1
|
Stanková J, Jurášek M, Hajdúch M, Džubák P. Terpenes and Terpenoids Conjugated with BODIPYs: An Overview of Biological and Chemical Properties. JOURNAL OF NATURAL PRODUCTS 2024; 87:1306-1319. [PMID: 38482846 PMCID: PMC11061839 DOI: 10.1021/acs.jnatprod.3c00961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 05/03/2024]
Abstract
Advancements in small-molecule research have created the need for sensitive techniques to accurately study biological processes in living systems. Fluorescent-labeled probes have become indispensable tools, particularly those that use boron-dipyrromethene (BODIPY) dyes. Terpenes and terpenoids are organic compounds found in nature that offer diverse biological activities, and BODIPY-based probes play a crucial role in studying these compounds. Monoterpene-BODIPY conjugates have exhibited potential for staining bacterial and fungal cells. Sesquiterpene-BODIPY derivatives have been used to study sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA), indicating their potential for drug development. Owing to their unique properties, diterpenes have been investigated using BODIPY conjugates to evaluate their mechanisms of action. Triterpene-BODIPY conjugates have been synthesized for biological studies, with different spacers affecting their cytotoxicity. Fluorescent probes, inspired by terpenoid-containing vitamins, have also been developed. Derivatives of tocopherol, coenzyme Q10, and vitamin K1 can provide insights into their oxidation-reduction abilities. All these probes have diverse applications, including the study of cell membranes to investigate immune responses and antioxidant properties. Further research in this field can help better understand and use terpenes and terpenoids in various biological contexts.
Collapse
Affiliation(s)
- Jarmila Stanková
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 77900 Olomouc, Czech Republic
| | - Michal Jurášek
- Department
of Chemistry of Natural Compounds, University
of Chemistry and Technology Prague, 16628 Prague, Czech Republic
| | - Marián Hajdúch
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 77900 Olomouc, Czech Republic
- Laboratory
of Experimental Medicine, Institute of Molecular and Translational
Medicine, University Hospital Olomouc, 77900 Olomouc, Czech Republic
| | - Petr Džubák
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 77900 Olomouc, Czech Republic
- Laboratory
of Experimental Medicine, Institute of Molecular and Translational
Medicine, University Hospital Olomouc, 77900 Olomouc, Czech Republic
| |
Collapse
|
2
|
Jia Y, Xu L, Wang L, Yan K, Chen J, Xu P, Di B, Yan F, Hu C. A light-up fluorescence probe for wash-free analysis of Mu-opioid receptor and ligand-binding events. Anal Chim Acta 2023; 1261:341220. [PMID: 37147056 DOI: 10.1016/j.aca.2023.341220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/01/2023] [Accepted: 04/13/2023] [Indexed: 05/07/2023]
Abstract
With the aggravated burden of opioid use disorder spreading worldwide, demands for new forms of opioid receptor agonist/antagonist constitute immense research interest. The Mu-opioid receptor (MOR) is currently in the spotlight on account of its general involvement in opioid-induced antinociception, tolerance and dependence. MOR binding assay, however, is often complicated by difficulty in MOR separation and purification, as well as the tedious procedure in standard biolayer interferometry and surface plasmon resonance measurements. To this end, we present TPE2N as a light-up fluorescent probe for MOR, which exhibits satisfactory performance in both live cells and lysates. TPE2N was elaborately designed based on the synergistic effect of twisted intramolecular charge-transfer and aggregation-induced emission by incorporating a tetraphenylethene unit to emit strong fluorescence in a restrained environment upon binding with MOR through the naloxone pharmacore. The developed assay enabled high-throughput screening of a compound library, and successfully identified three ligands as lead compounds for further development.
Collapse
Affiliation(s)
- Yan Jia
- China National Narcotics Control Commission-China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, No. 24 Tongjiaxiang Road, Nanjing, 210009, China
| | - Lili Xu
- China National Narcotics Control Commission-China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, No. 24 Tongjiaxiang Road, Nanjing, 210009, China
| | - Lancheng Wang
- China National Narcotics Control Commission-China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, No. 24 Tongjiaxiang Road, Nanjing, 210009, China
| | - Kun Yan
- China National Narcotics Control Commission-China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, No. 24 Tongjiaxiang Road, Nanjing, 210009, China
| | - Jieru Chen
- China National Narcotics Control Commission-China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, No. 24 Tongjiaxiang Road, Nanjing, 210009, China
| | - Pengcheng Xu
- China National Narcotics Control Commission-China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, No. 24 Tongjiaxiang Road, Nanjing, 210009, China
| | - Bin Di
- China National Narcotics Control Commission-China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, No. 24 Tongjiaxiang Road, Nanjing, 210009, China.
| | - Fang Yan
- China National Narcotics Control Commission-China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, No. 24 Tongjiaxiang Road, Nanjing, 210009, China.
| | - Chi Hu
- China National Narcotics Control Commission-China Pharmaceutical University Joint Laboratory on Key Technologies of Narcotics Control, No. 24 Tongjiaxiang Road, Nanjing, 210009, China.
| |
Collapse
|
3
|
Leon Duque MA, Vallavoju N, Woo CM. Chemical tools for the opioids. Mol Cell Neurosci 2023; 125:103845. [PMID: 36948231 PMCID: PMC10247539 DOI: 10.1016/j.mcn.2023.103845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/09/2023] [Accepted: 03/15/2023] [Indexed: 03/24/2023] Open
Abstract
The opioids are potent and widely used pain management medicines despite also possessing severe liabilities that have fueled the opioid crisis. The pharmacological properties of the opioids primarily derive from agonism or antagonism of the opioid receptors, but additional effects may arise from specific compounds, opioid receptors, or independent targets. The study of the opioids, their receptors, and the development of remediation strategies has benefitted from derivatization of the opioids as chemical tools. While these studies have primarily focused on the opioids in the context of the opioid receptors, these chemical tools may also play a role in delineating mechanisms that are independent of the opioid receptors. In this review, we describe recent advances in the development and applications of opioid derivatives as chemical tools and highlight opportunities for the future.
Collapse
Affiliation(s)
- Mark Anthony Leon Duque
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St, Cambridge, MA 02138, United States of America
| | - Nandini Vallavoju
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St, Cambridge, MA 02138, United States of America
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St, Cambridge, MA 02138, United States of America.
| |
Collapse
|
4
|
The chronological evolution of fluorescent GPCR probes for bioimaging. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
5
|
Giakomidi D, Bird MF, Guerrini R, Calo G, Lambert DG. Fluorescent opioid receptor ligands as tools to study opioid receptor function. J Pharmacol Toxicol Methods 2021; 113:107132. [PMID: 34728348 DOI: 10.1016/j.vascn.2021.107132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/12/2021] [Accepted: 10/25/2021] [Indexed: 11/30/2022]
Abstract
Opioid receptors are divided into the three classical types: MOP(μ:mu), DOP(δ:delta) and KOP(κ:kappa) that are naloxone-sensitive and an additional naloxone-insensitive nociceptin/orphanin FQ(N/OFQ) peptide receptor(NOP). Studies to determine opioid receptor location and turnover variably rely on; (i) measuring receptor mRNA, (ii) genetically tagging receptors, (iii) labelling receptors with radioligands, (iv) use of antibodies in immunohistochemistry/Western Blotting or (v) measuring receptor function coupled with the use of selective antagonists. All have their drawbacks with significant issues relating to mRNA not necessarily predicting protein, poor antibody selectivity and utility of radiolabels in low expression systems. In this minireview we discuss use of fluorescently labelled opioid receptor ligands. To maintain the pharmacological properties of the corresponding parent ligand fluorescently labelled ligands must take into account fluorophore (brightness and propensity to bleach), linker length and chemistry, and site to which the linker (and hence probe) will be attached. Use of donor and acceptor fluorophores with spectral overlap facilitates use in FRET type assays to determine proximity of ligand or tagged receptor pairs. There is a wide range of probes of agonist and antagonist nature for all four opioid receptor types; caution is needed with agonist probes due to the possibility for internalization. We have produced two novel ATTO based probes; DermorphinATTO488 (MOP) and N/OFQATTO594 (NOP). These probes label MOP and NOP in a range of preparations and using N/OFQATTO594 we demonstrate internalization and ligand-receptor interaction by FRET. Fluorescent opioid probes offer potential methodological advantages over more traditional use of antibodies and radiolabels.
Collapse
Affiliation(s)
- Despina Giakomidi
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Hodgkin Building, Leicester LE1 9HN. UK
| | - Mark F Bird
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Hodgkin Building, Leicester LE1 9HN. UK
| | - Remo Guerrini
- Department of Chemical, Pharmaceutical and Agricultural Sciences and LTTA, University of Ferrara, Italy
| | - Girolamo Calo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - David G Lambert
- Department of Cardiovascular Sciences (Anaesthesia, Critical Care and Pain Management), University of Leicester, Hodgkin Building, Leicester LE1 9HN. UK.
| |
Collapse
|
6
|
Borgarelli C, Klingl YE, Escamilla-Ayala A, Munck S, Van Den Bosch L, De Borggraeve WM, Ismalaj E. Lighting Up the Plasma Membrane: Development and Applications of Fluorescent Ligands for Transmembrane Proteins. Chemistry 2021; 27:8605-8641. [PMID: 33733502 DOI: 10.1002/chem.202100296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Indexed: 12/16/2022]
Abstract
Despite the fact that transmembrane proteins represent the main therapeutic targets for decades, complete and in-depth knowledge about their biochemical and pharmacological profiling is not fully available. In this regard, target-tailored small-molecule fluorescent ligands are a viable approach to fill in the missing pieces of the puzzle. Such tools, coupled with the ability of high-precision optical techniques to image with an unprecedented resolution at a single-molecule level, helped unraveling many of the conundrums related to plasma proteins' life-cycle and druggability. Herein, we review the recent progress made during the last two decades in fluorescent ligand design and potential applications in fluorescence microscopy of voltage-gated ion channels, ligand-gated ion channels and G-coupled protein receptors.
Collapse
Affiliation(s)
- Carlotta Borgarelli
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven Campus Arenberg Celestijnenlaan 200F -, box 2404, 3001, Leuven, Belgium
| | - Yvonne E Klingl
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Neurobiology, VIB, Center for Brain &, Disease Research, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Abril Escamilla-Ayala
- Center for Brain & Disease Research, & VIB BioImaging Core, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Department of Neurosciences, Leuven Brain Institute, KU Leuven, Campus Gasthuisberg O&N5 - box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Sebastian Munck
- Center for Brain & Disease Research, & VIB BioImaging Core, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Department of Neurosciences, Leuven Brain Institute, KU Leuven, Campus Gasthuisberg O&N5 - box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Neurobiology, VIB, Center for Brain &, Disease Research, VIB-KU Leuven Campus Gasthuisberg O&N5 -, box 602 Herestraat 49, 3000, Leuven, Belgium
| | - Wim M De Borggraeve
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven Campus Arenberg Celestijnenlaan 200F -, box 2404, 3001, Leuven, Belgium
| | - Ermal Ismalaj
- Department of Chemistry, Molecular Design and Synthesis, KU Leuven Campus Arenberg Celestijnenlaan 200F -, box 2404, 3001, Leuven, Belgium
| |
Collapse
|
7
|
New small molecule fluorescent probes for G protein-coupled receptors: valuable tools for drug discovery. Future Med Chem 2020; 13:63-90. [PMID: 33319586 DOI: 10.4155/fmc-2019-0327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are essential signaling proteins and tractable therapeutic targets. To develop new drug candidates, GPCR drug discovery programs require versatile, sensitive pharmacological tools for ligand binding and compound screening. With the availability of new imaging modalities and proximity-based ligand binding technologies, fluorescent ligands offer many advantages and are increasingly being used, yet labeling small molecules remains considerably more challenging relative to peptides. Focusing on recent fluorescent small molecule studies for family A GPCRs, this review addresses some of the key challenges, synthesis approaches and structure-activity relationship considerations, and discusses advantages of using high-resolution GPCR structures to inform conjugation strategies. While no single approach guarantees successful labeling without loss of affinity or selectivity, the choice of fluorophore, linker type and site of attachment have proved to be critical factors that can significantly affect their utility in drug discovery programs, and as discussed, can sometimes lead to very unexpected results.
Collapse
|
8
|
Drakopoulos A, Decker M. Development and Biological Applications of Fluorescent Opioid Ligands. Chempluschem 2020; 85:1354-1364. [DOI: 10.1002/cplu.202000212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/30/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Antonios Drakopoulos
- Pharmaceutical and Medicinal ChemistryInstitute of Pharmacy and Food ChemistryJulius Maximilian University of Würzburg 97074 Würzburg Germany
| | - Michael Decker
- Pharmaceutical and Medicinal ChemistryInstitute of Pharmacy and Food ChemistryJulius Maximilian University of Würzburg 97074 Würzburg Germany
| |
Collapse
|
9
|
Gentzsch C, Seier K, Drakopoulos A, Jobin M, Lanoiselée Y, Koszegi Z, Maurel D, Sounier R, Hübner H, Gmeiner P, Granier S, Calebiro D, Decker M. Selective and Wash-Resistant Fluorescent Dihydrocodeinone Derivatives Allow Single-Molecule Imaging of μ-Opioid Receptor Dimerization. Angew Chem Int Ed Engl 2020; 59:5958-5964. [PMID: 31808251 PMCID: PMC7125027 DOI: 10.1002/anie.201912683] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Indexed: 12/21/2022]
Abstract
μ-Opioid receptors (μ-ORs) play a critical role in the modulation of pain and mediate the effects of the most powerful analgesic drugs. Despite extensive efforts, it remains insufficiently understood how μ-ORs produce specific effects in living cells. We developed new fluorescent ligands based on the μ-OR antagonist E-p-nitrocinnamoylamino-dihydrocodeinone (CACO), that display high affinity, long residence time and pronounced selectivity. Using these ligands, we achieved single-molecule imaging of μ-ORs on the surface of living cells at physiological expression levels. Our results reveal a high heterogeneity in the diffusion of μ-ORs, with a relevant immobile fraction. Using a pair of fluorescent ligands of different color, we provide evidence that μ-ORs interact with each other to form short-lived homodimers on the plasma membrane. This approach provides a new strategy to investigate μ-OR pharmacology at single-molecule level.
Collapse
Affiliation(s)
- Christian Gentzsch
- Pharmaceutical and Medicinal ChemistryInstitute of Pharmacy and Food ChemistryJulius Maximilian University of WürzburgAm Hubland97074WürzburgGermany
| | - Kerstin Seier
- Institute of Pharmacology and ToxicologyJulius Maximilian University of WürzburgVersbacher Strasse 997078WürzburgGermany
| | - Antonios Drakopoulos
- Pharmaceutical and Medicinal ChemistryInstitute of Pharmacy and Food ChemistryJulius Maximilian University of WürzburgAm Hubland97074WürzburgGermany
| | - Marie‐Lise Jobin
- Institute of Pharmacology and ToxicologyJulius Maximilian University of WürzburgVersbacher Strasse 997078WürzburgGermany
| | - Yann Lanoiselée
- Institute of Metabolism and Systems Research & Centre of Membrane Proteins and ReceptorsUniversity of BirminghamIBR Tower, Level 2, EdgbastonBirminghamB152TTUK
| | - Zsombor Koszegi
- Institute of Metabolism and Systems Research & Centre of Membrane Proteins and ReceptorsUniversity of BirminghamIBR Tower, Level 2, EdgbastonBirminghamB152TTUK
| | - Damien Maurel
- ARPEGE (Pharmacology Screening Interactome) platform facilityInstitut de Génomique FonctionnelleUniversité de Montpellier, CNRS, INSERM141, rue de la Cardonille34094Montpellier Cedex 05France
| | - Rémy Sounier
- Institut de Génomique FonctionnelleUniversité de Montpellier, CNRS, INSERM141, rue de la Cardonille34094Montpellier Cedex 05France
| | - Harald Hübner
- Medicinal ChemistryDepartment of Chemistry and PharmacyFriedrich-Alexander University of Erlangen-Nuremberg91058ErlangenGermany
| | - Peter Gmeiner
- Medicinal ChemistryDepartment of Chemistry and PharmacyFriedrich-Alexander University of Erlangen-Nuremberg91058ErlangenGermany
| | - Sébastien Granier
- Institut de Génomique FonctionnelleUniversité de Montpellier, CNRS, INSERM141, rue de la Cardonille34094Montpellier Cedex 05France
| | - Davide Calebiro
- Institute of Pharmacology and ToxicologyJulius Maximilian University of WürzburgVersbacher Strasse 997078WürzburgGermany
- Institute of Metabolism and Systems Research & Centre of Membrane Proteins and ReceptorsUniversity of BirminghamIBR Tower, Level 2, EdgbastonBirminghamB152TTUK
| | - Michael Decker
- Pharmaceutical and Medicinal ChemistryInstitute of Pharmacy and Food ChemistryJulius Maximilian University of WürzburgAm Hubland97074WürzburgGermany
| |
Collapse
|
10
|
Gentzsch C, Seier K, Drakopoulos A, Jobin M, Lanoiselée Y, Koszegi Z, Maurel D, Sounier R, Hübner H, Gmeiner P, Granier S, Calebiro D, Decker M. Selective and Wash‐Resistant Fluorescent Dihydrocodeinone Derivatives Allow Single‐Molecule Imaging of μ‐Opioid Receptor Dimerization. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201912683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Christian Gentzsch
- Pharmaceutical and Medicinal ChemistryInstitute of Pharmacy and Food ChemistryJulius Maximilian University of Würzburg Am Hubland 97074 Würzburg Germany
| | - Kerstin Seier
- Institute of Pharmacology and ToxicologyJulius Maximilian University of Würzburg Versbacher Strasse 9 97078 Würzburg Germany
| | - Antonios Drakopoulos
- Pharmaceutical and Medicinal ChemistryInstitute of Pharmacy and Food ChemistryJulius Maximilian University of Würzburg Am Hubland 97074 Würzburg Germany
| | - Marie‐Lise Jobin
- Institute of Pharmacology and ToxicologyJulius Maximilian University of Würzburg Versbacher Strasse 9 97078 Würzburg Germany
| | - Yann Lanoiselée
- Institute of Metabolism and Systems Research & Centre of Membrane Proteins and ReceptorsUniversity of Birmingham IBR Tower, Level 2, Edgbaston Birmingham B152TT UK
| | - Zsombor Koszegi
- Institute of Metabolism and Systems Research & Centre of Membrane Proteins and ReceptorsUniversity of Birmingham IBR Tower, Level 2, Edgbaston Birmingham B152TT UK
| | - Damien Maurel
- ARPEGE (Pharmacology Screening Interactome) platform facilityInstitut de Génomique FonctionnelleUniversité de Montpellier, CNRS, INSERM 141, rue de la Cardonille 34094 Montpellier Cedex 05 France
| | - Rémy Sounier
- Institut de Génomique FonctionnelleUniversité de Montpellier, CNRS, INSERM 141, rue de la Cardonille 34094 Montpellier Cedex 05 France
| | - Harald Hübner
- Medicinal ChemistryDepartment of Chemistry and PharmacyFriedrich-Alexander University of Erlangen-Nuremberg 91058 Erlangen Germany
| | - Peter Gmeiner
- Medicinal ChemistryDepartment of Chemistry and PharmacyFriedrich-Alexander University of Erlangen-Nuremberg 91058 Erlangen Germany
| | - Sébastien Granier
- Institut de Génomique FonctionnelleUniversité de Montpellier, CNRS, INSERM 141, rue de la Cardonille 34094 Montpellier Cedex 05 France
| | - Davide Calebiro
- Institute of Pharmacology and ToxicologyJulius Maximilian University of Würzburg Versbacher Strasse 9 97078 Würzburg Germany
- Institute of Metabolism and Systems Research & Centre of Membrane Proteins and ReceptorsUniversity of Birmingham IBR Tower, Level 2, Edgbaston Birmingham B152TT UK
| | - Michael Decker
- Pharmaceutical and Medicinal ChemistryInstitute of Pharmacy and Food ChemistryJulius Maximilian University of Würzburg Am Hubland 97074 Würzburg Germany
| |
Collapse
|
11
|
Arttamangkul S, Plazek A, Platt EJ, Jin H, Murray TF, Birdsong WT, Rice KC, Farrens DL, Williams JT. Visualizing endogenous opioid receptors in living neurons using ligand-directed chemistry. eLife 2019; 8:49319. [PMID: 31589142 PMCID: PMC6809603 DOI: 10.7554/elife.49319] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/06/2019] [Indexed: 12/11/2022] Open
Abstract
Identifying neurons that have functional opioid receptors is fundamental for the understanding of the cellular, synaptic and systems actions of opioids. Current techniques are limited to post hoc analyses of fixed tissues. Here we developed a fluorescent probe, naltrexamine-acylimidazole (NAI), to label opioid receptors based on a chemical approach termed ‘traceless affinity labeling’. In this approach, a high affinity antagonist naltrexamine is used as the guide molecule for a transferring reaction of acylimidazole at the receptor. This reaction generates a fluorescent dye covalently linked to the receptor while naltrexamine is liberated and leaves the binding site. The labeling induced by this reagent allowed visualization of opioid-sensitive neurons in rat and mouse brains without loss of function of the fluorescently labeled receptors. The ability to locate endogenous receptors in living tissues will aid considerably in establishing the distribution and physiological role of opioid receptors in the CNS of wild type animals.
Collapse
Affiliation(s)
- Seksiri Arttamangkul
- The Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Andrew Plazek
- Medicinal Chemistry Core, Oregon Health & Science University, Portland, United States
| | - Emily J Platt
- Department of Biochemistry and Molecular Biology, School of Medicine, Oregon Health & Science University, Portland, United States
| | - Haihong Jin
- Medicinal Chemistry Core, Oregon Health & Science University, Portland, United States
| | - Thomas F Murray
- Department of Pharmacology, School of Medicine, Creighton University, Omaha, United States
| | - William T Birdsong
- The Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Kenner C Rice
- Drug Design and Synthesis Section, Intramural Research Program, NIDA and NIAAA, Bethesda, United States
| | - David L Farrens
- Department of Biochemistry and Molecular Biology, School of Medicine, Oregon Health & Science University, Portland, United States
| | - John T Williams
- The Vollum Institute, Oregon Health & Science University, Portland, United States
| |
Collapse
|
12
|
Lam R, Gondin AB, Canals M, Kellam B, Briddon SJ, Graham B, Scammells PJ. Fluorescently Labeled Morphine Derivatives for Bioimaging Studies. J Med Chem 2018; 61:1316-1329. [DOI: 10.1021/acs.jmedchem.7b01811] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Raymond Lam
- School
of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Arisbel B. Gondin
- Cell Signalling
Research Group, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, U.K
| | | | - Barrie Kellam
- School
of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Stephen J. Briddon
- Cell Signalling
Research Group, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, U.K
| | | | | |
Collapse
|
13
|
Leong C, Neumann C, Ramasamy S, Rout B, Yi Wee L, Bigliardi-Qi M, Bigliardi PL. Investigating endogenous µ-opioid receptors in human keratinocytes as pharmacological targets using novel fluorescent ligand. PLoS One 2017; 12:e0188607. [PMID: 29211767 PMCID: PMC5718609 DOI: 10.1371/journal.pone.0188607] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 11/09/2017] [Indexed: 12/14/2022] Open
Abstract
Opioids in skin function during stress response, regeneration, ageing and, particularly in regulating sensation. In chronic pruritus, topical treatment with Naltrexone changes μ-opioid receptor (μ-OR) localization to relieve itch. The molecular mechanisms behind the effects of Naltrexone on μ-OR function in reduction of itching behavior has not been studied. There is an immediate need to understand the endogenous complexity of μ-OR dynamics in normal and pathological skin conditions. Here we evaluate real-time behavior of μ-OR-Endomorphine complexes in the presence of agonist and antagonists. The μ-OR ligand Endomorphine-1 (EM) was conjugated to the fluorescent dye Tetramethylrhodamine (TAMRA) to investigate the effects of agonist and antagonists in N/TERT-1 keratinocytes. The cellular localization of the EM-TAMRA was followed through time resolved confocal microscopy and population analysis was performed by flow cytometry. The in vitro analyses demonstrate fast internalization and trafficking of the endogenous EM-TAMRA-μ-OR interactions in a qualitative manner. Competition with Endomorphine-1, Naltrexone and CTOP show both canonical and non-canonical effects in basal and differentiated keratinocytes. Acute and chronic treatment with Naltrexone and Endomorphine-1 increases EM-TAMRA binding to skin cells. Although Naltrexone is clinically effective in relieving itch, the mechanisms behind re-distribution of μ-ORs during clinical treatments are not known. Our study has given insight into cellular mechanisms of μ-OR ligand-receptor interactions after opioid agonist and antagonist treatments in vitro. These findings potentially offer opportunities in using novel treatment strategies for skin and peripheral sensory disorders.
Collapse
Affiliation(s)
- Cheryl Leong
- Institute of Medical Biology, Agency for Science Technology & Research (A*STAR), Singapore, Singapore
| | - Christine Neumann
- Institute of Medical Biology, Agency for Science Technology & Research (A*STAR), Singapore, Singapore
| | - Srinivas Ramasamy
- Institute of Medical Biology, Agency for Science Technology & Research (A*STAR), Singapore, Singapore
| | - Bhimsen Rout
- Institute of Medical Biology, Agency for Science Technology & Research (A*STAR), Singapore, Singapore
| | - Lim Yi Wee
- Institute of Chemical and Engineering Sciences, Agency for Science Technology & Research (A*STAR), Singapore, Singapore
| | - Mei Bigliardi-Qi
- Institute of Medical Biology, Agency for Science Technology & Research (A*STAR), Singapore, Singapore
- * E-mail: , (PB); , (MB)
| | - Paul L. Bigliardi
- Institute of Medical Biology, Agency for Science Technology & Research (A*STAR), Singapore, Singapore
- National University Hospital, Division of Rheumatology, University Medicine Cluster, Singapore, Singapore
- * E-mail: , (PB); , (MB)
| |
Collapse
|
14
|
Schembri LS, Stoddart LA, Briddon SJ, Kellam B, Canals M, Graham B, Scammells PJ. Synthesis, Biological Evaluation, and Utility of Fluorescent Ligands Targeting the μ-Opioid Receptor. J Med Chem 2015; 58:9754-67. [DOI: 10.1021/acs.jmedchem.5b01664] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
| | - Leigh A. Stoddart
- Cell
Signaling Research Group, School of Life Sciences, Queen’s
Medical Centre, University of Nottingham, Nottingham NG7 2UH, U.K
| | - Stephen J. Briddon
- Cell
Signaling Research Group, School of Life Sciences, Queen’s
Medical Centre, University of Nottingham, Nottingham NG7 2UH, U.K
| | - Barrie Kellam
- School
of Pharmacy, Centre for Biomolecular Sciences, University of Nottingham, Nottingham NG7 2RD, U.K
| | | | | | | |
Collapse
|
15
|
Sridharan R, Zuber J, Connelly SM, Mathew E, Dumont ME. Fluorescent approaches for understanding interactions of ligands with G protein coupled receptors. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1838:15-33. [PMID: 24055822 PMCID: PMC3926105 DOI: 10.1016/j.bbamem.2013.09.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 09/03/2013] [Accepted: 09/08/2013] [Indexed: 11/18/2022]
Abstract
G protein coupled receptors are responsible for a wide variety of signaling responses in diverse cell types. Despite major advances in the determination of structures of this class of receptors, the underlying mechanisms by which binding of different types of ligands specifically elicits particular signaling responses remain unclear. The use of fluorescence spectroscopy can provide important information about the process of ligand binding and ligand dependent conformational changes in receptors, especially kinetic aspects of these processes that can be difficult to extract from X-ray structures. We present an overview of the extensive array of fluorescent ligands that have been used in studies of G protein coupled receptors and describe spectroscopic approaches for assaying binding and probing the environment of receptor-bound ligands with particular attention to examples involving yeast pheromone receptors. In addition, we discuss the use of fluorescence spectroscopy for detecting and characterizing conformational changes in receptors induced by the binding of ligands. Such studies have provided strong evidence for diversity of receptor conformations elicited by different ligands, consistent with the idea that GPCRs are not simple on and off switches. This diversity of states constitutes an underlying mechanistic basis for biased agonism, the observation that different stimuli can produce different responses from a single receptor. It is likely that continued technical advances will allow fluorescence spectroscopy to play an important role in continued probing of structural transitions in G protein coupled receptors. This article is part of a Special Issue entitled: Structural and biophysical characterisation of membrane protein-ligand binding.
Collapse
Affiliation(s)
- Rajashri Sridharan
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Jeffrey Zuber
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Sara M. Connelly
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Elizabeth Mathew
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
| | - Mark E. Dumont
- Department of Biochemistry and Biophysics, P.O. Box 712, University of Rochester Medical Center, Rochester, NY 14642
- Department of Pediatrics, P.O. Box 777, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
16
|
Sugimoto H, Muto M, Tanaka T, Osuka A. Synthesis of BODIPY-Appended Subporphyrins. European J Org Chem 2010. [DOI: 10.1002/ejoc.201001188] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
17
|
Baker JG, Middleton R, Adams L, May LT, Briddon SJ, Kellam B, Hill SJ. Influence of fluorophore and linker composition on the pharmacology of fluorescent adenosine A1 receptor ligands. Br J Pharmacol 2010; 159:772-86. [PMID: 20105183 PMCID: PMC2829203 DOI: 10.1111/j.1476-5381.2009.00488.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background and purpose: The introduction of fluorescence-based techniques, and in particular the development of fluorescent ligands, has allowed the study of G protein-coupled receptor pharmacology at the single cell and single molecule level. This study evaluated how the physicochemical nature of the linker and the fluorophore affected the pharmacological properties of fluorescent agonists and antagonists. Experimental approach: Chinese hamster ovary cells stably expressing the human adenosine A1 receptor and a cyclic 3′,5′ adenosine monophosphate response element-secreted placental alkaline phosphatase (CRE-SPAP) reporter gene, together with whole cell [3H]-8-cyclopentyl-1,3-dipropylxanthine (DPCPX) radioligand binding, were used to evaluate the pharmacological properties of a range of fluorescent ligands based on the antagonist xanthine amine congener (XAC) and the agonist 5′ (N-ethylcarboxamido) adenosine (NECA). Key results: Derivatives of NECA and XAC with different fluorophores, but equivalent linker length, showed significant differences in their binding properties to the adenosine A1 receptor. The BODIPY 630/650 derivatives had the highest affinity. Linker length also affected the pharmacological properties, depending on the fluorophore used. Particularly in fluorescent agonists, higher agonist potency could be achieved with large or small linkers for dansyl and BODIPY 630/650 derivatives, respectively. Conclusions and implications: The pharmacology of a fluorescent ligand was critically influenced by both the fluorophore and the associated linker. Furthermore, our data strongly suggest that the physicochemical properties of the fluorophore/linker pairing determine where in the environment of the target receptor the fluorophore is placed, and this, together with the environmental sensitivity of the resulting fluorescence, may finally decide its utility as a fluorescent probe. This article is part of a themed section on Imaging in Pharmacology. To view the editorial for this themed section visit http://dx.doi.org/10.1111/j.1476-5381.2010.00685.x
Collapse
Affiliation(s)
- Jillian G Baker
- Institute of Cell Signalling, School of Biomedical Sciences, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | | | | | | | | | | | | |
Collapse
|
18
|
|
19
|
Middleton RJ, Briddon SJ, Cordeaux Y, Yates AS, Dale CL, George MW, Baker JG, Hill SJ, Kellam B. New fluorescent adenosine A1-receptor agonists that allow quantification of ligand-receptor interactions in microdomains of single living cells. J Med Chem 2007; 50:782-93. [PMID: 17249651 DOI: 10.1021/jm061279i] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fluorescence spectroscopy is becoming a valuable addition to the array of techniques available for scrutinizing ligand-receptor interactions in biological systems. In particular, scanning confocal microscopy and fluorescence correlation spectroscopy (FCS) allow the noninvasive imaging and quantification of these interactions in single living cells. To address the emerging need for fluorescently labeled ligands to support these technologies, we have developed a series of red-emitting agonists for the human adenosine A1-receptor that, collectively, are N6-aminoalkyl derivatives of adenosine or adenosine 5'-N-ethyl carboxamide. The agonists, which incorporate the commercially available fluorophore BODIPY [630/650], retain potent and efficacious agonist activity, as demonstrated by their ability to inhibit cAMP accumulation in chinese hamster ovary cells expressing the human adenosine A1-receptor. Visualization and confirmation of ligand-receptor interactions at the cell membrane were accomplished using confocal microscopy, and their suitability for use in FCS was demonstrated by quantification of agonist binding in small areas of cell membrane.
Collapse
Affiliation(s)
- Richard J Middleton
- School of Pharmacy, Centre for Biomolecular Sciences, and School of Chemistry, University of Nottingham, University Park, Nottingham, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
|
21
|
Abstract
[reaction: see text] Photochromic dithienylethene moieties were covalently attached to fluorescent 4,4-difluoro-8-(4'-iodophenyl)-1,3,5,7-tetramethyl-4-bora-3a,4a-diaza-s-indacene (iodo-BODIPY) via a phenylacetylene linker. UV light induced isomerization of the photochrome results in significant decrease in fluorescence intensity. This fluorescence can be recovered with visible light. Steady-state fluorescence measurements demonstrate that the emission of the dye can be modulated by external light. An intramolecular energy transfer mechanism accounts for the fluorescence quenching in the UV light produced isomers.
Collapse
Affiliation(s)
- Tatiana A Golovkova
- Center for Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, USA
| | | | | |
Collapse
|
22
|
Houghten RA, Dooley CT, Appel JR. De novo identification of highly active fluorescent kappa opioid ligands from a rhodamine labeled tetrapeptide positional scanning library. Bioorg Med Chem Lett 2004; 14:1947-51. [PMID: 15050634 DOI: 10.1016/j.bmcl.2004.01.090] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2003] [Accepted: 01/26/2004] [Indexed: 11/22/2022]
Abstract
Highly active fluorescent compounds having kappa opioid activity were identified following the screening in a kappa-specific radioligand binding assay of a positional scanning tetrapeptide combinatorial library in which every tetrapeptide was fluorescently labeled. Lissamine rhodamine B sulfonyl chloride was coupled to the N terminal of a mixture-based tetrapeptide positional scanning library made up of over 7.3 million tetrapeptides. Upon determination of the most active mixtures for each position of the library in the kappa binding assay, individual rhodamine labeled tetrapeptides were then synthesized and tested to determine their activities. Eight individual rhodamine labeled peptides were identified that were specific for the kappa opioid receptor, having binding affinities ranging from 5-20 nM. These peptides were poor inhibitors at the mu and delta receptors (K(i)>5,000 nM). Furthermore, neither rhodamine itself nor these same tetrapeptides lacking the N-terminal rhodamine had any significant activity at the kappa receptor, indicating that both the tetrapeptide sequence and the rhodamine moiety are required for receptor binding. This study has demonstrated that novel fluorescent compounds with intrinsic activity can be identified through the use of combinatorial chemistry.
Collapse
Affiliation(s)
- Richard A Houghten
- Torrey Pines Institute for Molecular Studies, 3550 General Atomics Court, San Diego, CA 92121, USA.
| | | | | |
Collapse
|
23
|
Briddon SJ, Middleton RJ, Cordeaux Y, Flavin FM, Weinstein JA, George MW, Kellam B, Hill SJ. Quantitative analysis of the formation and diffusion of A1-adenosine receptor-antagonist complexes in single living cells. Proc Natl Acad Sci U S A 2004; 101:4673-8. [PMID: 15070776 PMCID: PMC384805 DOI: 10.1073/pnas.0400420101] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2003] [Indexed: 11/18/2022] Open
Abstract
The A1-adenosine receptor (A1-AR) is a G protein-coupled receptor that mediates many of the physiological effects of adenosine in the brain, heart, kidney, and adipocytes. Currently, ligand interactions with the A1-AR can be quantified on large cell populations only by using radioligand binding. To increase the resolution of these measurements, we have designed and characterized a previously undescribed fluorescent antagonist for the A1-AR, XAC-BY630, based on xanthine amine congener (XAC). This compound has been used to quantify ligand-receptor binding at a single cell level using fluorescence correlation spectroscopy (FCS). XAC-BY630 was a competitive antagonist of A1-AR-mediated inhibition of cAMP accumulation [log10 of the affinity constant (pKb) = 6.7)] and stimulation of inositol phosphate accumulation (pKb = 6.5). Specific binding of XAC-BY630 to cell surface A1-AR could also be visualized in living Chinese hamster ovary (CHO)-A1 cells by using confocal microscopy. FCS analysis of XAC-BY630 binding to the membrane of CHO-A1 cells revealed three components with diffusion times (tauD) of 62 micros (tauD1, free ligand), 17 ms (tauD2, A1-AR-ligand), and 320 ms (tauD3). Confirmation that tauD2 resulted from diffusion of ligand-receptor complexes came from the similar diffusion time observed for the fluorescent A1-AR-Topaz fusion protein (15 ms). Quantification of tauD2 showed that the number of receptor-ligand complexes increased with increasing free ligand concentration and was decreased by the selective A1-AR antagonist, 8-cyclopentyl-1,3-dipropylxanthine. The combination of FCS with XAC-BY630 will be a powerful tool for the characterization of ligand-A1-AR interactions in single living cells in health and disease.
Collapse
Affiliation(s)
- S J Briddon
- Institute of Cell Signalling, Medical School, University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
24
|
|
25
|
Phillips TM. Analysis of single-cell cultures by immunoaffinity capillary electrophoresis with laser-induced fluorescence detection. LUMINESCENCE 2001; 16:145-52. [PMID: 11312540 DOI: 10.1002/bio.645] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Neuropeptide regulation of immunological activity is becoming an important issue in both basic and clinical sciences, necessitating the need for analysis to be performed at the single-cell level. A microsampling procedure has been developed for studying secretion of biologically important peptides from neuropeptide-stimulated lymphocytes, based on microdialysis sampling coupled to immunoaffinity capillary electrophoresis (ICE), with laser-induced fluorescence (LIF) detection using a fibre-optic spectrometer and diode laser excitation. The system demonstrated a limit of detection in the high attomole (10(-18) mol/L) range with pure standards and was capable of monitoring secretion from a single cell over time. Using this system it was possible to differentiate the effects of four neuropeptides on both T and B cell release of regulatory cytokines. CD4(+) lymphocytes demonstrated a 7.5-fold increase in cytokine secretion over baseline following stimulation with substance P (SP) and calcitonin gene-related peptide (CGRP). B cells responded to CGRP and vasoactive intestinal peptide (VIP) stimulation (5.5-fold increase), but not to SP. These changes took place 12--20 h post-stimulation and, once the peak secretion had been reached, remained at that level for the duration of the experiment. This system demonstrates the ability to perform high sensitivity measurements on microsamples of biological fluids.
Collapse
Affiliation(s)
- T M Phillips
- Immunochemistry Laboratory, George Washington University Medical Centre, Washington, DC 20037, USA.
| |
Collapse
|
26
|
Kumar V, Murray TF, Aldrich JV. Extended TIP(P) analogues as precursors for labeled delta-opioid receptor ligands. J Med Chem 2000; 43:5050-4. [PMID: 11150177 DOI: 10.1021/jm000362h] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tyr-Tic-Phe-Phe-OH (TIPP) and the shorter Tyr-Tic-Phe-OH (TIP) peptides are potent and highly selective antagonists at the delta-opioid receptor and, therefore, are ideal candidates for the attachment of labels to assist in the study of delta-opioid receptors. Peptides extended at the C-terminus with residues which can be used as handles for further modification and/or labeling (i.e. Asx, Glx, and Lys) were synthesized. The TIPP-D/L-Asx/Glx derivatives exhibited similar delta-receptor affinity to TIPP (K(i) = 5-10 nM vs K(i) = 6 nM), and neither the location of the carboxylic acid moiety nor the stereochemistry of the C-terminal residue significantly affected the delta-receptor affinity of these derivatives. Extension of TIPP with an additional residue did not increase mu-receptor affinity, even though the position of the acidic group, which imparts delta-receptor selectivity to TIPP, was shifted relative to the carboxylic acid moiety of TIPP. The delta-receptor affinities of the TIP-D/L-Asx/Glx derivatives were found to be influenced mainly by the position of the carboxylic acid function rather than the stereochemistry of the C-terminal residue. TIP(P)-D/L-Lys(Ac)-OH derivatives exhibited moderate delta-receptor affinity (K(i)(delta) = 16-28 nM). The most potent compounds found in the extended TIP(P) series were TIPP-D-Gln-OH and TIP-D-Gln-OH (K(i)(delta) = 5 nM) which had similar affinities to TIPP.
Collapse
Affiliation(s)
- V Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201, USA
| | | | | |
Collapse
|
27
|
Arttamangkul S, Alvarez-Maubecin V, Thomas G, Williams JT, Grandy DK. Binding and internalization of fluorescent opioid peptide conjugates in living cells. Mol Pharmacol 2000; 58:1570-80. [PMID: 11093798 DOI: 10.1124/mol.58.6.1570] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The dynamics of agonist-stimulated opioid receptor internalization and trafficking have been difficult to study in living cells in part because the available probes were inadequate. To overcome this obstacle, six new fluorescent opioid peptides were developed. Dermorphin (DERM), deltorphin (DELT), TIPP, and endomorphin were conjugated to BODIPY TR or Alexa Fluor 488, two fluorescent dyes with distinct hydrophobic properties. In membrane binding assays the fluorescent conjugates DERM-A488 or -BTR, DELT-A488 or -BTR, and TIPP-A488 displayed good binding affinity and selectivity for mu- and delta-opioid receptor subtypes. Furthermore, the fluorescent conjugates of dermorphin and deltorphin were biologically active as demonstrated by their ability to hyperpolarize locus coeruleus neurons (DERM-A488 or -BTR) and inhibit calcium currents in NG108-15 (DELT-A488). Both of these responses were antagonized by naloxone. In conjunction with confocal fluorescent microscopy the trafficking of these fluorescent ligands was monitored in real-time. The internalization of these ligands by mu- and delta-opioid receptors was found to be naloxone-sensitive and temperature-dependent. Interestingly, once these ligands were internalized the fluorescent puncta that formed became distributed in one of two patterns. In Chinese hamster ovary cells heterologously expressing either mu- or delta-opioid receptors the intracellular puncta were concentrated in the perinuclear region of the cell, whereas they were distributed throughout the cytoplasm in cells derived from either NG108-15 or SH-SY5Y cells. In summary, we have demonstrated that these novel, fluorescent opioid peptide conjugates permit real-time visual tracking of receptor-ligand complexes, including their internalization and trafficking, in living cells.
Collapse
Affiliation(s)
- S Arttamangkul
- Department of Physiology and Pharmacology, Oregon Health Sciences University, Portland, Oregon 97201, USA
| | | | | | | | | |
Collapse
|
28
|
Madsen BW, Beglan CL, Spivak CE. Fluorescein-labeled naloxone binding to mu opioid receptors on live Chinese hamster ovary cells using confocal fluorescent microscopy. J Neurosci Methods 2000; 97:123-31. [PMID: 10788666 DOI: 10.1016/s0165-0270(00)00175-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A general method of confocal laser scanning microscopy was used to demonstrate specific binding of fluorescein-labeled naloxone (FNAL, 10-50 nM) to stably transfected mu opioid receptors on live Chinese hamster ovary cells. Nonspecific binding was visually indistinguishable from autofluorescence in cells with intact cell membranes. Fluorescent labeling of cell perimeters, not present in control nontransfected cells, reversed in transfected cells upon washout of FNAL or following the addition of either unlabeled naloxone (25 microM) or the mu specific antagonist CTOP (1 microM). The addition of the delta and kappa specific agonists DPDPE (1 microM) and U50488 (1 microM), respectively, failed to reverse the labeling. Further evidence of specific binding was obtained from kinetic experiments, where it was observed that only transfected cells showed a time-dependent exponential change in fluorescence that permitted estimation of association and dissociation binding rate constants of (5.8+/-0.5, mean+/-S.E.M.)x10(5) M(-1) s(-1) and (3.3+/-0.6)x10(-3) s(-1), respectively and a kinetically derived dissociation constant of 5.7+/-1.4 nM. These estimates were comparable to those obtained under similar conditions in radioligand binding experiments using [3H]-naloxone.
Collapse
MESH Headings
- Animals
- Artifacts
- Binding, Competitive
- CHO Cells
- Cloning, Molecular
- Contrast Media/metabolism
- Contrast Media/pharmacology
- Cricetinae
- Fluorescein/metabolism
- Fluorescein/pharmacology
- Kinetics
- Microscopy, Confocal/methods
- Microscopy, Fluorescence/methods
- Naloxone/metabolism
- Naloxone/pharmacology
- Narcotic Antagonists/metabolism
- Narcotic Antagonists/pharmacology
- Radioligand Assay
- Receptors, Opioid, mu/analysis
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Sensitivity and Specificity
- Transfection
- Tritium
Collapse
Affiliation(s)
- B W Madsen
- Department of Pharmacology, University of Western Australia, Nedlands, Australia
| | | | | |
Collapse
|
29
|
Emmerson PJ, Clark MJ, Medzihradsky F, Remmers AE. Membrane microviscosity modulates mu-opioid receptor conformational transitions and agonist efficacy. J Neurochem 1999; 73:289-300. [PMID: 10386982 DOI: 10.1046/j.1471-4159.1999.0730289.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The influence of membrane microviscosity on mu-opioid agonist and antagonist binding, as well as agonist efficacy, was examined in membranes prepared from SH-SY5Y cells and from a C6 glioma cell line stably expressing the rat mu-opioid receptor (C6mu). Addition of cholesteryl hemisuccinate (CHS) to cell membranes increased membrane microviscosity and reduced the inhibitory effect of sodium and guanine nucleotides on the affinity of the full agonists sufentanil and [D-Ala2,N-MePhe4,Gly-ol5]enkephalin (DAMGO) for the mu-opioid receptor. Binding of the antagonists [3H]naltrexone and [3H]diprenorphine and the partial agonist nalbuphine was unaffected by CHS. The effect of CHS on agonist binding was reversed by subsequent addition of cis-vaccenic acid, suggesting that the effect of CHS is the result of increased membrane microviscosity and not a specific sterol-receptor interaction. CHS addition increased the potency of DAMGO to stimulate guanosine-5'-O-(3-[35S]thio)triphosphate binding by fourfold, whereas the potency of nalbuphine was unaffected. However, nalbuphine efficacy relative to that of the full agonist DAMGO was strongly increased in CHS-treated membranes compared with that in control membranes. Membrane rigidification also resulted in an increased efficacy for the partial agonists meperidine, profadol, and butorphanol relative to that of DAMGO as measured by agonist-stimulated GTPase activity in control and CHS-modified membranes. These findings support a regulatory role for membrane microviscosity in receptor-mediated G protein activation.
Collapse
Affiliation(s)
- P J Emmerson
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, USA
| | | | | | | |
Collapse
|
30
|
Beaudet A, Nouel D, Stroh T, Vandenbulcke F, Dal-Farra C, Vincent JP. Fluorescent ligands for studying neuropeptide receptors by confocal microscopy. Braz J Med Biol Res 1998; 31:1479-89. [PMID: 9921286 DOI: 10.1590/s0100-879x1998001100017] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This paper reviews the use of confocal microscopy as it pertains to the identification of G-protein coupled receptors and the study of their dynamic properties in cell cultures and in mammalian brain following their tagging with specific fluorescent ligands. Principles that should guide the choice of suitable ligands and fluorophores are discussed. Examples are provided from the work carried out in the authors' laboratory using custom synthetized fluoresceinylated or BODIPY-tagged bioactive peptides. The results show that confocal microscopic detection of specifically bound fluorescent ligands permits high resolution appraisal of neuropeptide receptor distribution both in cell culture and in brain sections. Within the framework of time course experiments, it also allows for a dynamic assessment of the internalization and subsequent intracellular trafficking of bound fluorescent molecules. Thus, it was found that neurotensin, somatostatin and mu- and delta-selective opioid peptides are internalized in a receptor-dependent fashion and according to receptor-specific patterns into their target cells. In the case of neurotensin, this internalization process was found to be clathrin-mediated, to proceed through classical endosomal pathways and, in neurons, to result in a mobilization of newly formed endosomes from neural processes to nerve cell bodies and from the periphery of cell bodies towards the perinuclear zone. These mechanisms are likely to play an important role for ligand inactivation, receptor regulation and perhaps also transmembrane signaling.
Collapse
Affiliation(s)
- A Beaudet
- Montreal Neurological Institute, McGill University, Quebec, Canada.
| | | | | | | | | | | |
Collapse
|