1
|
Hanske A, Nazaré M, Grether U. Chemical Probes for Investigating the Endocannabinoid System. Curr Top Behav Neurosci 2025. [PMID: 39747798 DOI: 10.1007/7854_2024_563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Cannabis sativa has been used therapeutically since early civilizations, with key cannabinoids Δ9-tetrahydrocannabinol (THC) 3.1 and cannabidiol characterized in the 1960s, leading to the discovery of cannabinoid receptors type 1 (CB1R) and type 2 (CB2R) and the endocannabinoid system (ECS) in the 1990s. The ECS, involving endogenous ligands like 2-arachidonoylglycerol (2-AG) 1.1, anandamide (N-arachidonoylethanolamine (AEA)) 1.2, and various proteins, regulates vital processes such as sleep, appetite, and memory, and holds significant therapeutic potential, especially for neurological disorders. Small molecule-derived pharmacological tools, or chemical probes, target key components of the ECS and are crucial for target validation, mechanistic studies, pathway elucidation, phenotypic screening, and drug discovery. These probes selectively interact with specific proteins or pathways, enabling researchers to modulate target activity and observe biological effects. When they carry an additional reporter group, they are referred to as labeled chemical probes. Developed through medicinal chemistry, structural biology, and high-throughput screening, effective chemical probes must be selective, potent, and depending on their purpose meet additional criteria such as cell permeability and metabolic stability.This chapter describes high-quality labeled and unlabeled chemical probes targeting ECS constituents that have been successfully applied for various research purposes. CB1R and CB2R, class A G protein-coupled receptors, are activated by 2-AG 1.1, AEA 1.2, and THC 3.1, with numerous ligands developed for these receptors. Imaging techniques like single-photon emission computed tomography, positron emission tomography, and fluorescently labeled CB1R and CB2R probes have enhanced CB receptor studies. CB2R activation generally results in immunosuppressive effects, limiting tissue injury. AEA 1.2 is mainly degraded by fatty acid amide hydrolase (FAAH) or N-acylethanolamine acid amidase (NAAA) into ethanolamine and arachidonic acid (AA) 1.3. FAAH inhibitors increase endogenous fatty acid amides, providing analgesic effects without adverse effects. NAAA inhibitors reduce inflammation and pain in animal models. Diacylglycerol lipase (DAGL) is essential for 2-AG 1.1 biosynthesis, while monoacylglycerol lipase (MAGL) degrades 2-AG 1.1 into AA 1.3, thus regulating cannabinoid signaling. Multiple inhibitors targeting FAAH and MAGL have been generated, though NAAA and DAGL probe development lags behind. Similarly, advancements in inhibitors targeting endocannabinoid (eCB) cellular uptake or trafficking proteins like fatty acid-binding proteins have been slower. The endocannabinoidome (eCBome) includes the ECS and related molecules and receptors, offering therapeutic opportunities from non-THC cannabinoids and eCBome mediators. Ongoing research aims to refine chemical tools for ECS and eCBome study, addressing unmet medical needs in central nervous system disorders and beyond.
Collapse
Affiliation(s)
- Annaleah Hanske
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP, Berlin, Germany
| | - Marc Nazaré
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP, Berlin, Germany
| | - Uwe Grether
- Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| |
Collapse
|
2
|
Gazzi T, Brennecke B, Olikauskas V, Hochstrasser R, Wang H, Keen Chao S, Atz K, Mostinski Y, Topp A, Heer D, Kaufmann I, Ritter M, Gobbi L, Hornsperger B, Wagner B, Richter H, O'Hara F, Wittwer MB, Jul Hansen D, Collin L, Kuhn B, Benz J, Grether U, Nazaré M. Development of a Highly Selective NanoBRET Probe to Assess MAGL Inhibition in Live Cells. Chembiochem 2024:e202400704. [PMID: 39607084 DOI: 10.1002/cbic.202400704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/23/2024] [Indexed: 11/29/2024]
Abstract
Cell-free enzymatic assays are highly useful tools in early compound profiling due to their robustness and scalability. However, their inadequacy to reflect the complexity of target engagement in a cellular environment may lead to a significantly divergent pharmacology that is eventually observed in cells. The discrepancy that emerges from properties like permeability and unspecific protein binding may largely mislead lead compound selection to undergo further chemical optimization. We report the development of a new intracellular NanoBRET assay to assess MAGL inhibition in live cells. Based on a reverse design approach, a highly potent, reversible preclinical inhibitor was conjugated to the cell-permeable BODIPY590 acceptor fluorophore while retaining its overall balanced properties. An engineered MAGL-nanoluciferase (Nluc) fusion protein provided a suitable donor counterpart for the facile interrogation of intracellular ligand activity. Validation of assay conditions using a selection of known MAGL inhibitors set the stage for the evaluation of over 1'900 MAGL drug candidates derived from our discovery program. This evaluation enabled us to select compounds for further development based not only on target engagement, but also on favorable physicochemical parameters like permeability and protein binding. This study highlights the advantages of cell-based target engagement assays for accelerating compound profiling and progress at the early stages of drug discovery programs.
Collapse
Affiliation(s)
- Thais Gazzi
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Benjamin Brennecke
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Valentas Olikauskas
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Remo Hochstrasser
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Haiyan Wang
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Suzan Keen Chao
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Kenneth Atz
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Yelena Mostinski
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Andreas Topp
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Dominik Heer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Isabelle Kaufmann
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Martin Ritter
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Luca Gobbi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Benoit Hornsperger
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Bjoern Wagner
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Hans Richter
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Fionn O'Hara
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Matthias B Wittwer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Dennis Jul Hansen
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Ludovic Collin
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Bernd Kuhn
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Joerg Benz
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Uwe Grether
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel, 4070, Switzerland
| | - Marc Nazaré
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| |
Collapse
|
3
|
Wąsińska-Kałwa M, Omran A, Mach L, Scipioni L, Bouma J, Li X, Radetzki S, Mostinski Y, Schippers M, Gazzi T, van der Horst C, Brennecke B, Hanske A, Kolomeets Y, Guba W, Sykes D, von Kries JP, Broichhagen J, Hua T, Veprintsev D, Heitman LH, Oddi S, Maccarrone M, Grether U, Nazare M. Visualization of membrane localization and the functional state of CB 2R pools using matched agonist and inverse agonist probe pairs. Chem Sci 2024:d4sc00402g. [PMID: 39430942 PMCID: PMC11485011 DOI: 10.1039/d4sc00402g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/28/2024] [Indexed: 10/22/2024] Open
Abstract
The diversity of physiological roles of the endocannabinoid system has turned it into an attractive yet elusive therapeutic target. However, chemical probes with various functionalities could pave the way for a better understanding of the endocannabinoid system at the cellular level. Notably, inverse agonists of CB2R - a key receptor of the endocannabinoid system - lagged behind despite the evidence regarding the therapeutic potential of its antagonism. Herein, we report a matched fluorescent probe pair based on a common chemotype to address and visualize both the active and inactive states of CB2R, selectively. Alongside extensive cross-validation by flow cytometry, time-lapse confocal microscopy, and super-resolution microscopy, we successfully visualize the intracellular localization of CB2R pools in live cells. The synthetic simplicity, together with the high CB2R-selectivity and specificity of our probes, turns them into valuable tools in chemical biology and drug development that can benefit the clinical translatability of CB2R-based drugs.
Collapse
Affiliation(s)
- M Wąsińska-Kałwa
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| | - A Omran
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| | - L Mach
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| | - L Scipioni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila 67100 L'Aquila Italy
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation via del Fosso di Fiorano 64 00143 Rome Italy
| | - J Bouma
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University 2333 CC Leiden The Netherlands
| | - X Li
- iHuman Institute, ShanghaiTech University Shanghai 201210 China
- School of Life Science and Technology, ShanghaiTech University Shanghai 201210 China
| | - S Radetzki
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| | - Y Mostinski
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| | - M Schippers
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd 4070 Basel Switzerland
| | - T Gazzi
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| | - C van der Horst
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University 2333 CC Leiden The Netherlands
| | - B Brennecke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| | - A Hanske
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| | - Y Kolomeets
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| | - W Guba
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd 4070 Basel Switzerland
| | - D Sykes
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham Nottingham NG7 2UH UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, University of Nottingham Midlands UK
| | - J P von Kries
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| | - J Broichhagen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| | - T Hua
- iHuman Institute, ShanghaiTech University Shanghai 201210 China
- School of Life Science and Technology, ShanghaiTech University Shanghai 201210 China
| | - D Veprintsev
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham Nottingham NG7 2UH UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, University of Nottingham Midlands UK
| | - L H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University 2333 CC Leiden The Netherlands
| | - S Oddi
- Department of Veterinary Medicine, University of Teramo Via R. Balzarini 1 64100 Teramo Italy
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation via del Fosso di Fiorano 64 00143 Rome Italy
| | - M Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila 67100 L'Aquila Italy
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation via del Fosso di Fiorano 64 00143 Rome Italy
| | - U Grether
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd 4070 Basel Switzerland
| | - M Nazare
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie FMP Campus Berlin-Buch 13125 Berlin Germany
| |
Collapse
|
4
|
Kosar M, Mach L, Carreira EM, Nazaré M, Pacher P, Grether U. Patent review of cannabinoid receptor type 2 (CB 2R) modulators (2016-present). Expert Opin Ther Pat 2024; 34:665-700. [PMID: 38886185 DOI: 10.1080/13543776.2024.2368745] [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: 02/08/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
INTRODUCTION Cannabinoid receptor type 2 (CB2R), predominantly expressed in immune tissues, is believed to play a crucial role within the body's protective mechanisms. Its modulation holds immense therapeutic promise for addressing a wide spectrum of dysbiotic conditions, including cardiovascular, gastrointestinal, liver, kidney, neurodegenerative, psychiatric, bone, skin, and autoimmune diseases, as well as lung disorders, cancer, and pain management. AREAS COVERED This review is an account of patents from 2016 up to 2023 which describes novel CB2R ligands, therapeutic applications, synthesis, as well as formulations of CB2R modulators. EXPERT OPINION The patents cover a vast, structurally diverse chemical space. The focus of CB2R ligand development has shifted from unselective dual-cannabinoid receptor type 1 (CB1R) and 2 agonists toward agonists with high selectivity over CB1R, particularly for indications associated with inflammation and tissue injury. Currently, there are at least eight CB2R agonists and one antagonist in active clinical development. A better understanding of the endocannabinoid system (ECS) and in particular of CB2R pharmacology is required to unlock the receptor's full therapeutic potential.
Collapse
Affiliation(s)
- Miroslav Kosar
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Leonard Mach
- Medicinal Chemistry, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Berlin, Berlin, Germany
| | - Erick M Carreira
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Marc Nazaré
- Medicinal Chemistry, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Berlin, Berlin, Germany
| | - Pal Pacher
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| | - Uwe Grether
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
5
|
Mach L, Omran A, Bouma J, Radetzki S, Sykes DA, Guba W, Li X, Höffelmeyer C, Hentsch A, Gazzi T, Mostinski Y, Wasinska-Kalwa M, de Molnier F, van der Horst C, von Kries JP, Vendrell M, Hua T, Veprintsev DB, Heitman LH, Grether U, Nazare M. Highly Selective Drug-Derived Fluorescent Probes for the Cannabinoid Receptor Type 1 (CB 1R). J Med Chem 2024; 67:11841-11867. [PMID: 38990855 DOI: 10.1021/acs.jmedchem.4c00465] [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: 07/13/2024]
Abstract
The cannabinoid receptor type 1 (CB1R) is pivotal within the endocannabinoid system regulating various signaling cascades with effects in appetite regulation, pain perception, memory formation, and thermoregulation. Still, understanding of CB1R's cellular signaling, distribution, and expression dynamics is very fragmentary. Real-time visualization of CB1R is crucial for addressing these questions. Selective drug-like CB1R ligands with a defined pharmacological profile were investigated for the construction of CB1R fluorescent probes using a reverse design-approach. A modular design concept with a diethyl glycine-based building block as the centerpiece allowed for the straightforward synthesis of novel probe candidates. Validated by computational docking studies, radioligand binding, and cAMP assay, this systematic approach allowed for the identification of novel pyrrole-based CB1R fluorescent probes. Application in fluorescence-based target-engagement studies and live cell imaging exemplify the great versatility of the tailored CB1R probes for investigating CB1R localization, trafficking, pharmacology, and its pathological implications.
Collapse
Affiliation(s)
- Leonard Mach
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Anahid Omran
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Jara Bouma
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University and Oncode Institute, 2333 CC Leiden, The Netherlands
| | - Silke Radetzki
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - David A Sykes
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, NG7 2UH Nottingham, U.K
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Edgbaston, B15 2TT Birmingham, Midlands, U.K
| | - Wolfgang Guba
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Xiaoting Li
- iHuman Institute, ShanghaiTech University, 201210 Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China
| | - Calvin Höffelmeyer
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Axel Hentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Thais Gazzi
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Yelena Mostinski
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | | | - Fabio de Molnier
- IRR Chemistry Hub and Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, EH16 4UU Edinburgh, U.K
| | - Cas van der Horst
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University and Oncode Institute, 2333 CC Leiden, The Netherlands
| | - Jens Peter von Kries
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Marc Vendrell
- IRR Chemistry Hub and Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, EH16 4UU Edinburgh, U.K
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, 201210 Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China
| | - Dmitry B Veprintsev
- Division of Physiology, Pharmacology & Neuroscience, School of Life Sciences, University of Nottingham, NG7 2UH Nottingham, U.K
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Edgbaston, B15 2TT Birmingham, Midlands, U.K
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University and Oncode Institute, 2333 CC Leiden, The Netherlands
| | - Uwe Grether
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Marc Nazare
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| |
Collapse
|
6
|
Stockinger F, Poc P, Möhwald A, Karch S, Häfner S, Alzheimer C, Sandoz G, Huth T, Broichhagen J. Multicolor, Cell-Impermeable, and High Affinity BACE1 Inhibitor Probes Enable Superior Endogenous Staining and Imaging of Single Molecules. J Med Chem 2024; 67:10152-10167. [PMID: 38842406 PMCID: PMC11215771 DOI: 10.1021/acs.jmedchem.4c00339] [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: 02/07/2024] [Revised: 05/10/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
The prevailing but not undisputed amyloid cascade hypothesis places the β-site of APP cleaving enzyme 1 (BACE1) center stage in Alzheimer's Disease pathogenesis. Here, we investigated functional properties of BACE1 with novel tag- and antibody-free labeling tools, which are conjugates of the BACE1-inhibitor IV (also referred to as C3) linked to different impermeable Alexa Fluor dyes. We show that these fluorescent small molecules bind specifically to BACE1, with a 1:1 labeling stoichiometry at their orthosteric site. This is a crucial property especially for single-molecule and super-resolution microscopy approaches, allowing characterization of the dyes' labeling capabilities in overexpressing cell systems and in native neuronal tissue. With multiple colors at hand, we evaluated BACE1-multimerization by Förster resonance energy transfer (FRET) acceptor-photobleaching and single-particle imaging of native BACE1. In summary, our novel fluorescent inhibitors, termed Alexa-C3, offer unprecedented insights into protein-protein interactions and diffusion behavior of BACE1 down to the single molecule level.
Collapse
Affiliation(s)
- Florian Stockinger
- Institut
für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Pascal Poc
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Heidelberg 69120, Germany
- Leibniz-Forschungsinstitut
für Molekulare Pharmakologie, Berlin 13125, Germany
| | - Alexander Möhwald
- Institut
für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Sandra Karch
- Institut
für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Stephanie Häfner
- Université
Côte d’Azur, CNRS, INSERM,
iBV, Nice 06108, Cedex 2, France
- Laboratories
of Excellence, Ion Channel Science and Therapeutics, Nice 06108, Cedex 2, France
| | - Christian Alzheimer
- Institut
für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Guillaume Sandoz
- Université
Côte d’Azur, CNRS, INSERM,
iBV, Nice 06108, Cedex 2, France
- Laboratories
of Excellence, Ion Channel Science and Therapeutics, Nice 06108, Cedex 2, France
| | - Tobias Huth
- Institut
für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Johannes Broichhagen
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Heidelberg 69120, Germany
- Leibniz-Forschungsinstitut
für Molekulare Pharmakologie, Berlin 13125, Germany
| |
Collapse
|
7
|
Kosar M, Sarott RC, Sykes DA, Viray AEG, Vitale RM, Tomašević N, Li X, Ganzoni RLZ, Kicin B, Reichert L, Patej KJ, Gómez-Bouzó U, Guba W, McCormick PJ, Hua T, Gruber CW, Veprintsev DB, Frank JA, Grether U, Carreira EM. Flipping the GPCR Switch: Structure-Based Development of Selective Cannabinoid Receptor 2 Inverse Agonists. ACS CENTRAL SCIENCE 2024; 10:956-968. [PMID: 38799662 PMCID: PMC11117691 DOI: 10.1021/acscentsci.3c01461] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 05/29/2024]
Abstract
We report a blueprint for the rational design of G protein coupled receptor (GPCR) ligands with a tailored functional response. The present study discloses the structure-based design of cannabinoid receptor type 2 (CB2R) selective inverse agonists (S)-1 and (R)-1, which were derived from privileged agonist HU-308 by introduction of a phenyl group at the gem-dimethylheptyl side chain. Epimer (R)-1 exhibits high affinity for CB2R with Kd = 39.1 nM and serves as a platform for the synthesis of a wide variety of probes. Notably, for the first time these fluorescent probes retain their inverse agonist functionality, high affinity, and selectivity for CB2R independent of linker and fluorophore substitution. Ligands (S)-1, (R)-1, and their derivatives act as inverse agonists in CB2R-mediated cAMP as well as G protein recruitment assays and do not trigger β-arrestin-receptor association. Furthermore, no receptor activation was detected in live cell ERK1/2 phosphorylation and Ca2+-release assays. Confocal fluorescence imaging experiments with (R)-7 (Alexa488) and (R)-9 (Alexa647) probes employing BV-2 microglial cells visualized CB2R expressed at endogenous levels. Finally, molecular dynamics simulations corroborate the initial docking data in which inverse agonists restrict movement of toggle switch Trp2586.48 and thereby stabilize CB2R in its inactive state.
Collapse
Affiliation(s)
- Miroslav Kosar
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Roman C. Sarott
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - David A. Sykes
- Faculty
of Medicine & Health Sciences, University
of Nottingham, Nottingham NG7 2UH, U.K.
- Centre
of Membrane Proteins and Receptors (COMPARE), University of Birmingham
and University of Nottingham, https://www.birmingham-nottingham.ac.uk/compare
| | - Alexander E. G. Viray
- Department
of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon 97239-3098, United States
| | - Rosa Maria Vitale
- Institute
of Biomolecular Chemistry, National Research
Council, Via Campi Flegrei
34, 80078 Pozzuoli, Italy
| | - Nataša Tomašević
- Center for
Physiology and Pharmacology, Medical University
of Vienna, Schwarzspanierstrasse
17, 1090 Vienna, Austria
| | - Xiaoting Li
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Rudolf L. Z. Ganzoni
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Bilal Kicin
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Lisa Reichert
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Kacper J. Patej
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Uxía Gómez-Bouzó
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Wolfgang Guba
- Roche
Pharma Research & Early Development, Roche Innovation Center Basel,
F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Peter J. McCormick
- Department
of Pharmacology and Therapeutics, University
of Liverpool, Ashton
Street, Liverpool L69 3GE, U.K.
| | - Tian Hua
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Christian W. Gruber
- Center for
Physiology and Pharmacology, Medical University
of Vienna, Schwarzspanierstrasse
17, 1090 Vienna, Austria
| | - Dmitry B. Veprintsev
- Faculty
of Medicine & Health Sciences, University
of Nottingham, Nottingham NG7 2UH, U.K.
- Centre
of Membrane Proteins and Receptors (COMPARE), University of Birmingham
and University of Nottingham, https://www.birmingham-nottingham.ac.uk/compare
| | - James A. Frank
- Department
of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon 97239-3098, United States
- Vollum
Institute, Oregon Health & Science University, Portland, Oregon 97239-3098, United States
| | - Uwe Grether
- Roche
Pharma Research & Early Development, Roche Innovation Center Basel,
F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Erick M. Carreira
- Laboratorium
für Organische Chemie, Eidgenössische
Technische Hochschule Zürich, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| |
Collapse
|
8
|
De Paus LV, An Y, Janssen APA, van den Berg RJBHN, Heitman LH, van der Stelt M. Discovery of a Photoaffinity Probe that Captures the Active Conformation of the Cannabinoid CB 2 Receptor. Chembiochem 2024; 25:e202300785. [PMID: 38372466 DOI: 10.1002/cbic.202300785] [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: 11/23/2023] [Revised: 01/26/2024] [Accepted: 02/13/2024] [Indexed: 02/20/2024]
Abstract
The cannabinoid receptor type 2 (CB2R) is a G protein-coupled receptor with therapeutic potential for the treatment of inflammatory disorders. Fluorescent probes are desirable to study its receptor localization, expression and occupancy. Previously, we have reported a photoaffinity probe LEI-121 that stabilized the inactive conformation of the CB2R. Here, we report the structure-based design of a novel bifunctional probe that captures the active conformation of the CB2R upon irradiation with light. An alkyne handle was incorporated to visualize the receptor using click-chemistry with fluorophore-azides. These probes may hold promise to study different receptor conformations in relation to their cellular localization and function.
Collapse
Affiliation(s)
- Laura V De Paus
- Molecular Physiology, Leiden University, Einsteinweg 55, Leiden, The Netherlands
| | - Yu An
- Molecular Physiology, Leiden University, Einsteinweg 55, Leiden, The Netherlands
| | - Antonius P A Janssen
- Molecular Physiology, Leiden University, Einsteinweg 55, Leiden, The Netherlands
| | | | - Laura H Heitman
- Molecular Pharmacology, Leiden University, Einsteinweg 55, Leiden, The Netherlands
| | - Mario van der Stelt
- Molecular Physiology, Leiden University, Einsteinweg 55, Leiden, The Netherlands
| |
Collapse
|
9
|
Navarro G, Sotelo E, Raïch I, Loza MI, Brea J, Majellaro M. A Robust and Efficient FRET-Based Assay for Cannabinoid Receptor Ligands Discovery. Molecules 2023; 28:8107. [PMID: 38138600 PMCID: PMC10745346 DOI: 10.3390/molecules28248107] [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: 10/31/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
The identification of new modulators for Cannabinoid Receptors (CBRs) has garnered significant attention in drug discovery over recent years, owing to their manifold pathophysiological implications. In the context of hit identification, the availability of robust and sensitive high-throughput screening assays is essential to enhance the likelihood of success. In this study, we present the development and validation of a Tag-lite® binding assay designed for screening hCB1/hCB2 binding, employing a dual fluorescent ligand, CELT-335. Representative ligands for CBRs, exhibiting diverse affinity and functional profiles, were utilized as reference compounds to validate the robustness and efficiency of the newly developed Tag-lite® binding assay protocol. The homogeneous format, coupled with the sensitivity and optimal performance of the fluorescent ligand CELT-335, establishes this assay as a viable and reliable method for screening in hit and lead identification campaigns.
Collapse
Affiliation(s)
- Gemma Navarro
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience of the University of Barcelona, 08035 Barcelona, Spain
| | - Eddy Sotelo
- Department of Organic Chemistry, Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain;
| | - Iu Raïch
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience of the University of Barcelona, 08035 Barcelona, Spain
| | - María Isabel Loza
- Research Center in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain;
| | - Jose Brea
- Research Center in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain;
| | - Maria Majellaro
- Celtarys Research SL, Avda. Mestre Mateo, 2, 15706 Santiago de Compostela, Spain
| |
Collapse
|
10
|
Maccarrone M, Di Marzo V, Gertsch J, Grether U, Howlett AC, Hua T, Makriyannis A, Piomelli D, Ueda N, van der Stelt M. Goods and Bads of the Endocannabinoid System as a Therapeutic Target: Lessons Learned after 30 Years. Pharmacol Rev 2023; 75:885-958. [PMID: 37164640 PMCID: PMC10441647 DOI: 10.1124/pharmrev.122.000600] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/12/2023] Open
Abstract
The cannabis derivative marijuana is the most widely used recreational drug in the Western world and is consumed by an estimated 83 million individuals (∼3% of the world population). In recent years, there has been a marked transformation in society regarding the risk perception of cannabis, driven by its legalization and medical use in many states in the United States and worldwide. Compelling research evidence and the Food and Drug Administration cannabis-derived cannabidiol approval for severe childhood epilepsy have confirmed the large therapeutic potential of cannabidiol itself, Δ9-tetrahydrocannabinol and other plant-derived cannabinoids (phytocannabinoids). Of note, our body has a complex endocannabinoid system (ECS)-made of receptors, metabolic enzymes, and transporters-that is also regulated by phytocannabinoids. The first endocannabinoid to be discovered 30 years ago was anandamide (N-arachidonoyl-ethanolamine); since then, distinct elements of the ECS have been the target of drug design programs aimed at curing (or at least slowing down) a number of human diseases, both in the central nervous system and at the periphery. Here a critical review of our knowledge of the goods and bads of the ECS as a therapeutic target is presented to define the benefits of ECS-active phytocannabinoids and ECS-oriented synthetic drugs for human health. SIGNIFICANCE STATEMENT: The endocannabinoid system plays important roles virtually everywhere in our body and is either involved in mediating key processes of central and peripheral diseases or represents a therapeutic target for treatment. Therefore, understanding the structure, function, and pharmacology of the components of this complex system, and in particular of key receptors (like cannabinoid receptors 1 and 2) and metabolic enzymes (like fatty acid amide hydrolase and monoacylglycerol lipase), will advance our understanding of endocannabinoid signaling and activity at molecular, cellular, and system levels, providing new opportunities to treat patients.
Collapse
Affiliation(s)
- Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Vincenzo Di Marzo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Jürg Gertsch
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Uwe Grether
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Allyn C Howlett
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Tian Hua
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Alexandros Makriyannis
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Daniele Piomelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Natsuo Ueda
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Mario van der Stelt
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| |
Collapse
|
11
|
Bhattacharjee P, Iyer MR. Rational Design, Synthesis, and Evaluation of Fluorescent CB 2 Receptor Ligands for Live-Cell Imaging: A Comprehensive Review. Pharmaceuticals (Basel) 2023; 16:1235. [PMID: 37765043 PMCID: PMC10534640 DOI: 10.3390/ph16091235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
The cannabinoid receptors CB1 and CB2 are class A G protein-coupled receptors (GPCRs) that are activated via endogenous lipids called endocannabinoids. The endocannabinoid system (ECS) plays a critical role in the regulation of several physiological states and a wide range of diseases. In recent years, drug discovery approaches targeting the cannabinoid type 2 receptor (CB2R) have gained prominence. Particular attention has been given to selective agonists targeting the CB2 receptors to circumvent the neuropsychotropic side effects associated with CB1 receptors. The pharmacological modulation of CB2R holds therapeutic promise for various diseases, such as inflammatory disorders and immunological conditions, as well as pain management and cancer treatment. Recently, the utilization of fluorescent probes has emerged as a valuable technique for investigating the interactions between ligands and proteins at an exceptional level of spatial and temporal precision. In this review, we aim to examine the progress made in the development of fluorescent probes targeting CB2 receptors and highlight their significance in facilitating the successful clinical translation of CB2R-based therapies.
Collapse
Affiliation(s)
| | - Malliga R. Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, MD 20852, USA
| |
Collapse
|
12
|
Scipioni L, Tortolani D, Ciaramellano F, Fanti F, Gazzi T, Sergi M, Nazaré M, Oddi S, Maccarrone M. Aβ Chronic Exposure Promotes an Activation State of Microglia through Endocannabinoid Signalling Imbalance. Int J Mol Sci 2023; 24:ijms24076684. [PMID: 37047663 PMCID: PMC10095368 DOI: 10.3390/ijms24076684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/12/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Dysfunctional phenotype of microglia, the primary brain immune cells, may aggravate Alzheimer’s disease (AD) pathogenesis by releasing proinflammatory factors, such as nitric oxide (NO). The endocannabinoids N-arachidonoylethanolamine (AEA) and 2-arachidonoylglycerol (2-AG) are bioactive lipids increasingly recognised for their essential roles in regulating microglial activity both under normal and AD-driven pathological conditions. To investigate the possible impact of chronic exposure to β-amyloid peptides (Aβ) on the microglial endocannabinoid signalling, we characterised the functional expression of the endocannabinoid system on neonatal microglia isolated from wild-type and Tg2576 mice, an AD-like model, which overexpresses Aβ peptides in the developing brain. We found that Aβ-exposed microglia produced 2-fold more 2-AG than normal microglia. Accordingly, the expression levels of diacylglycerol lipase-α (DAGLα) and monoacylglycerol lipase (MAGL), the main enzymes responsible for synthesising and hydrolysing 2-AG, respectively, were consistently modified in Tg2576 microglia. Furthermore, compared to wild-type cells, transgenic microglia basally showed increased expression of the cannabinoid 2 receptor, typically upregulated in an activated proinflammatory phenotype. Indeed, following inflammatory stimulus, Aβ-exposed microglia displayed an enhanced production of NO, which was abolished by pharmacological inhibition of DAGLα. These findings suggested that exposure to Aβ polarises microglial cells towards a pro-AD phenotype, possibly by enhancing 2-AG signalling.
Collapse
Affiliation(s)
- Lucia Scipioni
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio Snc, 67100 L’Aquila, Italy
- European Center for Brain Research-IRCCS Santa Lucia Foundation (FSL), Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Daniel Tortolani
- European Center for Brain Research-IRCCS Santa Lucia Foundation (FSL), Via del Fosso di Fiorano 64, 00143 Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy
| | - Francesca Ciaramellano
- European Center for Brain Research-IRCCS Santa Lucia Foundation (FSL), Via del Fosso di Fiorano 64, 00143 Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy
| | - Federico Fanti
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy
| | - Thais Gazzi
- Leibniz Research Institute for Molecular Pharmacology (FMP), Campus Berlin-Buch, 13125 Berlin, Germany
| | - Manuel Sergi
- Department of Chemistry, Sapienza University of Rome, 00185 Rome, Italy
| | - Marc Nazaré
- Leibniz Research Institute for Molecular Pharmacology (FMP), Campus Berlin-Buch, 13125 Berlin, Germany
| | - Sergio Oddi
- European Center for Brain Research-IRCCS Santa Lucia Foundation (FSL), Via del Fosso di Fiorano 64, 00143 Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio Snc, 67100 L’Aquila, Italy
- European Center for Brain Research-IRCCS Santa Lucia Foundation (FSL), Via del Fosso di Fiorano 64, 00143 Rome, Italy
| |
Collapse
|
13
|
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]
|
14
|
Zimmerberg J, Soubias O, Pastor RW. Special issue for Klaus Gawrisch. Biophys J 2023; 122:E1-E8. [PMID: 36921597 PMCID: PMC10111273 DOI: 10.1016/j.bpj.2023.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 03/17/2023] Open
Affiliation(s)
- Joshua Zimmerberg
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Olivier Soubias
- Macromolecular NMR Section, Center for Structural Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Richard W Pastor
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
15
|
Deciphering Complex Interactions in Bioactive Lipid Signaling. Molecules 2023; 28:molecules28062622. [PMID: 36985594 PMCID: PMC10057854 DOI: 10.3390/molecules28062622] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Lipids are usually viewed as metabolic fuel and structural membrane components. Yet, in recent years, different families of lipids able to act as authentic messengers between cells and/or intracellularly have been discovered. Such lipid signals have been shown to exert their biological activity via specific receptors that, by triggering distinct signal transduction pathways, regulate manifold pathophysiological processes in our body. Here, endogenous bioactive lipids produced from arachidonic acid (AA) and other poly-unsaturated fatty acids will be presented, in order to put into better perspective the relevance of their mutual interactions for health and disease conditions. To this end, metabolism and signal transduction pathways of classical eicosanoids, endocannabinoids and specialized pro-resolving mediators will be described, and the intersections and commonalities of their metabolic enzymes and binding receptors will be discussed. Moreover, the interactions of AA-derived signals with other bioactive lipids such as shingosine-1-phosphate and steroid hormones will be addressed.
Collapse
|
16
|
Punt J, van der Vliet D, van der Stelt M. Chemical Probes to Control and Visualize Lipid Metabolism in the Brain. Acc Chem Res 2022; 55:3205-3217. [PMID: 36283077 PMCID: PMC9670861 DOI: 10.1021/acs.accounts.2c00521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Signaling lipids, such as the endocannabinoids, play an important role in the brain. They regulate synaptic transmission and control various neurophysiological processes, including pain sensation, appetite, memory formation, stress, and anxiety. Unlike classical neurotransmitters, lipid messengers are produced on demand and degraded by metabolic enzymes to control their lifespan and signaling actions. Chemical biology approaches have become one of the main driving forces to study and unravel the physiological role of lipid messengers in the brain. Here, we review how the development and use of chemical probes has allowed one to study endocannabinoid signaling by (i) inhibiting the biosynthetic and metabolic enzymes; (ii) visualizing the activity of these enzymes; and (iii) controlling the release and transport of the endocannabinoids. Activity-based probes were instrumental to guide the discovery of highly selective and in vivo active inhibitors of the biosynthetic (DAGL, NAPE-PLD) and metabolic (MAGL, FAAH) enzymes of endocannabinoids. These inhibitors allowed one to study the role of these enzymes in animal models of disease. For instance, the DAGL-MAGL axis was shown to control neuroinflammation and the NAPE-PLD-FAAH axis to regulate emotional behavior. Activity-based protein profiling and chemical proteomics were essential to guide the drug discovery and development of compounds targeting MAGL and FAAH, such as ABX-1431 (Lu AG06466) and PF-04457845, respectively. These experimental drugs are now in clinical trials for multiple indications, including multiple sclerosis and post-traumatic stress disorders. Activity-based probes have also been used to visualize the activity of these lipid metabolizing enzymes with high spatial resolution in brain slices, thereby showing the cell type-specific activity of these lipid metabolizing enzymes. The transport, release, and uptake of signaling lipids themselves cannot, however, be captured by activity-based probes in a spatiotemporal controlled manner. Therefore, bio-orthogonal lipids equipped with photoreactive, photoswitchable groups or photocages have been developed. These chemical probes were employed to investigate the protein interaction partners of the endocannabinoids, such as putative membrane transporters, as well as to study the functional cellular responses within milliseconds upon irradiation. Finally, genetically encoded sensors have recently been developed to monitor the real-time release of endocannabinoids with high spatiotemporal resolution in cultured neurons, acute brain slices, and in vivo mouse models. It is anticipated that the combination of chemical probes, highly selective inhibitors, and sensors with advanced (super resolution) imaging modalities, such as PharmacoSTORM and correlative light-electron microscopy, will uncover the fundamental basis of lipid signaling at nanoscale resolution in the brain. Furthermore, chemical biology approaches enable the translation of these fundamental discoveries into clinical solutions for brain diseases with aberrant lipid signaling.
Collapse
|