viral and host functions. Antiviral Res 2013; 100:120-32. [PMID: 23933115 DOI: 10.1016/j.antiviral.2013.07.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 07/23/2013] [Accepted: 07/26/2013] [Indexed: 01/08/2023]
Abstract
Current options for influenza antiviral therapy are limited to the neuraminidase inhibitors, and knowledge that high levels of oseltamivir resistance have been seen among previously circulating H1N1 viruses increases the urgency to find new influenza therapeutics. To feed this pipeline, assays that are appropriate for use in high-throughput screens are being developed and are discussed in this review. Particular emphasis is placed on cell-based assays that capture both inhibitors of viral functions as well as the host functions that facilitate optimal influenza virus replication. Success in this area has been fueled by a greater understanding of the genome structure of influenza viruses and the ability to generate replication-competent recombinant viruses that carry a reporter gene, allowing for easy monitoring of viral infection in a high-throughput setting. This article forms part of a symposium in Antiviral Research on "Treatment of influenza: targeting the virus or the host."
Collapse
Affiliation(s)
- Grant Beyleveld
- Department of Microbiology and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | |
Collapse
|