1
|
Investigating the ligand agonism and antagonism at the D 2long receptor by dynamic mass redistribution. Sci Rep 2022; 12:9637. [PMID: 35688965 PMCID: PMC9187652 DOI: 10.1038/s41598-022-14311-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/06/2022] [Indexed: 11/09/2022] Open
Abstract
The signalling of the D2 receptor (D2R), a G protein-coupled receptor (GPCR), is a complex process consisting of various components. For the screening of D2R ligands, methods quantifying distinct second messengers such as cAMP or the interaction of the receptor with β-arrestin, are commonly employed. In contrast, a label-free biosensor technology like dynamic mass redistribution (DMR), where it is mostly unknown how the individual signalling pathways contribute to the DMR signal, provides a holistic readout of the complex cellular response. In this study, we report the successful application of the DMR technology to CHO-K1 cells stably expressing the human dopamine D2long receptor. In real-time kinetic experiments, studies of D2R reference compounds yielded results for agonists and antagonists that were consistent with those obtained by conventional methods and also allowed a discrimination between partial and full agonists. Furthermore, investigations on the signalling pathway in CHO-K1 hD2longR cells identified the Gαi/o protein as the main proximal trigger of the observed DMR response. The present study has shown that the DMR technology is a valuable method for the characterisation of putative new ligands and, due to its label-free nature, suggests its use for deorphanisation studies of GPCRs.
Collapse
|
2
|
David F, Davis AM, Gossing M, Hayes MA, Romero E, Scott LH, Wigglesworth MJ. A Perspective on Synthetic Biology in Drug Discovery and Development-Current Impact and Future Opportunities. SLAS DISCOVERY 2021; 26:581-603. [PMID: 33834873 DOI: 10.1177/24725552211000669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The global impact of synthetic biology has been accelerating, because of the plummeting cost of DNA synthesis, advances in genetic engineering, growing understanding of genome organization, and explosion in data science. However, much of the discipline's application in the pharmaceutical industry remains enigmatic. In this review, we highlight recent examples of the impact of synthetic biology on target validation, assay development, hit finding, lead optimization, and chemical synthesis, through to the development of cellular therapeutics. We also highlight the availability of tools and technologies driving the discipline. Synthetic biology is certainly impacting all stages of drug discovery and development, and the recognition of the discipline's contribution can further enhance the opportunities for the drug discovery and development value chain.
Collapse
Affiliation(s)
- Florian David
- Department of Biology and Biological Engineering, Division of Systems and Synthetic Biology, Chalmers University of Technology, Gothenburg, Sweden
| | - Andrew M Davis
- Discovery Sciences, Biopharmaceutical R&D, AstraZeneca, Cambridge, UK
| | - Michael Gossing
- Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Martin A Hayes
- Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Elvira Romero
- Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Louis H Scott
- Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | |
Collapse
|
3
|
Kargaran PK, Mosqueira D, Kozicz T. Mitochondrial Medicine: Genetic Underpinnings and Disease Modeling Using Induced Pluripotent Stem Cell Technology. Front Cardiovasc Med 2021; 7:604581. [PMID: 33585579 PMCID: PMC7874022 DOI: 10.3389/fcvm.2020.604581] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial medicine is an exciting and rapidly evolving field. While the mitochondrial genome is small and differs from the nuclear genome in that it is circular and free of histones, it has been implicated in neurodegenerative diseases, type 2 diabetes, aging and cardiovascular disorders. Currently, there is a lack of efficient treatments for mitochondrial diseases. This has promoted the need for developing an appropriate platform to investigate and target the mitochondrial genome. However, developing these therapeutics requires a model system that enables rapid and effective studying of potential candidate therapeutics. In the past decade, induced pluripotent stem cells (iPSCs) have become a promising technology for applications in basic science and clinical trials, and have the potential to be transformative for mitochondrial drug development. Engineered iPSC-derived cardiomyocytes (iPSC-CM) offer a unique tool to model mitochondrial disorders. Additionally, these cellular models enable the discovery and testing of novel therapeutics and their impact on pathogenic mtDNA variants and dysfunctional mitochondria. Herein, we review recent advances in iPSC-CM models focused on mitochondrial dysfunction often causing cardiovascular diseases. The importance of mitochondrial disease systems biology coupled with genetically encoded NAD+/NADH sensors is addressed toward developing an in vitro translational approach to establish effective therapies.
Collapse
Affiliation(s)
- Parisa K Kargaran
- Department of Cardiovascular Medicine, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Diogo Mosqueira
- Division of Cancer & Stem Cells, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Tamas Kozicz
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
4
|
Yu X, Scott D, Dikici E, Joel S, Deo S, Daunert S. Multiplexing cytokine analysis: towards reducing sample volume needs in clinical diagnostics. Analyst 2019; 144:3250-3259. [PMID: 31049499 PMCID: PMC11401509 DOI: 10.1039/c9an00297a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2024]
Abstract
The trend for improved more precise diagnostics and management of disease heavily relies on the measurement of panels of biomarkers in physiological samples of patients. Ideally, the ultimate goal would be to detect as many clinically relevant biomarkers as possible in a single drop of blood, achieving quick, sensitive, reproducible, and affordable detection in small volume physiological samples. Bioluminescent (BL) proteins provide many of the desired characteristics required for such labels, including detection at extremely low concentrations, no interference from physiological fluids leading to excellent detection limits, and compatibility with many miniaturized systems. However, to date the use of BL proteins has been restricted by their limited multiplexing capabilities. BL proteins typically exhibit a single emission profile and decay kinetics making the simultaneous detection of multiple analytes difficult. Recent progresses in this area include the use of two different engineered luminescent proteins to achieve resolved signals via one-dimensional time resolution. This approach, however, to date only lead to a dual analyte detection. Herein, we have demonstrated that using a two-dimensional approach that combines both temporal and spatial resolution, we can expand the multiplexing capabilities of bioluminescent proteins. To that end, the photoprotein aequorin (AEQ) has been employed for the simultaneous detection of three separate analytes in a single well, differentiated through the use of three discrete time/wavelength windows. Through a combination of site-specific mutations and synthetic coelenterazines "semi-synthetic" AEQ variants have been developed with altered emission profiles and decay kinetics. In this study, two AEQ mutant proteins were genetically conjugated to three pro-inflammatory cytokines (tumor necrosis factor alpha, interleukins 6 and 8) resulting in AEQ-labeled cytokines. These fusion proteins were combined with synthetic coelenterazines resulting in proteins with differing emission maxima and half-lives to allow for the simultaneous detection of all three cytokines in a single sample. The validity of the assay was demonstrated in serum by employing human physiological samples and comparing our results with commercially available individual tests for each of the three cytokines.
Collapse
Affiliation(s)
- Xiaowen Yu
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136, USA.
| | | | | | | | | | | |
Collapse
|
5
|
Sharifian S, Homaei A, Hemmati R, B Luwor R, Khajeh K. The emerging use of bioluminescence in medical research. Biomed Pharmacother 2018; 101:74-86. [PMID: 29477474 DOI: 10.1016/j.biopha.2018.02.065] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 02/13/2018] [Accepted: 02/15/2018] [Indexed: 01/01/2023] Open
Abstract
Bioluminescence is the light produced by a living organism and is commonly emitted by sea life with Ca2+-regulated photoproteins being the most responsible for bioluminescence emission. Marine coelenterates provide important functions involved in essential purposes such as defense, feeding, and breeding. In this review, the main characteristics of marine photoproteins including aequorin, clytin, obelin, berovin, pholasin and symplectin from different marine organisms will be discussed. We will focused on the recent use of recombinant photoproteins in different biomedical research fields including the measurement of Ca2+ in different intracellular compartments of animal cells, as labels in the design and development of binding assays. This review will also outline how bioluminescent photoproteins have been used in a plethora of analytical methods including ultra-sensitive assays and in vivo imaging of cellular processes. Due to their unique properties including elective intracellular distribution, wide dynamic range, high signal-to-noise ratio and low Ca2+-buffering effect, recombinant photoproteins represent a promising future analytical tool in several in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Sana Sharifian
- Department of Marine Biology, Faculty of Sciences, University of Hormozgan, Bandar Abbas, Iran
| | - Ahmad Homaei
- Department of Biochemistry, Faculty of Sciences, University of Hormozgan, Bandar Abbas, Iran.
| | - Roohullah Hemmati
- Department of Biology, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Rodney B Luwor
- Department of Surgery, Level 5, Clinical Sciences Building, The University of Melbourne, The Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia
| | - Khosro Khajeh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
6
|
Abstract
In the past decade, automated microscopy has become an important tool for the drug discovery and development process. The establishment of imaging modalities as screening tools depended on technological breakthroughs in the domain of automated microscopy and automated image analysis. These types of assays are often referred to as high content screening or high content analysis (HCS/HCA). The driving force to adopt imaging for drug development is the quantity and quality of cellular information that can be collected and the enhanced physiological relevance of cellular screening compared to biochemical screening. Most imaging in drug development is performed on fixed cells as this allows uncoupling the preparation of the cells from the acquisition of the images. Live-cell imaging is technically challenging, but is very useful for many aspects of the drug development pipeline such as kinetic studies of compound mode of action or to analyze the motion of cellular components. Most vendors of HCS microscopy systems offer the option of environmental chambers and onboard pipetting on their platforms. This reflects the wish and desire of many customers to have the ability to perform live-cell assays on their HCS automated microscopes. This book chapter summarizes the challenges and advantages of live-cell imaging in drug discovery. Examples of applications are presented and the motivation to perform these assays in kinetic mode is discussed.
Collapse
Affiliation(s)
- Milan Esner
- High Throughput Technology Development Studio (HT-TDS), Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, 625 00, Brno, Czech Republic
| | - Felix Meyenhofer
- High Throughput Technology Development Studio (HT-TDS), Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Département de Médecine, Faculté des Sciences, University of Fribourg, 1, Rte., Albert Gockel, Fribourg, 1700, Switzerland
| | - Marc Bickle
- High Throughput Technology Development Studio (HT-TDS), Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany.
| |
Collapse
|
7
|
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
|
8
|
Alieva RR, Kudryasheva NS. Variability of fluorescence spectra of coelenteramide-containing proteins as a basis for toxicity monitoring. Talanta 2017; 170:425-431. [PMID: 28501192 DOI: 10.1016/j.talanta.2017.04.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 04/10/2017] [Accepted: 04/16/2017] [Indexed: 11/30/2022]
Abstract
Nowadays, physicochemical approach to understanding toxic effects remains underdeveloped. A proper development of such mode would be concerned with simplest bioassay systems. Coelenteramide-Containing Fluorescent Proteins (CLM-CFPs) can serve as proper tools for study primary physicochemical processes in organisms under external exposures. CLM-CFPs are products of bioluminescent reactions of marine coelenterates. As opposed to Green Fluorescent Proteins, the CLM-CFPs are not widely applied in biomedical research, and their potential as colored biomarkers is undervalued now. Coelenteramide, fluorophore of CLM-CFPs, is a photochemically active molecule; it acts as a proton donor in its electron-excited states, generating several forms of different fluorescent state energy and, hence, different fluorescence color, from violet to green. Contributions of the forms to the visible fluorescence depend on the coelenteramide microenvironment in proteins. Hence, CLM-CFPs can serve as fluorescence biomarkers with color differentiation to monitor results of destructive biomolecule exposures. The paper reviews experimental and theoretical studies of spectral-luminescent and photochemical properties of CLM-CFPs, as well as their variation under different exposures - chemicals, temperature, and ionizing radiation. Application of CLM-CFPs as toxicity bioassays of a new type is justified.
Collapse
Affiliation(s)
- Roza R Alieva
- Institute of Biophysics SB RAS, Federal Research Center "Krasnoyarsk Science Center SB RAS", Akademgorodok 50/50, Krasnoyarsk 660036, Russia; Siberian Federal University, Svobodny Prospect 79, Krasnoyarsk 660041, Russia
| | - Nadezhda S Kudryasheva
- Institute of Biophysics SB RAS, Federal Research Center "Krasnoyarsk Science Center SB RAS", Akademgorodok 50/50, Krasnoyarsk 660036, Russia; Siberian Federal University, Svobodny Prospect 79, Krasnoyarsk 660041, Russia
| |
Collapse
|
9
|
Grinstead K, Joel S, Zingg JM, Dikici E, Daunert S. Enabling Aequorin for Biotechnology Applications Through Genetic Engineering. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2015:149-179. [PMID: 26475468 DOI: 10.1007/10_2015_336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In recent years, luminescent proteins have been studied for their potential application in a variety of detection systems. Bioluminescent proteins, which do not require an external excitation source, are especially well-suited as reporters in analytical detection. The photoprotein aequorin is a bioluminescent protein that can be engineered for use as a molecular reporter under a wide range of conditions while maintaining its sensitivity. Herein, the characteristics of aequorin as well as the engineering and production of aequorin variants and their impact on signal detection in biological systems are presented. The structural features and activity of aequorin, its benefits as a label for sensing and applications in highly sensitive detection, as well as in gaining insight into biological processes are discussed. Among those, focus has been placed on the highly sensitive calcium detection in vivo, in vitro DNA and small molecule sensing, and development of in vivo imaging technologies. Graphical Abstract.
Collapse
Affiliation(s)
- Kristen Grinstead
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Smita Joel
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Jean-Marc Zingg
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
10
|
Chen Y, Tang H, Seibel W, Papoian R, Li X, Lambert NA, Palczewski K. A High-Throughput Drug Screening Strategy for Detecting Rhodopsin P23H Mutant Rescue and Degradation. Invest Ophthalmol Vis Sci 2015; 56:2553-67. [PMID: 25783607 DOI: 10.1167/iovs.14-16298] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Inherent instability of the P23H mutant opsin accounts for approximately 10% of autosomal dominant retinitis pigmentosa cases. Our purpose was to develop an overall set of reliable screening strategies to assess if either stabilization or enhanced degradation of mutant rhodopsin could rescue rod photoreceptors expressing this mutant protein. These strategies promise to reveal active compounds and clarify molecular mechanisms of biologically important processes, such as inhibition of target degradation or enhanced target folding. METHODS Cell-based bioluminescence reporter assays were developed and validated for high-throughput screening (HTS) of compounds that promote either stabilization or degradation of P23H mutant opsin. Such assays were further complemented by immunoblotting and image-based analyses. RESULTS Two stabilization assays of P23H mutant opsin were developed and validated, one based on β-galactosidase complementarity and a second assay involving bioluminescence resonance energy transfer (BRET) technology. Moreover, two additional assays evaluating mutant protein degradation also were employed, one based on the disappearance of luminescence and another employing the ALPHA immunoassay. Imaging of cells revealed the cellular localization of mutant rhodopsin, whereas immunoblots identified changes in the aggregation and glycosylation of P23H mutant opsin. CONCLUSIONS Our findings indicate that these initial HTS and following assays can identify active therapeutic compounds, even for difficult targets such as mutant rhodopsin. The assays are readily scalable and their function has been proven with model compounds. High-throughput screening, supported by automated imaging and classic immunoassays, can further characterize multiple steps and pathways in the biosynthesis and degradation of this essential visual system protein.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Department of Pharmacology Case Western Reserve University, Cleveland, Ohio, United States
| | - Hong Tang
- Drug Discovery Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States
| | - William Seibel
- Drug Discovery Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States
| | - Ruben Papoian
- Drug Discovery Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States
| | - Xiaoyu Li
- Department of Pharmacology Case Western Reserve University, Cleveland, Ohio, United States
| | - Nevin A Lambert
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, Georgia, United States
| | - Krzysztof Palczewski
- Department of Pharmacology Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
11
|
Propensity scores for prediction and characterization of bioluminescent proteins from sequences. PLoS One 2014; 9:e97158. [PMID: 24828431 PMCID: PMC4020813 DOI: 10.1371/journal.pone.0097158] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/15/2014] [Indexed: 01/19/2023] Open
Abstract
Bioluminescent proteins (BLPs) are a class of proteins with various mechanisms of light emission such as bioluminescence and fluorescence from luminous organisms. While valuable for commercial and medical applications, identification of BLPs, including luciferases and fluorescent proteins (FPs), is rather challenging, owing to their high variety of protein sequences. Moreover, characterization of BLPs facilitates mutagenesis analysis to enhance bioluminescence and fluorescence. Therefore, this study proposes a novel methodological approach to estimating the propensity scores of 400 dipeptides and 20 amino acids in order to design two prediction methods and characterize BLPs based on a scoring card method (SCM). The SCMBLP method for predicting BLPs achieves an accuracy of 90.83% for 10-fold cross-validation higher than existing support vector machine based methods and a test accuracy of 82.85%. A dataset consisting of 269 luciferases and 216 FPs is also established to design the SCMLFP prediction method, which achieves training and test accuracies of 97.10% and 96.28%, respectively. Additionally, four informative physicochemical properties of 20 amino acids are identified using the estimated propensity scores to characterize BLPs as follows: 1) high transfer free energy from inside to the protein surface, 2) high occurrence frequency of residues in the transmembrane regions of the protein, 3) large hydrophobicity scale from the native protein structure, and 4) high correlation coefficient (R = 0.921) between the amino acid compositions of BLPs and integral membrane proteins. Further analyzing BLPs reveals that luciferases have a larger value of R (0.937) than FPs (0.635), suggesting that luciferases tend to locate near the cell membrane location rather than FPs for convenient receipt of extracellular ions. Importantly, the propensity scores of dipeptides and amino acids and the identified properties facilitate efforts to predict, characterize, and apply BLPs, including luciferases, photoproteins, and FPs. The web server is available at http://iclab.life.nctu.edu.tw/SCMBLP/index.html.
Collapse
|
12
|
Alieva RR, Belogurova NV, Petrova AS, Kudryasheva NS. Effects of alcohols on fluorescence intensity and color of a discharged-obelin-based biomarker. Anal Bioanal Chem 2014; 406:2965-74. [PMID: 24618986 DOI: 10.1007/s00216-014-7685-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/05/2014] [Accepted: 02/07/2014] [Indexed: 10/25/2022]
Abstract
Photoproteins are responsible for bioluminescence of marine coelenterates; bioluminescent and fluorescent biomarkers based on photoproteins are useful for monitoring of calcium-dependent processes in medical investigations. Here, we present the analysis of intensity and color of light-induced fluorescence of Ca(2+)-discharged photoprotein obelin in the presence of alcohols (ethanol and glycerol). Complex obelin spectra obtained at different concentrations of the alcohols at 350- and 280-nm excitation (corresponding to polypeptide-bound coelenteramide and tryptophan absorption regions) were deconvoluted into Gaussian components; fluorescent intensity and contributions of the components to experimental spectra were analyzed. Five Gaussian components were found in different spectral regions-ultraviolet (tryptophan emission), blue-green (coelenteramide emission), and red (hypothetical indole-coelenteramide exciplex emission). Inhibition coefficients and contributions of the components to experimental fluorescent spectra showed that presence of alcohols increased contributions of ultraviolet, violet, and red components, but decreased contributions of components in the blue-green region. The effects were related to (1) changes of proton transfer efficiency in fluorescent S*1 state of coelenteramide in the obelin active center and (2) formation of indole-coelenteramide exciplex at 280-nm photoexcitation. The data show that variation of fluorescence color and intensity in the presence of alcohols and dependence of emission spectra on excitation wavelength should be considered while applying the discharged obelin as a fluorescence biomarker.
Collapse
Affiliation(s)
- Roza R Alieva
- Siberian Federal University, Svobodny Prospect 79, 660041, Krasnoyarsk, Russia
| | | | | | | |
Collapse
|
13
|
Bioluminescent properties of obelin and aequorin with novel coelenterazine analogues. Anal Bioanal Chem 2014; 406:2695-707. [DOI: 10.1007/s00216-014-7656-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/28/2013] [Accepted: 01/23/2014] [Indexed: 10/25/2022]
|
14
|
Natashin PV, Ding W, Eremeeva EV, Markova SV, Lee J, Vysotski ES, Liu ZJ. Structures of the Ca2+-regulated photoprotein obelin Y138F mutant before and after bioluminescence support the catalytic function of a water molecule in the reaction. ACTA ACUST UNITED AC 2014; 70:720-32. [DOI: 10.1107/s1399004713032434] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 11/28/2013] [Indexed: 11/11/2022]
Abstract
Ca2+-regulated photoproteins, which are responsible for light emission in a variety of marine coelenterates, are a highly valuable tool for measuring Ca2+inside living cells. All of the photoproteins are a single-chain polypeptide to which a 2-hydroperoxycoelenterazine molecule is tightly but noncovalently bound. Bioluminescence results from the oxidative decarboxylation of 2-hydroperoxycoelenterazine, generating protein-bound coelenteramide in an excited state. Here, the crystal structures of the Y138F obelin mutant before and after bioluminescence are reported at 1.72 and 1.30 Å resolution, respectively. The comparison of the spatial structures of the conformational states of Y138F obelin with those of wild-type obelin gives clear evidence that the substitution of Tyr by Phe does not affect the overall structure of both Y138F obelin and its product following Ca2+discharge compared with the corresponding conformational states of wild-type obelin. Despite the similarity of the overall structures and internal cavities of Y138F and wild-type obelins, there is a substantial difference: in the cavity of Y138F obelin a water molecule corresponding to W2in wild-type obelin is not found. However, in Ca2+-discharged Y138F obelin this water molecule now appears in the same location. This finding, together with the observed much slower kinetics of Y138F obelin, clearly supports the hypothesis that the function of a water molecule in this location is to catalyze the 2-hydroperoxycoelenterazine decarboxylation reaction by protonation of a dioxetanone anion before its decomposition into the excited-state product. Although obelin differs from other hydromedusan Ca2+-regulated photoproteins in some of its properties, they are believed to share a common mechanism.
Collapse
|
15
|
Chen L, Jin L, Zhou N. An update of novel screening methods for GPCR in drug discovery. Expert Opin Drug Discov 2012; 7:791-806. [DOI: 10.1517/17460441.2012.699036] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
16
|
Abstract
G-protein-coupled receptors (GPCRs) mediate many important physiological functions and
are considered as one of the most successful therapeutic targets for a broad spectrum of
diseases. The design and implementation of high-throughput GPCR assays that allow the
cost-effective screening of large compound libraries to identify novel drug candidates are
critical in early drug discovery. Early functional GPCR assays depend primarily on the
measurement of G-protein-mediated 2nd messenger generation. Taking advantage of the
continuously deepening understanding of GPCR signal transduction, many
G-protein-independent pathways are utilized to detect the activity of GPCRs, and may
provide additional information on functional selectivity of candidate compounds. With the
combination of automated imaging systems and label-free detection systems, such assays are
now suitable for high-throughput screening (HTS). In this review, we summarize the most
widely used GPCR assays and recent advances in HTS technologies for GPCR drug
discovery.
Collapse
|
17
|
Markova SV, Burakova LP, Golz S, Malikova NP, Frank LA, Vysotski ES. The light-sensitive photoprotein berovin from the bioluminescent ctenophore Beroe abyssicola: a novel type of Ca2+-regulated photoprotein. FEBS J 2012; 279:856-70. [DOI: 10.1111/j.1742-4658.2012.08476.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
18
|
Bruton JD, Cheng AJ, Westerblad H. Methods to Detect Ca2+ in Living Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:27-43. [DOI: 10.1007/978-94-007-2888-2_2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
19
|
Eglen R, Reisine T. Primary cells and stem cells in drug discovery: emerging tools for high-throughput screening. Assay Drug Dev Technol 2010; 9:108-24. [PMID: 21186936 DOI: 10.1089/adt.2010.0305] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Many drug discovery screening programs employ immortalized cells, recombinantly engineered to express a defined molecular target. Several technologies are now emerging that render it feasible to employ more physiologically, and clinically relevant, cell phenotypes. Consequently, numerous approaches use primary cells, which retain many functions seen in vivo, as well as endogenously expressing the target of interest. Furthermore, stem cells, of either embryonic or adult origin, as well as those derived from differentiated cells, are now finding a place in drug discovery. Collectively, these cells are expanding the utility of authentic human cells, either as screening tools or as therapeutics, as well as providing cells derived directly from patients. Nonetheless, the growing use of phenotypically relevant cells (including primary cells or stem cells) is not without technical difficulties, particularly when their envisioned use lies in high-throughput screening (HTS) protocols. In particular, the limited availability of homogeneous primary or stem cell populations for HTS mandates that novel technologies be developed to accelerate their adoption. These technologies include detection of responses with very few cells as well as protocols to generate cell lines in abundant, homogeneous populations. In parallel, the growing use of changes in cell phenotype as the assay readout is driving greater use of high-throughput imaging techniques in screening. Taken together, the greater availability of novel primary and stem cell phenotypes as well as new detection technologies is heralding a new era of cellular screening. This convergence offers unique opportunities to identify drug candidates for disorders at which few therapeutics are presently available.
Collapse
Affiliation(s)
- Richard Eglen
- Bio-discovery, PerkinElmer, Waltham, Massachusetts 02451-1457, USA.
| | | |
Collapse
|
20
|
Charlton SJ, Vauquelin G. Elusive equilibrium: the challenge of interpreting receptor pharmacology using calcium assays. Br J Pharmacol 2010; 161:1250-65. [PMID: 20977466 PMCID: PMC3000651 DOI: 10.1111/j.1476-5381.2010.00863.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 03/29/2010] [Accepted: 04/12/2010] [Indexed: 11/28/2022] Open
Abstract
UNLABELLED Calcium is a key intracellular signal that controls manifold cellular processes over a wide temporal range. The development of calcium-sensitive fluorescent dyes and proteins revolutionized our ability to visualize this important second messenger and its complex signalling characteristics. The subsequent advent of high throughput plate-based fluorescence readers has resulted in the calcium assay becoming the most widely utilized assay system for the characterization of novel receptor ligands. In this review we discuss common approaches to calcium assays, paying particular attention to the potential issues associated with interpretation of receptor pharmacology using this system. Topics covered include dye saturation and forced-coupling of receptors to the calcium pathway, but special consideration is given to the influence of non-equilibrium conditions in this rapid signalling system. Modelling the calcium transient in a kinetic mode allows the influence of ligand kinetics, receptor reserve and read time to be explored. This demonstrates that observed ligand pharmacology at very early time points can be quite different to that determined after longer incubations, even resulting in reversal of agonist potency orders that may be misinterpreted as agonist biased signalling. It also shows that estimates of antagonist affinity, whether by Schild analysis or inhibition curves, are similarly affected by hemi-equilibrium conditions. Finally we end with a discussion on practical approaches to accurately estimate the affinity of insurmountable antagonists using calcium assays. LINKED ARTICLES This article is part of a themed section on Analytical Receptor Pharmacology in Drug Discovery. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2010.161.issue-6.
Collapse
|
21
|
Cainarca S, Fenu S, Bovolenta S, Arioli P, Menegon A, Lohmer S, Corazza S. From c-Photina® Mouse Embryonic Stem Cells to High-Throughput Screening of Differentiated Neural Cells via an Intermediate Step Enriched in Neural Precursor Cells. ACTA ACUST UNITED AC 2010; 15:1132-43. [DOI: 10.1177/1087057110379267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The use of engineered mouse embryonic stem (mES) cells in high-throughput screening (HTS) can offer new opportunities for studying complex targets in their native environment, increasing the probability of discovering more meaningful hits. The authors have generated and developed a mouse embryonic stem cell line called c-Photina® mES stably expressing a Ca2+-activated photoprotein as a reporter gene. This reporter cell line retains the ability to differentiate into any cell lineage and can be used for miniaturized screening processes in 384-well microplates. The c-Photina® mES cell line is particularly well suited for the study of the pharmacological modulation of target genes that induce Ca2+ mobilization. The authors differentiated this mES reporter cell line into neuronal cells and screened the LOPAC1280™ library monitoring the agonistic or antagonistic activities of compounds. They also demonstrate the possibility to generate and freeze bulk preparations of cells at an intermediate stage of differentiation and enriched in neural precursor cells, which retain the ability to form fully functional neural networks once thawed. The proposed cell model is of high value for HTS purposes because it offers a more physiological environment to the targets of interest and the possibility of using frozen batches of neural precursor cells.
Collapse
Affiliation(s)
| | - Simone Fenu
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | | | | | - Andrea Menegon
- NeuroTechnology Lab. Alembic (Advanced Light and Electron Microscopy Bio-Imaging Centre), San Raffaele Scientific Institute, Milan, Italy
| | | | | |
Collapse
|
22
|
Abstract
Ever since it was shown that maintenance of muscle contraction required the presence of extracellular Ca(2+), evidence has accumulated that Ca(2+) plays a crucial role in excitation-contraction coupling. This culminated in the use of the photoprotein aequorin to demonstrate that [Ca(2+)](i) increased after depolarization but before contraction in barnacle muscle. Green fluorescent protein was extracted from the same jellyfish as aequorin, so this work also has important historical links to the use of fluorescent proteins as markers in living cells. The subsequent development of cell-permeant Ca(2+) indicators resulted in a dramatic increase in related research, revealing Ca(2+) to be a ubiquitous cell signal. High-speed, confocal Ca(2+) imaging has now revealed subcellular detail not previously apparent, with the identification of Ca(2+) sparks. These act as building blocks for larger transients during excitation-contraction coupling in cardiac muscle, but their function in smooth muscle appears more diverse, with evidence suggesting both 'excitatory' and 'inhibitory' roles. Sparks can activate Ca(2+)-sensitive Cl() and K(+) currents, which exert positive and negative feedback, respectively, on global Ca(2+) signalling, through changes in membrane potential and activation of voltage-operated Ca(2+) channels. Calcium imaging has also demonstrated that agonists that appear to evoke relatively tonic increases in average [Ca(2+)](i) at the whole tissue level often stimulate much higher frequency phasic Ca(2+) oscillations at the cellular level. These findings may require re-evaluation of some of our models of Ca(2+) signalling to account for newly revealed cellular and subcellular detail. Future research in the field is likely to make increasing use of genetically coded Ca(2+) indicators expressed in an organelle- or tissue-specific manner.
Collapse
Affiliation(s)
- J Graham McGeown
- Centre for Vision and Vascular Sciences, Queen's University of Belfast, Institute of Clinical Sciences, Grosvenor Road, Royal Victoria Hospital, Belfast BT12 6BA, UK.
| |
Collapse
|
23
|
Discharged photoprotein obelin: fluorescence peculiarities. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2010; 101:103-8. [PMID: 20678944 DOI: 10.1016/j.jphotobiol.2010.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2010] [Revised: 06/30/2010] [Accepted: 07/01/2010] [Indexed: 11/20/2022]
Abstract
Photoprotein obelin, the enzyme-substrate complex of polypeptide with 2-hydroperoxycoelenterazine, is responsible for bioluminescence of marine hydroid Obelia longissima. Addition of Ca(2+) to the photoprotein triggers the bioluminescent reaction with light emission. The product of the bioluminescent reaction--enzyme-bound coelenteramide--is a fluorescent protein called 'discharged' obelin. It is stable and highly fluorescent. The paper considers dependence of its light-induced fluorescence on Ca(2+) concentration. Increase of Ca(2+) concentration enhanced the fluorescence intensity of discharged obelin; the dependence was found as linear in double logarithmic coordinates at Ca(2+) concentration range 10(-7)-10(-6) M, both in excitation and emission spectra. The spectra were divided into components; contributions of the components to experimental excitation and emission spectra depended on Ca(2+) concentration. The data suggest enzymatic conformational transition in discharged obelin at approximately 5 x 10(-7) M of Ca(2+) concentration. Spectra variations were attributed to acidity changes of discharged obelin chromophore (coelenteramide) in its fluorescent state S(1)*.
Collapse
|
24
|
Jones B, Holskin B, Meyer S, Ung T, Dupriez V, Flores SY, Burgeon E, Ator M, Duzic E. Aequorin functional assay for characterization of G-protein-coupled receptors: implementation with cryopreserved transiently transfected cells. Anal Biochem 2010; 400:184-9. [PMID: 20109436 DOI: 10.1016/j.ab.2010.01.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 01/06/2010] [Accepted: 01/21/2010] [Indexed: 10/19/2022]
Abstract
Assay technologies that measure intracellular Ca(2+) release are among the predominant methods for evaluation of GPCR function. These measurements have historically been performed using cell-permeable fluorescent dyes, although the use of the recombinant photoprotein aequorin (AEQ) as a Ca(2+) sensor has gained popularity with recent advances in instrumentation. The requirement of the AEQ system for cells expressing both the photoprotein and the GPCR target of interest has necessitated the labor-intensive development of cell lines stably expressing both proteins. With the goal of streamlining this process, transient transfections were used to either (1) introduce AEQ into cells stably expressing the GPCR of interest or (2) introduce the GPCR into cells stably expressing the AEQ protein, employing the human muscarinic M(1) receptor as a model system. Robust results were obtained from cryopreserved cells prepared by both strategies, yielding agonist and antagonist pharmacology in good agreement with literature values. Good reproducibility was observed between multiple transient transfection events. These results indicate that transient transfection is a viable and efficient method for production of cellular reagents for use in AEQ assays.
Collapse
Affiliation(s)
- Bruce Jones
- Cephalon, Inc., Worldwide Discovery Research, 145 Brandywine Parkway, West Chester, PA 19380, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|