1
|
Ertl F, Kopanchuk S, Dijon NC, Veikšina S, Tahk MJ, Laasfeld T, Schettler F, Gattor AO, Hübner H, Archipowa N, Köckenberger J, Heinrich MR, Gmeiner P, Kutta RJ, Holliday ND, Rinken A, Keller M. Dually Labeled Neurotensin NTS 1R Ligands for Probing Radiochemical and Fluorescence-Based Binding Assays. J Med Chem 2024; 67:16664-16691. [PMID: 39261089 PMCID: PMC11440508 DOI: 10.1021/acs.jmedchem.4c01470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
The determination of ligand-receptor binding affinities plays a key role in the development process of pharmaceuticals. While the classical radiochemical binding assay uses radioligands, fluorescence-based binding assays require fluorescent probes. Usually, radio- and fluorescence-labeled ligands are dissimilar in terms of structure and bioactivity, and can be used in either radiochemical or fluorescence-based assays. Aiming for a close comparison of both assay types, we synthesized tritiated fluorescent neurotensin receptor ligands ([3H]13, [3H]18) and their nontritiated analogues (13, 18). The labeled probes were studied in radiochemical and fluorescence-based (high-content imaging, flow cytometry, fluorescence anisotropy) binding assays. Equilibrium saturation binding yielded well-comparable ligand-receptor affinities, indicating that all these setups can be used for the screening of new drugs. In contrast, discrepancies were found in the kinetic behavior of the probes, which can be attributed to technical differences of the methods and require further studies with respect to the elucidation of the underlying mechanisms.
Collapse
Affiliation(s)
- Fabian
J. Ertl
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraβe 31, D-93053 Regensburg, Germany
| | - Sergei Kopanchuk
- Institute
of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Nicola C. Dijon
- School
of Life Sciences, University of Nottingham,
Queen’s Medical Centre, Nottingham NG7 2UH, U.K.
| | - Santa Veikšina
- Institute
of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Maris-Johanna Tahk
- Institute
of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Tõnis Laasfeld
- Institute
of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Franziska Schettler
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraβe 31, D-93053 Regensburg, Germany
| | - Albert O. Gattor
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraβe 31, D-93053 Regensburg, Germany
| | - Harald Hübner
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straβe 10, D-91058 Erlangen, Germany
| | - Nataliya Archipowa
- Institute
of Biophysics and Physical Biochemistry, Faculty of Biology and Preclinical
Medicine, University of Regensburg, Universitätsstraβe
31, D-93053 Regensburg, Germany
| | - Johannes Köckenberger
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straβe 10, D-91058 Erlangen, Germany
| | - Markus R. Heinrich
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straβe 10, D-91058 Erlangen, Germany
| | - Peter Gmeiner
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straβe 10, D-91058 Erlangen, Germany
| | - Roger J. Kutta
- Institute
of Physical and Theoretical Chemistry, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraβe 31, D-93053 Regensburg, Germany
| | - Nicholas D. Holliday
- School
of Life Sciences, University of Nottingham,
Queen’s Medical Centre, Nottingham NG7 2UH, U.K.
| | - Ago Rinken
- Institute
of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Max Keller
- Institute
of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraβe 31, D-93053 Regensburg, Germany
| |
Collapse
|
2
|
Xie J, Mao S, Zhao Y, Zhang G, Yao J, Guan Y, Yan J, Zhang H. Quantification of binding capacity of natural products to target proteins by sensors integrating SERS labeling and photocrosslinked molecular probes. Anal Chim Acta 2024; 1317:342911. [PMID: 39030011 DOI: 10.1016/j.aca.2024.342911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/03/2024] [Accepted: 06/24/2024] [Indexed: 07/21/2024]
Abstract
Natural products-based screening of active ingredients and their interactions with target proteins is an important ways to discover new drugs. Assessing the binding capacity of target proteins, particularly when multiple components are involved, presents a significant challenge for sensors. As far as we know, there is currently no sensor that can accomplish high-throughput quantitative analysis of natural product-target protein binding capacity based on Raman spectroscopy. In this study, a novel sensor model has been developed for the quantitative analysis of binding capacity based on Surface-Enhanced Raman Spectroscopy (SERS) and Photocrosslinked Molecular Probe (PCMP) technology. This sensor, named SERS-PCMP, leverages the high throughput of molecular probe technology to investigate the active ingredients in natural products, along with the application of SERS labelling technology for target proteins. Thus it significantly improves the efficiency and accuracy of target protein identification. Based on the novel strategy, quantitative analysis of the binding capacity of 20 components from Shenqi Jiangtang Granules (SJG) to α-Glucosidase were completed. Ultimately, the binding capacity of these active ingredients was ranked based on the detected Raman Intensity. The compounds with higher binding capacity were Astragaloside IV (Intensity, 138.17), Ginsenoside Rh2 (Intensity, 87.46), Ginsenoside Rg3 (Intensity, 73.92) and Ginsenoside Rh1 (Intensity, 64.37), which all exceeded the binding capacity of the positive drug Acarbose (Intensity, 28.75). Furthermore, this strategy also performed a high detection sensitivity. The limit of detection for the enzyme using 0.1 mg of molecular probe magnetic nanoparticles (MP MNPs) was determined to be no less than 0.375 μg/mL. SERS-PCMP sensor integrating SERS labeling and photocrosslinked molecular probes which offers a fresh perspective for future drug discovery studies. Such as high-throughput drug screening and the exploration of small molecule-target protein interactions in vitro.
Collapse
Affiliation(s)
- Jianhui Xie
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou, 310014, China; School of Pharmaceutical and Chemical Engineering, Taizhou University, Taizhou, 318000, China
| | - Shuying Mao
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou, 310014, China
| | - Yanglan Zhao
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou, 310014, China
| | - Guimin Zhang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Shandong, 276006, China
| | - Jingchun Yao
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Shandong, 276006, China
| | - Yongxia Guan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co., Ltd., Shandong, 276006, China
| | - Jizhong Yan
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou, 310014, China.
| | - Hui Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou, 310014, China.
| |
Collapse
|
3
|
Sakamoto K, Yamamoto Y, Inaba H, Matsuura K. Strategy toward In-Cell Self-Assembly of an Artificial Viral Capsid from a Fluorescent Protein-Modified β-Annulus Peptide. ACS Synth Biol 2024; 13:1842-1850. [PMID: 38729919 DOI: 10.1021/acssynbio.4c00135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
In-cell self-assembly of natural viral capsids is an event that can be visualized under transmission electron microscopy (TEM) observations. By mimicking the self-assembly of natural viral capsids, various artificial protein- and peptide-based nanocages were developed; however, few studies have reported the in-cell self-assembly of such nanocages. Our group developed a β-Annulus peptide that can form a nanocage called artificial viral capsid in vitro, but in-cell self-assembly of the capsid has not been achieved. Here, we designed an artificial viral capsid decorated with a fluorescent protein, StayGold, to visualize in-cell self-assembly. Fluorescence anisotropy measurements and fluorescence resonance energy transfer imaging, in addition to TEM observations of the cells and super-resolution microscopy, revealed that StayGold-conjugated β-Annulus peptides self-assembled into the StayGold-decorated artificial viral capsid in a cell. Using these techniques, we achieved the in-cell self-assembly of an artificial viral capsid.
Collapse
Affiliation(s)
- Kentarou Sakamoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
| | - Yuka Yamamoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
| | - Hiroshi Inaba
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
- Centre for Research on Green Sustainable Chemistry, Tottori University, Tottori 680-8552, Japan
| | - Kazunori Matsuura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
- Centre for Research on Green Sustainable Chemistry, Tottori University, Tottori 680-8552, Japan
| |
Collapse
|
4
|
Archipowa N, Wittmann L, Köckenberger J, Ertl FJ, Gleixner J, Keller M, Heinrich MR, Kutta RJ. Characterization of Fluorescent Dyes Frequently Used for Bioimaging: Photophysics and Photocatalytical Reactions with Proteins. J Phys Chem B 2023; 127:9532-9542. [PMID: 37903729 DOI: 10.1021/acs.jpcb.3c04484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Derivatives of the rhodamine-based dye 5-TAMRA (5-carboxy-tetramethylrhodamine) and the indocarbocyanine-type Cy3B (cyclized derivative of the cyanine dye Cy3), both representing important fluorophores frequently used for the labeling of biomolecules (proteins, nucleic acids) and bioactive compounds, such as receptor ligands, were photophysically investigated in aqueous solution, i.e., in neat phosphate-buffered saline (PBS) and in PBS supplemented with 1 wt % bovine serum albumin (BSA). The dyes exhibit comparable absorption (λabs,max: 550-569 nm) and emission wavelengths (λem,max: 580-582 nm), and similar S1 lifetimes (2.27-2.75 ns), and their excited state deactivation proceeds mainly via the lowest excited singlet state (triplet quantum yield ca. 1%). However, the probes show marked differences with respect to their fluorescence quantum yield and photostability. While 5-TAMRA shows a lower quantum yield (37-39%) than the Cy3B derivative (ca. 57%), its photostability is considerably higher compared to Cy3B. Generally, the impact of the protein on the photophysics is low. However, on prolonged illumination, both fluorescent dyes undergo a photocatalytic reaction with tryptophan residues of BSA mediated by sensitized singlet oxygen resulting in a tryptophan photoproduct with an absorption maximum around 330 nm. The overall results of this work will assist in choosing the right dye for the labeling of bioactive compounds, and the study demonstrates that experiments performed with 5-TAMRA or Cy3B-labeled compounds in a biological environment may be influenced by photochemical modification of experimentally relevant proteins at aromatic amino acid residues.
Collapse
Affiliation(s)
- Nataliya Archipowa
- Institute of Biophysics and Physical Biochemistry, Faculty of Biology and Preclinical Medicine, University of Regensburg, D-93053 Regensburg, Germany
| | - Lukas Wittmann
- Institute of Physical and Theoretical Chemistry, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Johannes Köckenberger
- Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-University Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Fabian J Ertl
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Jakob Gleixner
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Markus R Heinrich
- Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-University Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Roger Jan Kutta
- Institute of Physical and Theoretical Chemistry, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| |
Collapse
|
5
|
Scheuplein NJ, Lohr T, Vivoli Vega M, Ankrett D, Seufert F, Kirchner L, Harmer NJ, Holzgrabe U. Fluorescent probe for the identification of potent inhibitors of the macrophage infectivity potentiator (Mip) protein of Burkholderia pseudomallei. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:211-222. [PMID: 37001588 DOI: 10.1016/j.slasd.2023.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/02/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023]
Abstract
The macrophage infectivity potentiator (Mip) protein belongs to the immunophilin superfamily. This class of enzymes catalyzes the interconversion between the cis and trans configuration of proline-containing peptide bonds. Mip has been shown to be important for the virulence of a wide range of pathogenic microorganisms, including the Gram-negative bacterium Burkholderia pseudomallei. Small molecules derived from the natural product rapamycin, lacking its immunosuppression-inducing moiety, inhibit Mip's peptidyl-prolyl cis-trans isomerase (PPIase) activity and lead to a reduction in pathogen load in vitro. Here, a fluorescence polarization assay (FPA) to enable the screening and effective development of BpMip inhibitors was established. A fluorescent probe was prepared, derived from previous pipecolic scaffold Mip inhibitors labeled with fluorescein. This probe showed moderate affinity for BpMip and enabled a highly robust FPA suitable for screening large compound libraries with medium- to high-throughput (Z factor ∼ 0.89) to identify potent new inhibitors. The FPA results are consistent with data from the protease-coupled PPIase assay. Analysis of the temperature dependence of the probe's binding highlighted that BpMip's ligand binding is driven by enthalpic rather than entropic effects. This has considerable consequences for the use of low-temperature kinetic assays.
Collapse
Affiliation(s)
- Nicolas Julian Scheuplein
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Theresa Lohr
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | | | - Dyan Ankrett
- Living Systems Institute, Stocker Road, Exeter EX4 4QD, UK
| | - Florian Seufert
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Lukas Kirchner
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | | | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, Würzburg 97074, Germany.
| |
Collapse
|
6
|
Cruz P, Paredes N, Asela I, Kolimi N, Molina JA, Ramírez-Sarmiento CA, Goutam R, Huang G, Medina E, Sanabria H. Domain tethering impacts dimerization and DNA-mediated allostery in the human transcription factor FoxP1. J Chem Phys 2023; 158:2890482. [PMID: 37184020 DOI: 10.1063/5.0138782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/25/2023] [Indexed: 05/16/2023] Open
Abstract
Transcription factors are multidomain proteins with specific DNA binding and regulatory domains. In the human FoxP subfamily (FoxP1, FoxP2, FoxP3, and FoxP4) of transcription factors, a 90 residue-long disordered region links a Leucine Zipper (ZIP)-known to form coiled-coil dimers-and a Forkhead (FKH) domain-known to form domain swapping dimers. We used replica exchange discrete molecular dynamics simulations, single-molecule fluorescence experiments, and other biophysical tools to understand how domain tethering in FoxP1 impacts dimerization at ZIP and FKH domains and how DNA binding allosterically regulates their dimerization. We found that domain tethering promotes FoxP1 dimerization but inhibits a FKH domain-swapped structure. Furthermore, our findings indicate that the linker mediates the mutual organization and dynamics of ZIP and FKH domains, forming closed and open states with and without interdomain contacts, thus highlighting the role of the linkers in multidomain proteins. Finally, we found that DNA allosterically promotes structural changes that decrease the dimerization propensity of FoxP1. We postulate that, upon DNA binding, the interdomain linker plays a crucial role in the gene regulatory function of FoxP1.
Collapse
Affiliation(s)
- Perla Cruz
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Casilla 653, Santiago 7800003, Chile
| | - Nicolás Paredes
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Casilla 653, Santiago 7800003, Chile
| | - Isabel Asela
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Casilla 653, Santiago 7800003, Chile
| | - Narendar Kolimi
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, USA
| | - José Alejandro Molina
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Santiago 7820436, Chile
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Santiago 7820436, Chile
| | - César A Ramírez-Sarmiento
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Santiago 7820436, Chile
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Santiago 7820436, Chile
| | - Rajen Goutam
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, USA
| | - Gangton Huang
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, USA
| | - Exequiel Medina
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Casilla 653, Santiago 7800003, Chile
| | - Hugo Sanabria
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, USA
| |
Collapse
|
7
|
Tahk MJ, Laasfeld T, Meriste E, Brea J, Loza MI, Majellaro M, Contino M, Sotelo E, Rinken A. Fluorescence based HTS-compatible ligand binding assays for dopamine D3 receptors in baculovirus preparations and live cells. Front Mol Biosci 2023; 10:1119157. [PMID: 37006609 PMCID: PMC10062709 DOI: 10.3389/fmolb.2023.1119157] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Dopamine receptors are G-protein-coupled receptors that are connected to severe neurological disorders. The development of new ligands targeting these receptors enables gaining a deeper insight into the receptor functioning, including binding mechanisms, kinetics and oligomerization. Novel fluorescent probes allow the development of more efficient, cheaper, reliable and scalable high-throughput screening systems, which speeds up the drug development process. In this study, we used a novel Cy3B labelled commercially available fluorescent ligand CELT-419 for developing dopamine D3 receptor-ligand binding assays with fluorescence polarization and quantitative live cell epifluorescence microscopy. The fluorescence anisotropy assay using 384-well plates achieved Z’ value of 0.71, which is suitable for high-throughput screening of ligand binding. The assay can also be used to determine the kinetics of both the fluorescent ligand as well as some reference unlabeled ligands. Furthermore, CELT-419 was also used with live HEK293-D3R cells in epifluorescence microscopy imaging for deep-learning-based ligand binding quantification. This makes CELT-419 quite a universal fluorescence probe which has the potential to be also used in more advanced microscopy techniques resulting in more comparable studies.
Collapse
Affiliation(s)
| | - Tõnis Laasfeld
- Institute of Chemistry, University of Tartu, Tartu, Estonia
- Department of Computer Science, University of Tartu, Tartu, Estonia
| | - Elo Meriste
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Jose Brea
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Universidade de Santiago de Compostela, Santiago, Spain
| | - Maria Isabel Loza
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Universidade de Santiago de Compostela, Santiago, Spain
| | - Maria Majellaro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago, Spain
- Celtarys Research S.L., Santiago, Spain
| | - Marialessandra Contino
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Eddy Sotelo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago, Spain
| | - Ago Rinken
- Institute of Chemistry, University of Tartu, Tartu, Estonia
- *Correspondence: Ago Rinken,
| |
Collapse
|
8
|
Lavogina D, Nasirova N, Sõrmus T, Tähtjärv T, Enkvist E, Viht K, Haljasorg T, Herodes K, Jaal J, Uri A. Conjugates of adenosine mimetics and arginine-rich peptides serve as inhibitors and fluorescent probes but not as long-lifetime photoluminescent probes for protein arginine methyltransferases. J Pept Sci 2023; 29:e3456. [PMID: 36208424 DOI: 10.1002/psc.3456] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
Abstract
The conjugates of an adenosine mimetic and oligo-l-arginine or oligo-d-arginine (ARCs) were initially designed in our research group as inhibitors and photoluminescent probes targeting basophilic protein kinases. Here, we explored a panel of ARCs and their fluorescent derivatives in biochemical assays with members of the protein arginine methyltransferase (PRMT) family, focusing specifically on PRMT1. In the binding/displacement assay with detection of fluorescence anisotropy, we found that ARCs and arginine-rich peptides could serve as high-affinity ligands for PRMT1, whereas the equilibrium dissociation constant values depended dramatically on the number of arginine residues within the compounds. The fluorescently labeled probe ARC-1081 was displaced from its complex with PRMT1 by both S-adenosyl-l-methionine (SAM) and S-adenosyl-l-homocysteine (SAH), indicating binding of the adenosine mimetic of ARCs to the SAM/SAH-binding site within PRMT1. The ARCs that had previously shown microsecond-lifetime photoluminescence in complex with protein kinases did not feature such property in complex with PRMT1, demonstrating the selectivity of the time-resolved readout format. When tested against a panel of PRMT family members in single-dose inhibition experiments, a micromolar concentration of ARC-902 was required for the inhibition of PRMT1 and PRMT7. Overall, our results suggest that the compounds containing multiple arginine residues (including the well-known cell-penetrating peptides) are likely to inhibit PRMT and thus interfere with the epigenetic modification status in complex biological systems, which should be taken into consideration during interpretation of the experimental data.
Collapse
Affiliation(s)
- Darja Lavogina
- Institute of Chemistry, University of Tartu, Tartu, Estonia.,Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Naila Nasirova
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Tanel Sõrmus
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Taavo Tähtjärv
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Erki Enkvist
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Kaido Viht
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Tõiv Haljasorg
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Koit Herodes
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Jana Jaal
- Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.,Department of Radiotherapy and Oncological Therapy, Tartu University Hospital, Tartu, Estonia
| | - Asko Uri
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| |
Collapse
|
9
|
Zhang Y, Tang H, Chen W, Zhang J. Nanomaterials Used in Fluorescence Polarization Based Biosensors. Int J Mol Sci 2022; 23:8625. [PMID: 35955779 PMCID: PMC9369394 DOI: 10.3390/ijms23158625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Fluorescence polarization (FP) has been applied in detecting chemicals and biomolecules for early-stage diagnosis, food safety analyses, and environmental monitoring. Compared to organic dyes, inorganic nanomaterials such as quantum dots have special fluorescence properties that can enhance the photostability of FP-based biosensing. In addition, nanomaterials, such as metallic nanoparticles, can be used as signal amplifiers to increase fluorescence polarization. In this review paper, different types of nanomaterials used in in FP-based biosensors have been reviewed. The role of each type of nanomaterial, acting as a fluorescent element and/or the signal amplifier, has been discussed. In addition, the advantages of FP-based biosensing systems have been discussed and compared with other fluorescence-based techniques. The integration of nanomaterials and FP techniques allows biosensors to quickly detect analytes in a sensitive and cost-effective manner and positively impact a variety of different fields including early-stage diagnoses.
Collapse
Affiliation(s)
- Yingqi Zhang
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada; (Y.Z.); (W.C.)
| | - Howyn Tang
- School of Biomedical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada;
| | - Wei Chen
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada; (Y.Z.); (W.C.)
| | - Jin Zhang
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada; (Y.Z.); (W.C.)
- School of Biomedical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada;
| |
Collapse
|
10
|
Tahk MJ, Torp J, Ali MAS, Fishman D, Parts L, Grätz L, Müller C, Keller M, Veiksina S, Laasfeld T, Rinken A. Live-cell microscopy or fluorescence anisotropy with budded baculoviruses-which way to go with measuring ligand binding to M 4 muscarinic receptors? Open Biol 2022; 12:220019. [PMID: 35674179 PMCID: PMC9175271 DOI: 10.1098/rsob.220019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/27/2022] [Indexed: 01/04/2023] Open
Abstract
M4 muscarinic acetylcholine receptor is a G protein-coupled receptor (GPCR) that has been associated with alcohol and cocaine abuse, Alzheimer's disease, and schizophrenia which makes it an interesting drug target. For many GPCRs, the high-affinity fluorescence ligands have expanded the options for high-throughput screening of drug candidates and serve as useful tools in fundamental receptor research. Here, we explored two TAMRA-labelled fluorescence ligands, UR-MK342 and UR-CG072, for development of assays for studying ligand-binding properties to M4 receptor. Using budded baculovirus particles as M4 receptor preparation and fluorescence anisotropy method, we measured the affinities and binding kinetics of both fluorescence ligands. Using the fluorescence ligands as reporter probes, the binding affinities of unlabelled ligands could be determined. Based on these results, we took a step towards a more natural system and developed a method using live CHO-K1-hM4R cells and automated fluorescence microscopy suitable for the routine determination of unlabelled ligand affinities. For quantitative image analysis, we developed random forest and deep learning-based pipelines for cell segmentation. The pipelines were integrated into the user-friendly open-source Aparecium software. Both image analysis methods were suitable for measuring fluorescence ligand saturation binding and kinetics as well as for screening binding affinities of unlabelled ligands.
Collapse
Affiliation(s)
- Maris-Johanna Tahk
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Jane Torp
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Mohammed A. S. Ali
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
| | - Dmytro Fishman
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
| | - Leopold Parts
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, UK
| | - Lukas Grätz
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Christoph Müller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Santa Veiksina
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Tõnis Laasfeld
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
- Department of Computer Science, University of Tartu, Narva Street 20, 51009 Tartu, Estonia
| | - Ago Rinken
- Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| |
Collapse
|
11
|
Müller C, Gleixner J, Tahk MJ, Kopanchuk S, Laasfeld T, Weinhart M, Schollmeyer D, Betschart MU, Lüdeke S, Koch P, Rinken A, Keller M. Structure-Based Design of High-Affinity Fluorescent Probes for the Neuropeptide Y Y 1 Receptor. J Med Chem 2022; 65:4832-4853. [PMID: 35263541 DOI: 10.1021/acs.jmedchem.1c02033] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The recent crystallization of the neuropeptide Y Y1 receptor (Y1R) in complex with the argininamide-type Y1R selective antagonist UR-MK299 (2) opened up a new approach toward structure-based design of nonpeptidic Y1R ligands. We designed novel fluorescent probes showing excellent Y1R selectivity and, in contrast to previously described fluorescent Y1R ligands, considerably higher (∼100-fold) binding affinity. This was achieved through the attachment of different fluorescent dyes to the diphenylacetyl moiety in 2 via an amine-functionalized linker. The fluorescent ligands exhibited picomolar Y1R binding affinities (pKi values of 9.36-9.95) and proved to be Y1R antagonists, as validated in a Fura-2 calcium assay. The versatile applicability of the probes as tool compounds was demonstrated by flow cytometry- and fluorescence anisotropy-based Y1R binding studies (saturation and competition binding and association and dissociation kinetics) as well as by widefield and total internal reflection fluorescence (TIRF) microscopy of live tumor cells, revealing that fluorescence was mainly localized at the plasma membrane.
Collapse
Affiliation(s)
- Christoph Müller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Jakob Gleixner
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Maris-Johanna Tahk
- Institute of Chemistry, Faculty of Bioorganic Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Sergei Kopanchuk
- Institute of Chemistry, Faculty of Bioorganic Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Tõnis Laasfeld
- Institute of Chemistry, Faculty of Bioorganic Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Michael Weinhart
- Institute of Inorganic Chemistry, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Dieter Schollmeyer
- Department of Chemistry, Johannes-Gutenberg-University Mainz, Düsbergweg 10-14, 55099 Mainz, Germany
| | - Martin U Betschart
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstrasse 25, 79104 Freiburg, Germany
| | - Steffen Lüdeke
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstrasse 25, 79104 Freiburg, Germany
| | - Pierre Koch
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Ago Rinken
- Institute of Chemistry, Faculty of Bioorganic Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| |
Collapse
|
12
|
Lingasamy P, Põšnograjeva K, Kopanchuk S, Tobi A, Rinken A, General IJ, Asciutto EK, Teesalu T. PL1 Peptide Engages Acidic Surfaces on Tumor-Associated Fibronectin and Tenascin Isoforms to Trigger Cellular Uptake. Pharmaceutics 2021; 13:pharmaceutics13121998. [PMID: 34959279 PMCID: PMC8707168 DOI: 10.3390/pharmaceutics13121998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/09/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
Tumor extracellular matrix (ECM) is a high-capacity target for the precision delivery of affinity ligand-guided drugs and imaging agents. Recently, we developed a PL1 peptide (sequence: PPRRGLIKLKTS) for systemic targeting of malignant ECM. Here, we map the dynamics of PL1 binding to its receptors Fibronectin Extra Domain B (FN-EDB) and Tenascin C C-isoform (TNC-C) by computational modeling and cell-free binding studies on mutated receptor proteins, and study cellular binding and internalization of PL1 nanoparticles in cultured cells. Molecular dynamics simulation and docking analysis suggested that the engagement of PL1 peptide with both receptors is primarily driven by electrostatic interactions. Substituting acidic amino acid residues with neutral amino acids at predicted PL1 binding sites in FN-EDB (D52N-D49N-D12N) and TNC-C (D39N-D45N) resulted in the loss of binding of PL1 nanoparticles. Remarkably, PL1-functionalized nanoparticles (NPs) were not only deposited on the target ECM but bound the cells and initiated a robust cellular uptake via a pathway resembling macropinocytosis. Our studies establish the mode of engagement of the PL1 peptide with its receptors and suggest applications for intracellular delivery of nanoscale payloads. The outcomes of this work can be used for the development of PL1-derived peptides with improved stability, affinity, and specificity for precision targeting of the tumor ECM and malignant cells.
Collapse
Affiliation(s)
- Prakash Lingasamy
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (P.L.); (K.P.); (A.T.)
| | - Kristina Põšnograjeva
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (P.L.); (K.P.); (A.T.)
| | - Sergei Kopanchuk
- Institute of Chemistry, University of Tartu, Ravila 14, 50411 Tartu, Estonia; (S.K.); (A.R.)
| | - Allan Tobi
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (P.L.); (K.P.); (A.T.)
| | - Ago Rinken
- Institute of Chemistry, University of Tartu, Ravila 14, 50411 Tartu, Estonia; (S.K.); (A.R.)
| | - Ignacio J. General
- Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA;
| | - Eliana K. Asciutto
- School of Science and Technology, National University of San Martin (UNSAM), ICIFI and CONICET, 25 de Mayo y Francia, San Martín 1650, Argentina;
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (P.L.); (K.P.); (A.T.)
- Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA;
- Correspondence: Estonia
| |
Collapse
|
13
|
Koretz KS, McGraw CE, Stradley S, Elbaradei A, Malmstadt N, Robinson AS. Characterization of binding kinetics of A 2AR to Gα s protein by surface plasmon resonance. Biophys J 2021; 120:1641-1649. [PMID: 33675761 DOI: 10.1016/j.bpj.2021.02.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/11/2021] [Accepted: 02/17/2021] [Indexed: 01/09/2023] Open
Abstract
Because of their surface localization, G protein-coupled receptors (GPCRs) are often pharmaceutical targets as they respond to a variety of extracellular stimuli (e.g., light, hormones, small molecules) that may activate or inhibit a downstream signaling response. The adenosine A2A receptor (A2AR) is a well-characterized GPCR that is expressed widely throughout the human body, with over 10 crystal structures determined. Truncation of the A2AR C-terminus is necessary for crystallization as this portion of the receptor is long and unstructured; however, previous work suggests shortening of the A2AR C-terminus from 412 to 316 amino acids (A2AΔ316R) ablates downstream signaling, as measured by cAMP production, to below that of constitutive full-length A2AR levels. As cAMP production is downstream of the first activation event-coupling of G protein to its receptor-investigating that first step in activation is important in understanding how the truncation effects native GPCR function. Here, using purified receptor and Gαs proteins, we characterize the association of A2AR and A2AΔ316R to Gαs with and without GDP or GTPγs using surface plasmon resonance (SPR). Gαs affinity for A2AR was greatest for apo-Gαs, moderately affected in the presence of GDP and nearly completely ablated by the addition of GTPγs. Truncation of the A2AR C-terminus (A2AΔ316R) decreased the affinity of the unliganded receptor for Gαs by ∼20%, suggesting small changes to binding can greatly impact downstream signaling.
Collapse
Affiliation(s)
- Kirsten S Koretz
- Department of Chemical and Biomolecular Engineering, Tulane University, New Orleans, Louisiana; Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Claire E McGraw
- Department of Chemical and Biomolecular Engineering, Tulane University, New Orleans, Louisiana
| | - Steven Stradley
- Department of Chemical and Biomolecular Engineering, Tulane University, New Orleans, Louisiana
| | - Ahmed Elbaradei
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California
| | - Noah Malmstadt
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California
| | - Anne S Robinson
- Department of Chemical and Biomolecular Engineering, Tulane University, New Orleans, Louisiana; Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania.
| |
Collapse
|
14
|
Laasfeld T, Ehrminger R, Tahk MJ, Veiksina S, Kõlvart KR, Min M, Kopanchuk S, Rinken A. Budded baculoviruses as a receptor display system to quantify ligand binding with TIRF microscopy. NANOSCALE 2021; 13:2436-2447. [PMID: 33464268 DOI: 10.1039/d0nr06737g] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Studying mechanisms of receptor-ligand interactions has remained challenging due to several limitations of different measurement methods. Here we present a total internal reflection fluorescence microscopy-based method that maintains the right balance between retaining the receptors in the natural lipid environment, sufficient throughput for ligand screening, high sensitivity, and offering more detailed view into the ligand-binding process. The novel method combines G protein-coupled receptor display in budded baculovirus particles and the immobilization of the particles to a functionalized coverslip. We adapted and validated the functionalized coverslip preparation process to achieve selective immobilization of budded baculovirus particles. The selectivity of budded baculovirus immobilization was validated with budded baculovirus particles displaying either Frizzled 6 receptors labeled with mCherry or neuropeptide Y Y1 receptors. To scale the system for ligand binding assays, we developed both open-source multiwell systems and image analysis software SPOTNIC for flexible assay design. The neuropeptide Y Y1 receptor was used for further receptor-ligand binding studies with high-affinity TAMRA labeled fluorescent ligand UR-MC026. The affinities of the fluorescent ligand and four unlabeled ligands (BIBO3304, UR-MK299, PYY, pNPY) were obtained with the developed method and followed a similar trend with both the parallel measurements with fluorescence anisotropy method and the data published earlier. The novel method could be extended for various advanced assays utilizing multidimensional detection modes, integrating super-resolution methods for single molecule detection and microfluidic devices for kinetic measurements.
Collapse
Affiliation(s)
- Tõnis Laasfeld
- University of Tartu, Institute of Chemistry, Ravila 14a, 50411, Tartu, Estonia.
| | - Robin Ehrminger
- University of Tartu, Institute of Chemistry, Ravila 14a, 50411, Tartu, Estonia. and Tallinn University of Technology, Thomas Johann Seebeck Department of Electronics, Ehitajate Tee 5, 19086, Tallinn, Estonia
| | - Maris-Johanna Tahk
- University of Tartu, Institute of Chemistry, Ravila 14a, 50411, Tartu, Estonia.
| | - Santa Veiksina
- University of Tartu, Institute of Chemistry, Ravila 14a, 50411, Tartu, Estonia.
| | - Karl Rene Kõlvart
- University of Tartu, Institute of Chemistry, Ravila 14a, 50411, Tartu, Estonia.
| | - Mart Min
- Tallinn University of Technology, Thomas Johann Seebeck Department of Electronics, Ehitajate Tee 5, 19086, Tallinn, Estonia
| | - Sergei Kopanchuk
- University of Tartu, Institute of Chemistry, Ravila 14a, 50411, Tartu, Estonia.
| | - Ago Rinken
- University of Tartu, Institute of Chemistry, Ravila 14a, 50411, Tartu, Estonia.
| |
Collapse
|
15
|
Veiksina S, Tahk MJ, Laasfeld T, Link R, Kopanchuk S, Rinken A. Fluorescence Anisotropy-Based Assay for Characterization of Ligand Binding Dynamics to GPCRs: The Case of Cy3B-Labeled Ligands Binding to MC 4 Receptors in Budded Baculoviruses. Methods Mol Biol 2021; 2268:119-136. [PMID: 34085265 DOI: 10.1007/978-1-0716-1221-7_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
During the past decade, fluorescence methods have become valuable tools for characterizing ligand binding to G protein-coupled receptors (GPCRs). However, only a few of the assays enable studying wild-type receptors and monitor the ligand binding in real time. One of the approaches that is inherently suitable for this purpose is the fluorescence anisotropy (FA) assay. In the FA assay, the change of ligand's rotational freedom connected with its binding to the receptor can be monitored with a conventional fluorescence plate reader equipped with suitable optical filters. To achieve the high receptor concentration required for the assay and the low autofluorescence levels essential for reliable results, budded baculoviruses that display GPCRs on their surfaces can be used. The monitoring process generates a substantial amount of kinetic data, which is usually stored as a proprietary file format limiting the flexibility of data analysis. To solve this problem, we propose the use of the data curation software Aparecium ( http://gpcr.ut.ee/aparecium.html ), which integrates experimental data with metadata in a Minimum Information for Data Analysis in Systems Biology (MIDAS) format. Aparecium enables data export to different software packages for fitting to suitable kinetic or equilibrium models. A combination of the FA assay with the novel data analysis strategy is suitable for screening new active compounds, but also for modeling complex systems of ligand binding to GPCRs. We present the proposed approach using different fluorescent probes and assay types to characterize ligand binding to melanocortin 4 (MC4) receptor.
Collapse
Affiliation(s)
- Santa Veiksina
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | | | - Tõnis Laasfeld
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Reet Link
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | | | - Ago Rinken
- Institute of Chemistry, University of Tartu, Tartu, Estonia.
| |
Collapse
|
16
|
BRET- and fluorescence anisotropy-based assays for real-time monitoring of ligand binding to M 2 muscarinic acetylcholine receptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118930. [PMID: 33347921 DOI: 10.1016/j.bbamcr.2020.118930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/26/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022]
Abstract
BRET and fluorescence anisotropy (FA) are two fluorescence-based techniques used for the characterization of ligand binding to G protein-coupled receptors (GPCRs) and both allow monitoring of ligand binding in real time. In this study, we present the first direct comparison of BRET-based and FA-based binding assays using the human M2 muscarinic acetylcholine receptor (M2R) and two TAMRA (5-carboxytetramethylrhodamine)-labeled fluorescent ligands as a model system. The determined fluorescent ligand affinities from both assays were in good agreement with results obtained from radioligand competition binding experiments. The assays yielded real-time kinetic binding data revealing differences in the mechanism of binding for the investigated fluorescent probes. Furthermore, the investigation of various unlabeled M2R ligands yielded pharmacological profiles in accordance with earlier reported data. Taken together, this study showed that BRET- and FA-based binding assays represent valuable alternatives to radioactivity-based methods for screening purposes and for a precise characterization of binding kinetics supporting the exploration of binding mechanisms.
Collapse
|
17
|
Allikalt A, Purkayastha N, Flad K, Schmidt MF, Tabor A, Gmeiner P, Hübner H, Weikert D. Fluorescent ligands for dopamine D 2/D 3 receptors. Sci Rep 2020; 10:21842. [PMID: 33318558 PMCID: PMC7736868 DOI: 10.1038/s41598-020-78827-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022] Open
Abstract
Fluorescent ligands are versatile tools for the study of G protein-coupled receptors. Depending on the fluorophore, they can be used for a range of different applications, including fluorescence microscopy and bioluminescence or fluorescence resonance energy transfer (BRET or FRET) assays. Starting from phenylpiperazines and indanylamines, privileged scaffolds for dopamine D2-like receptors, we developed dansyl-labeled fluorescent ligands that are well accommodated in the binding pockets of D2 and D3 receptors. These receptors are the target proteins for the therapy for several neurologic and psychiatric disorders, including Parkinson’s disease and schizophrenia. The dansyl-labeled ligands exhibit binding affinities up to 0.44 nM and 0.29 nM at D2R and D3R, respectively. When the dansyl label was exchanged for sterically more demanding xanthene or cyanine dyes, fluorescent ligands 10a-c retained excellent binding properties and, as expected from their indanylamine pharmacophore, acted as agonists at D2R. While the Cy3B-labeled ligand 10b was used to visualize D2R and D3R on the surface of living cells by total internal reflection microscopy, ligand 10a comprising a rhodamine label showed excellent properties in a NanoBRET binding assay at D3R.
Collapse
Affiliation(s)
- Anni Allikalt
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Nirupam Purkayastha
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Khajidmaa Flad
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Maximilian F Schmidt
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Alina Tabor
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Dorothee Weikert
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany.
| |
Collapse
|
18
|
Fan YL, Liu ZY, Zeng YM, Huang LY, Li Z, Zhang ZL, Pang DW, Tian ZQ. A near-infrared-II fluorescence anisotropy strategy for separation-free detection of adenosine triphosphate in complex media. Talanta 2020; 223:121721. [PMID: 33303167 DOI: 10.1016/j.talanta.2020.121721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/22/2020] [Accepted: 09/28/2020] [Indexed: 10/23/2022]
Abstract
Fluorescence anisotropy (FA) has been widely applied for detecting and monitoring special targets in life sciences. However, matrix autofluorescence restricted its further application in complex biological samples. Herein, we report a near-infrared-II (NIR-II) FA strategy for detecting adenosine triphosphate (ATP) in human serum samples and breast cancer cell lysate, which employed NIR-II fluorescent Ag2Se quantum dots (QDs) as tags to reduce matrix autofluorescence effect and applied graphene oxide (GO) to enhance fluorescence anisotropy signals. In the presence of ATP, the recognition between NIR-II Ag2Se QDs labeled aptamer (QD-pDNA) and ATP led to the release of QD-pDNA from GO, resulting in the obvious decrease of FA values. ATP could be quantitatively detected in concentrations ranged from 3 nM to 2500 nM, with a detection limit down to 1.01 nM. This study showed that the developed NIR-II FA strategy could be applied for detecting targets in complex biological samples and had great potential for monitoring interactions between biomolecules in biomedical research.
Collapse
Affiliation(s)
- Ya-Ling Fan
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Zhen-Ya Liu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Yu-Mei Zeng
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Lu-Yao Huang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Zheng Li
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Zhi-Ling Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Dai-Wen Pang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Zhi-Quan Tian
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, PR China; College of Science, Tibet University, Lhasa, 850000, PR China.
| |
Collapse
|
19
|
Allikalt A, Laasfeld T, Ilisson M, Kopanchuk S, Rinken A. Quantitative analysis of fluorescent ligand binding to dopamine D 3 receptors using live-cell microscopy. FEBS J 2020; 288:1514-1532. [PMID: 32783364 DOI: 10.1111/febs.15519] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/14/2020] [Accepted: 07/30/2020] [Indexed: 12/29/2022]
Abstract
Dopamine receptors are G protein-coupled receptors that have several essential functions in the central nervous system. A better understanding of the regulatory mechanisms of ligand binding to the receptor may open new possibilities to affect the downstream signal transduction pathways. The majority of the available ligand binding assays use either membrane preparations, cell suspensions, or genetically modified receptors, which may give at least partially incorrect understanding of ligand binding. In this study, we implemented an assay combining fluorescence and bright-field microscopy to measure ligand binding to dopamine D3 receptors in live mammalian cells. For membrane fluorescence intensity quantification from microscopy images, we developed a machine learning-based user-friendly software membrane tools and incorporated it into a data management software aparecium that has been previously developed in our workgroup. For the experiments, a fluorescent ligand NAPS-Cy3B was synthesized by conjugating a dopaminergic antagonist N-(p-aminophenethyl)spiperone with a fluorophore Cy3B. The subnanomolar affinity of NAPS-Cy3B makes it a suitable ligand for the characterization of D3 receptors in live HEK293 cells. Using a microplate compatible automated widefield fluorescence microscope, together with the membrane tools software, enables the detection and quantification of ligand binding with a high-throughput. The live cell assay is suitable for the characterization of fluorescent ligand binding and also in the competition experiments for the screening of novel unlabeled dopaminergic ligands. We propose that this simple yet more native-like approach is feasible in GPCR research, as it enables the detection of ligand binding in an environment containing more components involved in the signal transduction cascade.
Collapse
Affiliation(s)
- Anni Allikalt
- Institute of Chemistry, University of Tartu, Estonia.,Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | | | | | | | - Ago Rinken
- Institute of Chemistry, University of Tartu, Estonia
| |
Collapse
|
20
|
Biselli S, Alencastre I, Tropmann K, Erdmann D, Chen M, Littmann T, Maia AF, Gomez-Lazaro M, Tanaka M, Ozawa T, Keller M, Lamghari M, Buschauer A, Bernhardt G. Fluorescent H 2 Receptor Squaramide-Type Antagonists: Synthesis, Characterization, and Applications. ACS Med Chem Lett 2020; 11:1521-1528. [PMID: 32832018 DOI: 10.1021/acsmedchemlett.0c00033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/20/2020] [Indexed: 02/08/2023] Open
Abstract
Fluorescence labeled ligands have been gaining importance as molecular tools, enabling receptor-ligand-binding studies by various fluorescence-based techniques. Aiming at red-emitting fluorescent ligands for the hH2R, a series of squaramides labeled with pyridinium or cyanine fluorophores (19-27) was synthesized and characterized. The highest hH2R affinities in radioligand competition binding assays were obtained in the case of pyridinium labeled antagonists 19-21 (pK i: 7.71-7.76) and cyanine labeled antagonists 23 and 25 (pK i: 7.67, 7.11). These fluorescent ligands proved to be useful tools for binding studies (saturation and competition binding as well as kinetic experiments), using confocal microscopy, flow cytometry, and high content imaging. Saturation binding experiments revealed pK d values comparable to the pK i values. The fluorescent probes 21, 23, and 25 could be used to localize H2 receptors in HEK cells and to determine the binding affinities of unlabeled compounds.
Collapse
Affiliation(s)
- Sabrina Biselli
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Inês Alencastre
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135 Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Katharina Tropmann
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Daniela Erdmann
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Mengya Chen
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Timo Littmann
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - André F. Maia
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135 Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria Gomez-Lazaro
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135 Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Miho Tanaka
- Department of Chemistry, School of Science, University of Tokyo, 7-3-1 Bunkyo-ku, Hongo, Tokyo 113-0033, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, University of Tokyo, 7-3-1 Bunkyo-ku, Hongo, Tokyo 113-0033, Japan
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Meriem Lamghari
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135 Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Armin Buschauer
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Günther Bernhardt
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| |
Collapse
|
21
|
Gruber CG, Pegoli A, Müller C, Grätz L, She X, Keller M. Differently fluorescence-labelled dibenzodiazepinone-type muscarinic acetylcholine receptor ligands with high M 2R affinity. RSC Med Chem 2020; 11:823-832. [PMID: 33479678 PMCID: PMC7650007 DOI: 10.1039/d0md00137f] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/20/2020] [Indexed: 11/21/2022] Open
Abstract
A series of fluorescent dibenzodiazepinone-type muscarinic acetylcholine M2 receptor (M2R) ligands was synthesized using various fluorescent dyes (5-TAMRA, λ ex/λ em ≈ 547/576 nm; BODIPY 630/650, λ ex/λ em ≈ 625/640 nm; pyridinium dye Py-1, λ ex/λ em ≈ 611/665 nm and pyridinium dye Py-5, λ ex/λ em ≈ 465/732 nm). All fluorescent probes exhibited high M2R affinity (pK i (radioligand competition binding): 8.75-9.62, pK d (flow cytometry): 8.36-9.19), a very low preference for the M2R over the M1 and M4 receptors and moderate to pronounced M2R selectivity compared to the M3 and M5 receptors. The presented fluorescent ligands are considered useful molecular tools for future studies using methods such as fluorescence anisotropy and BRET based MR binding assays.
Collapse
Affiliation(s)
- Corinna G Gruber
- Institute of Pharmacy , Faculty of Chemistry and Pharmacy , University of Regensburg , Universitätsstrasse 31 , D-93053 Regensburg , Germany .
| | - Andrea Pegoli
- Institute of Pharmacy , Faculty of Chemistry and Pharmacy , University of Regensburg , Universitätsstrasse 31 , D-93053 Regensburg , Germany .
| | - Christoph Müller
- Institute of Pharmacy , Faculty of Chemistry and Pharmacy , University of Regensburg , Universitätsstrasse 31 , D-93053 Regensburg , Germany .
| | - Lukas Grätz
- Institute of Pharmacy , Faculty of Chemistry and Pharmacy , University of Regensburg , Universitätsstrasse 31 , D-93053 Regensburg , Germany .
| | - Xueke She
- Institute of Pharmacy , Faculty of Chemistry and Pharmacy , University of Regensburg , Universitätsstrasse 31 , D-93053 Regensburg , Germany .
| | - Max Keller
- Institute of Pharmacy , Faculty of Chemistry and Pharmacy , University of Regensburg , Universitätsstrasse 31 , D-93053 Regensburg , Germany .
| |
Collapse
|
22
|
She X, Pegoli A, Gruber CG, Wifling D, Carpenter J, Hübner H, Chen M, Wan J, Bernhardt G, Gmeiner P, Holliday ND, Keller M. Red-Emitting Dibenzodiazepinone Derivatives as Fluorescent Dualsteric Probes for the Muscarinic Acetylcholine M2 Receptor. J Med Chem 2020; 63:4133-4154. [PMID: 32233403 DOI: 10.1021/acs.jmedchem.9b02172] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Xueke She
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Andrea Pegoli
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Corinna G. Gruber
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - David Wifling
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Jessica Carpenter
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Derby Road, Nottingham NG7 2UH, U.K
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Mengya Chen
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Jianfei Wan
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Günther Bernhardt
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Nicholas D. Holliday
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Derby Road, Nottingham NG7 2UH, U.K
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstr. 31, D-93053 Regensburg, Germany
| |
Collapse
|
23
|
Keller M, Mahuroof SA, Hong Yee V, Carpenter J, Schindler L, Littmann T, Pegoli A, Hübner H, Bernhardt G, Gmeiner P, Holliday ND. Fluorescence Labeling of Neurotensin(8-13) via Arginine Residues Gives Molecular Tools with High Receptor Affinity. ACS Med Chem Lett 2020; 11:16-22. [PMID: 31938457 PMCID: PMC6956362 DOI: 10.1021/acsmedchemlett.9b00462] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 11/19/2019] [Indexed: 02/07/2023] Open
Abstract
Fluorescence-labeled receptor ligands have emerged as valuable molecular tools, being indispensable for studying receptor-ligand interactions by fluorescence-based techniques such as high-content imaging, fluorescence microscopy, and fluorescence polarization. Through application of a new labeling strategy for peptides, a series of fluorescent neurotensin(8-13) derivatives was synthesized by attaching red-emitting fluorophores (indolinium- and pyridinium-type cyanine dyes) to carbamoylated arginine residues in neurotensin(8-13) analogues, yielding fluorescent probes with high NTS1R affinity (pK i values: 8.15-9.12) and potency (pEC50 values (Ca2+ mobilization): 8.23-9.43). Selected fluorescent ligands were investigated by flow cytometry and high-content imaging (saturation binding, kinetic studies, and competition binding) as well as by confocal microscopy using intact CHO-hNTS1R cells. The study demonstrates the applicability of the fluorescent probes as molecular tools to obtain, for example, information about the localization of receptors in cells and to determine binding affinities of nonlabeled ligands.
Collapse
Affiliation(s)
- Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Shahani A. Mahuroof
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Vivyanne Hong Yee
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Jessica Carpenter
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Lisa Schindler
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Timo Littmann
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Andrea Pegoli
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Günther Bernhardt
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University, Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Nicholas D. Holliday
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, United Kingdom
| |
Collapse
|
24
|
Link R, Veiksina S, Tahk MJ, Laasfeld T, Paiste P, Kopanchuk S, Rinken A. The constitutive activity of melanocortin-4 receptors in cAMP pathway is allosterically modulated by zinc and copper ions. J Neurochem 2019; 153:346-361. [PMID: 31792980 DOI: 10.1111/jnc.14933] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 11/06/2019] [Accepted: 11/29/2019] [Indexed: 01/25/2023]
Abstract
Melanocortin-4 receptors (MC4 R) are unique among G-protein-coupled receptors (GPCRs) as they have endogenous ligands that can exhibit inverse agonistic properties in the case of elevated basal activity. It is known that the constitutive activity of GPCRs strongly affects the ligand-dependent physiological responses, but little is known about these regulatory mechanisms. Since several metal ions have been shown to be important modulators of the signal transduction of GPCRs, we hypothesized that metal ions regulate the basal activity of MC4 Rs. Implementation of a fluorescence anisotropy assay and novel redshifted fluorescent peptides enabled kinetic characterization of ligand binding to MC4 R expressed on budded baculoviruses. We show that Ca2+ is required for high-affinity ligand binding, but Zn2+ and Cu2+ in the presence of Ca2+ behave as negative allosteric modulators of ligand binding to MC4 R. FRET-based cAMP biosensor was used to measure the activation of MC4 R stably expressed in CHO-K1 cells. At low micromolar concentrations, Zn2+ caused MC4 R-dependent activation of the cAMP pathway, whereas Cu2+ reduced the activity of MC4 R even below the basal level. These findings indicate that at physiologically relevant concentrations can Zn2+ and Cu2+ function as MC4 R agonists or inverse agonists, respectively. This means that depending on the level of constitutive activity induced by Zn2+ ions, the pharmacological effect of orthosteric ligands of MC4 R can be switched from a partial to an inverse agonist. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. More information about the Open Science badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Reet Link
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Santa Veiksina
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | | | - Tõnis Laasfeld
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Päärn Paiste
- Department of Geology, University of Tartu, Tartu, Estonia
| | | | - Ago Rinken
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| |
Collapse
|
25
|
Calmet P, Cullin C, Cortès S, Vang M, Caudy N, Baccouch R, Dessolin J, Maamar NT, Lecomte S, Tillier B, Alves ID. Cholesterol impacts chemokine CCR5 receptor ligand-binding activity. FEBS J 2019; 287:2367-2385. [PMID: 31738467 DOI: 10.1111/febs.15145] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/27/2019] [Accepted: 11/16/2019] [Indexed: 01/03/2023]
Abstract
The chemokine CCR5 receptor is target of maraviroc, a negative allosteric modulator of CCR5 that blocks the HIV protein gp120 from associating with the receptor, thereby inhibiting virus cellular entry. As noted with other G-protein-coupled receptor family members, the role of the lipid environment in CCR5 signaling remains obscure and very modestly investigated. Controversial literature on the impact of cholesterol (Chol) depletion in HIV infection and CCR5 signaling, including the hypothesis that Chol depletion could inhibit HIV infection, lead us to focus on the understanding of Chol impact in the first stages of receptor activation. To address this aim, the approach chosen was to employ reconstituted model lipid systems of controlled lipid composition containing CCR5 from two distinct expression systems: Pichia pastoris and cell-free expression. The characterization of receptor/ligand interaction in terms of total binding or competition binding assays was independently performed by plasmon waveguide resonance and fluorescence anisotropy, respectively. Maraviroc, a potent receptor antagonist, was the ligand investigated. Additionally, coarse-grained molecular dynamics simulation was employed to investigate Chol impact in the receptor-conformational flexibility and dynamics. Results obtained with receptor produced by different expression systems and using different biophysical approaches clearly demonstrate a considerable impact of Chol in the binding affinity of maraviroc to the receptor and receptor-conformational dynamics. Chol considerably decreases maraviroc binding affinity to the CCR5 receptor. The mechanisms by which this effect occurs seem to involve the adoption of distinct receptor-conformational states with restrained structural dynamics and helical motions in the presence of Chol.
Collapse
Affiliation(s)
- Pierre Calmet
- CBMN, UMR 5248 CNRS, University of Bordeaux, Pessac, France
| | | | | | - Maylou Vang
- CBMN, UMR 5248 CNRS, University of Bordeaux, Pessac, France
| | - Nada Caudy
- CBMN, UMR 5248 CNRS, University of Bordeaux, Pessac, France
| | - Rim Baccouch
- CBMN, UMR 5248 CNRS, University of Bordeaux, Pessac, France
| | - Jean Dessolin
- CBMN, UMR 5248 CNRS, University of Bordeaux, Pessac, France
| | | | - Sophie Lecomte
- CBMN, UMR 5248 CNRS, University of Bordeaux, Pessac, France
| | | | - Isabel D Alves
- CBMN, UMR 5248 CNRS, University of Bordeaux, Pessac, France
| |
Collapse
|
26
|
Uri A, Nonga OE. What is the current value of fluorescence polarization assays in small molecule screening? Expert Opin Drug Discov 2019; 15:131-133. [DOI: 10.1080/17460441.2020.1702966] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Asko Uri
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | | |
Collapse
|
27
|
Lopez A, Liu J. Fluorescence Polarization for Probing DNA Adsorption by Nanomaterials and Fluorophore/DNA Interactions. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:9954-9961. [PMID: 31271290 DOI: 10.1021/acs.langmuir.9b01678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Fluorescence polarization (FP) is attractive for measuring binding interactions and has been recently used to study DNA adsorption on nanomaterials. Since most nanomaterials are strong fluorescence quenchers, correlations among adsorption efficiency, quenching efficiency, and FP need to be interpreted carefully. In this work, carboxyfluorescein (FAM)-labeled DNA oligonucleotides were studied under various quenching conditions. First, quenching was induced by lowering the pH, taking advantage of the fact that FAM is almost nonfluorescent at a pH below 4. Strong interactions were observed between the FAM label and polyadenine DNA, as judged by the increased FP at low pH, while FAM-labeled polythymine DNA was less affected by the pH. Comparisons were also performed with FAM-labeled poly(ethylene glycol) and bovine serum albumin. An equation was derived to calculate the effect of fluorescence quenching and DNA adsorption by nanomaterials. For strongly quenching nanomaterials, such as graphene oxide, DNA adsorption alone does not change the measured FP. Light scattering and weak fluorescence from graphene oxide increase FP in these cases. For comparison, a strongly adsorbing but weak quenching material, Y2O3, was also studied and the result was consistent with a normal binding reaction. Overall, FP is a powerful technique for binding and adsorption assays, but quenched samples need to be interpreted with care.
Collapse
Affiliation(s)
- Anand Lopez
- Department of Chemistry, Waterloo Institute for Nanotechnology , University of Waterloo , Waterloo , Ontario N2L 3G1 , Canada
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology , University of Waterloo , Waterloo , Ontario N2L 3G1 , Canada
| |
Collapse
|
28
|
Viirlaid E, Ilisson M, Kopanchuk S, Mäeorg U, Rinken A, Rinken T. Immunoassay for rapid on-site detection of glyphosate herbicide. ENVIRONMENTAL MONITORING AND ASSESSMENT 2019; 191:507. [PMID: 31342281 DOI: 10.1007/s10661-019-7657-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 07/15/2019] [Indexed: 06/10/2023]
Abstract
Glyphosate is the most widespread herbicide and its global use is steadily increasing. Although glyphosate is considered to have low toxicity, its wide application has raised concerns about its effects on human health. The extensive use of glyphosate has risen a need of its continuous monitoring in drinking and surface waters to assure in accordance with the set standards. Within the present study, we have developed a novel assay for the on-site detection of glyphosate by combining flow-through technology with the high specificity of immunorecognition. The proposed biosensing system was based on the detection of fluorescence signal generated by the quantitative replacement of glyphosate in antigen-antibody complex with IgY-type anti-glyphosate antibodies on microbeads by synthetic 5-carboxytetramethylrhodamine (5-TAMRA) conjugated glyphosate. The working range of this assay was in low millimolar range and the time required for glyphosate detection around 0.5 h. The applicability of the immunoassay for glyphosate detection in surface water was tested and the biosensor results were validated with high-performance liquid chromatography.
Collapse
Affiliation(s)
- E Viirlaid
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia.
| | - M Ilisson
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| | - S Kopanchuk
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| | - U Mäeorg
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| | - A Rinken
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| | - T Rinken
- Institute of Chemistry, University of Tartu, Ravila 14A, 50411, Tartu, Estonia
| |
Collapse
|
29
|
Abstract
The accurate and precise determination of binding interactions plays a central role in fields such as drug discovery where structure-activity relationships guide the selection and optimization of drug leads. Binding is often assessed by monitoring the response caused by varying one of the binding partners in a functional assay or by using methods where the concentrations of free and/or bound ligand can be directly determined. In addition, there are also many approaches where binding leads to a change in the properties of the binding partner(s) that can be directly quantified such as an alteration in mass or in a spectroscopic signal. The analysis of data resulting from these techniques invariably relies on computer software that enable rapid fitting of the data to nonlinear multiparameter equations. The objective of this Perspective is to serve as a reminder of the basic assumptions that are used in deriving these equations and thus that should be considered during assay design and subsequent data analysis. The result is a set of guidelines for authors considering submitting their work to journals such as ACS Infectious Diseases.
Collapse
Affiliation(s)
- Peter J. Tonge
- Center for Advanced Study of Drug Action, Departments of Chemistry and Radiology, Stony Brook University, John S. Toll Drive, Stony Brook, New York 11794-3400, United States
| |
Collapse
|
30
|
Abstract
The nociceptin/orphanin FQ (N/OFQ) peptide receptor (NOP) is a G protein-coupled receptor involved in the regulation of several physiological functions and pathological conditions. Thus, researchers from academia and industry are pursuing NOP to discover and study novel pharmacological entities. In a multidisciplinary effort of pharmacologists, medicinal chemists, and molecular and structural biologists the mechanisms of NOP activation and inhibition have been, at least partially, disentangled. Here, we review the in vitro methodologies employed, which have contributed to our understanding of this target. We hope this chapter guides the reader through the mostly established assay platforms to investigate NOP pharmacology, and gives some hints taking advantage from what has already illuminated the function of other GPCRs. We analyzed the pharmacological results obtained with a large panel of NOP ligands investigated in several assays including receptor binding, stimulation of GTPγS binding, decrease of cAMP levels, calcium flux stimulation via chimeric G proteins, NOP/G protein and NOP/β-arrestin interaction, label-free assays such as dynamic mass redistribution, and bioassays such as the electrically stimulated mouse vas deferens.
Collapse
Affiliation(s)
- Davide Malfacini
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Girolamo Caló
- Section of Pharmacology, Department of Medical Sciences, National Institute of Neurosciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
31
|
Allikalt A, Kopanchuk S, Rinken A. Implementation of fluorescence anisotropy-based assay for the characterization of ligand binding to dopamine D1 receptors. Eur J Pharmacol 2018; 839:40-46. [DOI: 10.1016/j.ejphar.2018.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 01/11/2023]
|