1
|
Luo Y, Sun L, Peng Y. The structural basis of the G protein-coupled receptor and ion channel axis. Curr Res Struct Biol 2025; 9:100165. [PMID: 40083915 PMCID: PMC11904507 DOI: 10.1016/j.crstbi.2025.100165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/25/2025] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
Sensory neurons play an essential role in recognizing and responding to detrimental, irritating, and inflammatory stimuli from our surroundings, such as pain, itch, cough, and neurogenic inflammation. The transduction of these physiological signals is chiefly mediated by G protein-coupled receptors (GPCRs) and ion channels. The binding of ligands to GPCRs triggers a signaling cascade, recruiting G proteins or β-arrestins, which subsequently interact with ion channels (e.g., GIRK and TRP channels). This interaction leads to the sensitization and activation of these channels, initiating the neuron's protective mechanisms. This review delves into the complex interplay between GPCRs and ion channels that underpin these physiological processes, with a particular focus on the role of structural biology in enhancing our comprehension. Through unraveling the intricacies of the GPCR-ion channel axis, we aim to shed light on the sophisticated intermolecular dynamics within these pivotal membrane protein families, ultimately guiding the development of precise therapeutic interventions.
Collapse
Affiliation(s)
- Yulin Luo
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, L Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Liping Sun
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Yao Peng
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| |
Collapse
|
2
|
Villaescusa L, Azcune L, Landa A, Oiarbide M. Catalytic Asymmetric Synthesis of Bicyclic Isothioureas from a Common Enolizable Template. J Org Chem 2024; 89:15607-15622. [PMID: 39418573 DOI: 10.1021/acs.joc.4c01686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
While bicyclic isothiourea (ITU) moieties are found in biologically active molecules and organocatalysts, a catalytic asymmetric synthesis of ITUs is pending. Here, we report the catalytic, enantioselective functionalization of enolizable template II with a variety of electrophiles, including unsaturated ketones, esters, sulfones, nitro compounds, and thiolating reagents, as an entry to enantioenriched bicyclic ITUs III. Scope and limitations are shown, as well as examples of product derivatization.
Collapse
Affiliation(s)
- Leire Villaescusa
- Department of Organic Chemistry I, Faculty of Chemistry, University of the Basque Country UPV/EHU, Manuel Lardizabal Pasealekua 3, Donostia-San Sebastian 20018, Spain
| | - Laura Azcune
- Department of Organic Chemistry I, Faculty of Chemistry, University of the Basque Country UPV/EHU, Manuel Lardizabal Pasealekua 3, Donostia-San Sebastian 20018, Spain
| | - Aitor Landa
- Department of Organic Chemistry I, Faculty of Chemistry, University of the Basque Country UPV/EHU, Manuel Lardizabal Pasealekua 3, Donostia-San Sebastian 20018, Spain
| | - Mikel Oiarbide
- Department of Organic Chemistry I, Faculty of Chemistry, University of the Basque Country UPV/EHU, Manuel Lardizabal Pasealekua 3, Donostia-San Sebastian 20018, Spain
| |
Collapse
|
3
|
Hovah ME, Holzgrabe U. Bivalent and bitopic ligands of the opioid receptors: The prospects of a dual approach. Med Res Rev 2024; 44:2545-2599. [PMID: 38751227 DOI: 10.1002/med.22050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 04/09/2024] [Accepted: 05/05/2024] [Indexed: 10/05/2024]
Abstract
Opioid receptors belonging to the class A G-protein coupled receptors (GPCRs) are the targets of choice in the treatment of acute and chronic pain. However, their on-target side effects such as respiratory depression, tolerance and addiction have led to the advent of the 'opioid crisis'. In the search for safer analgesics, bivalent and more recently, bitopic ligands have emerged as valuable tool compounds to probe these receptors. The activity of bivalent and bitopic ligands rely greatly on the allosteric nature of the GPCRs. Bivalent ligands consist of two pharmacophores, each binding to the individual orthosteric binding site (OBS) of the monomers within a dimer. Bitopic or dualsteric ligands bridge the gap between the OBS and the spatially distinct, less conserved allosteric binding site (ABS) through the simultaneous occupation of these two sites. Bivalent and bitopic ligands stabilize distinct conformations of the receptors which ultimately translates into unique signalling and pharmacological profiles. Some of the interesting properties shown by these ligands include improved affinity and/or efficacy, subtype and/or functional selectivity and reduced side effects. This review aims at providing an overview of some of the bivalent and bitopic ligands of the opioid receptors and, their pharmacology in the hope of inspiring the design and discovery of the next generation of opioid analgesics.
Collapse
Affiliation(s)
- Marie Emilie Hovah
- Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany
| |
Collapse
|
4
|
Payne CM, Baltos JA, Langiu M, Sinh Lu C, Tyndall JDA, Gregory KJ, May LT, Vernall AJ. Development of Putative Bivalent Dicovalent Ligands for the Adenosine A1 Receptor. Chembiochem 2024; 25:e202400242. [PMID: 38777792 DOI: 10.1002/cbic.202400242] [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: 03/15/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/25/2024]
Abstract
Accumulating evidence suggests that G protein-coupled receptors (GPCRs) can exist and function in homodimer and heterodimer forms. The adenosine A1 receptor (A1R) has been shown to form both homodimers and heterodimers, but there is a lack of chemical tools to study these dimeric receptor populations. This work describes the synthesis and pharmacological evaluation of a novel class of bivalent GPCR chemical tools, where each ligand moiety of the bivalent compound contains a sulfonyl fluoride covalent warhead designed to be capable of simultaneously reacting with each A1R of an A1R homodimer. The novel compounds were characterised using radioligand binding assays, including washout assays, and functionally in cAMP assays. The bivalent dicovalent compounds were competitive A1R antagonists and showed evidence of covalent binding and simultaneous binding across an A1R homodimer. Greater selectivity for A1R over the adenosine A3 receptor was observed for bivalent dicovalent over the equivalent monovalent compounds, indicating subtype selectivity can be achieved with dual occupation by a bivalent dicovalent ligand.
Collapse
Affiliation(s)
- China M Payne
- Department of Chemistry, University of Otago, Dunedin, 9016, New Zealand
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| | - Monica Langiu
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| | - Cam Sinh Lu
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| | - Joel D A Tyndall
- School of Pharmacy, University of Otago, Dunedin, 9054, New Zealand
| | - Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash University, Parkville, VIC, 3052, Australia
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| | - Andrea J Vernall
- Department of Chemistry, University of Otago, Dunedin, 9016, New Zealand
| |
Collapse
|
5
|
Llinas Del Torrent C, Raïch I, Gonzalez A, Lillo J, Casajuana-Martin N, Franco R, Pardo L, Navarro G. Allosterism in the adenosine A 2A and cannabinoid CB 2 heteromer. Br J Pharmacol 2024. [PMID: 39044481 DOI: 10.1111/bph.16502] [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: 03/03/2023] [Revised: 01/24/2024] [Accepted: 03/18/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND AND PURPOSE Allosterism is a regulatory mechanism for GPCRs that can be attained by ligand-binding or protein-protein interactions with another GPCR. We have studied the influence of the dimer interface on the allosteric properties of the A2A receptor and CB2 receptor heteromer. EXPERIMENTAL APPROACH We have evaluated cAMP production, phosphorylation of signal-regulated kinases (pERK1/2), label-free dynamic mass redistribution, β-arrestin 2 recruitment and bimolecular fluorescence complementation assays in the absence and presence of synthetic peptides that disrupt the formation of the heteromer. Molecular dynamic simulations provided converging evidence that the heteromeric interface influences the allosteric properties of the A2AR-CB2R heteromer. KEY RESULTS Apo A2AR blocks agonist-induced signalling of CB2R. The disruptive peptides, with the amino acid sequence of transmembrane (TM) 6 of A2AR or CB2R, facilitate CB2R activation, suggesting that A2AR allosterically prevents the outward movement of TM 6 of CB2R for G protein binding. Significantly, binding of the selective antagonist SCH 58261 to A2AR also facilitated agonist-induced activation of CB2R. CONCLUSIONS AND IMPLICATIONS It is proposed that the A2AR-CB2R heteromer contains distinct dimerization interfaces that govern its functional properties. The molecular interface between protomers of the A2AR-CB2R heteromer interconverted from TM 6 for apo or agonist-bound A2AR, blocking CB2R activation, to mainly the TM 1/7 interface for antagonist-bound A2AR, facilitating the independent opening of intracellular cavities for G protein binding. These novel results shed light on a different type of allosteric mechanism and extend the repertoire of GPCR heteromer signalling.
Collapse
Affiliation(s)
- Claudia Llinas Del Torrent
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Iu Raïch
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, University of Barcelona (NeuroUB), Barcelona, Spain
| | - Angel Gonzalez
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Jaume Lillo
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Nil Casajuana-Martin
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Rafael Franco
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Leonardo Pardo
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Gemma Navarro
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Neuroscience, University of Barcelona (NeuroUB), Barcelona, Spain
| |
Collapse
|
6
|
Nagl M, Mönnich D, Rosier N, Schihada H, Sirbu A, Konar N, Reyes-Resina I, Navarro G, Franco R, Kolb P, Annibale P, Pockes S. Fluorescent Tools for the Imaging of Dopamine D 2 -Like Receptors. Chembiochem 2024; 25:e202300659. [PMID: 37942961 DOI: 10.1002/cbic.202300659] [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: 09/26/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/10/2023]
Abstract
The family of dopamine D2 -like receptors represents an interesting target for a variety of neurological diseases, e. g. Parkinson's disease (PD), addiction, or schizophrenia. In this study we describe the synthesis of a new set of fluorescent ligands as tools for visualization of dopamine D2 -like receptors. Pharmacological characterization in radioligand binding studies identified UR-MN212 (20) as a high-affinity ligand for D2 -like receptors (pKi (D2long R)=8.24, pKi (D3 R)=8.58, pKi (D4 R)=7.78) with decent selectivity towards D1 -like receptors. Compound 20 is a neutral antagonist in a Go1 activation assay at the D2long R, D3 R, and D4 R, which is an important feature for studies using whole cells. The neutral antagonist 20, equipped with a 5-TAMRA dye, displayed rapid association to the D2long R in binding studies using confocal microscopy demonstrating its suitability for fluorescence microscopy. Furthermore, in molecular brightness studies, the ligand's binding affinity could be determined in a single-digit nanomolar range that was in good agreement with radioligand binding data. Therefore, the fluorescent compound can be used for quantitative characterization of native D2 -like receptors in a broad variety of experimental setups.
Collapse
Affiliation(s)
- Martin Nagl
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Denise Mönnich
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Niklas Rosier
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35037, Marburg, Germany
| | - Alexei Sirbu
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany
| | - Nergis Konar
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany
| | - Irene Reyes-Resina
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Rafael Franco
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35037, Marburg, Germany
| | - Paolo Annibale
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany
- School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, Scotland
| | - Steffen Pockes
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN, 55414, USA
| |
Collapse
|
7
|
Qian M, Sun Z, Chen X, Van Calenbergh S. Study of G protein-coupled receptors dimerization: From bivalent ligands to drug-like small molecules. Bioorg Chem 2023; 140:106809. [PMID: 37651896 DOI: 10.1016/j.bioorg.2023.106809] [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: 04/12/2023] [Revised: 07/27/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023]
Abstract
In the past decades an increasing number of studies revealed that G protein-coupled receptors (GPCRs) are capable of forming dimers or even higher-ordered oligomers, which may modulate receptor function and act as potential drug targets. In this review, we briefly summarized the design strategy of bivalent GPCR ligands and mainly focused on how to use them to study and/or detect GPCP dimerization in vitro and in vivo. Bivalent ligands show specific properties relative to their corresponding monomeric ligands because they are able to bind to GPCR homodimers or heterodimers simultaneously. For example, bivalent ligands with optimal length of spacers often exhibited higher binding affinities for dimers compared to that of monomers. Furthermore, bivalent ligands displayed specific signal transduction compared to monovalent ligands. Finally, we give our perspective on targeting GPCR dimers from traditional bivalent ligands to more drug-like small molecules.
Collapse
Affiliation(s)
- Mingcheng Qian
- School of Pharmacy, Changzhou University, Changzhou 213164, Jiangsu, China; Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium.
| | - Zhengyang Sun
- School of Pharmacy, Changzhou University, Changzhou 213164, Jiangsu, China
| | - Xin Chen
- School of Pharmacy, Changzhou University, Changzhou 213164, Jiangsu, China
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium.
| |
Collapse
|
8
|
Bueschbell B, Magalhães PR, Barreto CA, Melo R, Schiedel AC, Machuqueiro M, Moreira IS. The World of GPCR dimers - Mapping dopamine receptor D 2 homodimers in different activation states and configuration arrangements. Comput Struct Biotechnol J 2023; 21:4336-4353. [PMID: 37711187 PMCID: PMC10497915 DOI: 10.1016/j.csbj.2023.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/29/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are known to dimerize, but the molecular and structural basis of GPCR dimers is not well understood. In this study, we developed a computational framework to generate models of symmetric and asymmetric GPCR dimers using different monomer activation states and identified their most likely interfaces with molecular details. We chose the dopamine receptor D2 (D2R) homodimer as a case study because of its biological relevance and the availability of structural information. Our results showed that transmembrane domains 4 and 5 (TM4 and TM5) are mostly found at the dimer interface of the D2R dimer and that these interfaces have a subset of key residues that are mostly nonpolar from TM4 and TM5, which was in line with experimental studies. In addition, TM2 and TM3 appear to be relevant for D2R dimers. In some cases, the inactive configuration is unaffected by the partnered protomer, whereas in others, the active protomer adopts the properties of an inactive receptor. Additionally, the β-arrestin configuration displayed the properties of an active receptor in the absence of an agonist, suggesting that a switch to another meta-state during dimerization occurred. Our findings are consistent with the experimental data, and this method can be adapted to study heterodimers and potentially extended to include additional proteins such as G proteins or β-arrestins. In summary, this approach provides insight into the impact of the conformational status of partnered protomers on the overall quaternary GPCR macromolecular structure and dynamics.
Collapse
Affiliation(s)
- Beatriz Bueschbell
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-456 Coimbra, Portugal
- IIIs-Institute for Interdisciplinary Research, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Pedro R. Magalhães
- BioISI - Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande C8 bdg, 1749-016 Lisboa, Portugal
| | - Carlos A.V. Barreto
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-456 Coimbra, Portugal
- IIIs-Institute for Interdisciplinary Research, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Rita Melo
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-456 Coimbra, Portugal
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, University of Coimbra, Coimbra, Portugal
| | - Anke C. Schiedel
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn, D-53121 Bonn, Germany
| | - Miguel Machuqueiro
- BioISI - Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande C8 bdg, 1749-016 Lisboa, Portugal
| | - Irina S. Moreira
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
9
|
Cervetto C, Maura G, Guidolin D, Amato S, Ceccoli C, Agnati LF, Marcoli M. Striatal astrocytic A2A-D2 receptor-receptor interactions and their role in neuropsychiatric disorders. Neuropharmacology 2023:109636. [PMID: 37321323 DOI: 10.1016/j.neuropharm.2023.109636] [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: 03/30/2023] [Revised: 05/26/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
It is now generally accepted that astrocytes are active players in synaptic transmission, so that a neurocentric perspective of the integrative signal communication in the central nervous system is shifting towards a neuro-astrocentric perspective. Astrocytes respond to synaptic activity, release chemical signals (gliotransmitters) and express neurotransmitter receptors (G protein-coupled and ionotropic receptors), thus behaving as co-actors with neurons in signal communication in the central nervous system. The ability of G protein-coupled receptors to physically interact through heteromerization, forming heteromers and receptor mosaics with new distinct signal recognition and transduction pathways, has been intensively studied at neuronal plasma membrane, and has changed the view of the integrative signal communication in the central nervous system. One of the best-known examples of receptor-receptor interaction through heteromerization, with relevant consequences for both the physiological and the pharmacological points of view, is given by adenosine A2A and dopamine D2 receptors on the plasma membrane of striatal neurons. Here we review evidence that native A2A and D2 receptors can interact through heteromerization at the plasma membrane of astrocytes as well. Astrocytic A2A-D2 heteromers were found able to control the release of glutamate from the striatal astrocyte processes. A2A-D2 heteromers on striatal astrocytes and astrocyte processes are discussed as far as their potential relevance in the control of glutamatergic transmission in striatum is concerned, including potential roles in glutamatergic transmission dysregulation in pathological conditions including schizophrenia or the Parkinson's disease.
Collapse
Affiliation(s)
- Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy; Center for Promotion of 3Rs in Teaching and Research (Centro 3R), Pisa, Italy.
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Italy.
| | - Sarah Amato
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Cristina Ceccoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Luigi F Agnati
- Department of Biochemical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, Modena, Italy.
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy; Center for Promotion of 3Rs in Teaching and Research (Centro 3R), Pisa, Italy; Center of Excellence for Biomedical Research, University of Genova, Italy.
| |
Collapse
|
10
|
Juza R, Musilek K, Mezeiova E, Soukup O, Korabecny J. Recent advances in dopamine D 2 receptor ligands in the treatment of neuropsychiatric disorders. Med Res Rev 2023; 43:55-211. [PMID: 36111795 DOI: 10.1002/med.21923] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 02/04/2023]
Abstract
Dopamine is a biologically active amine synthesized in the central and peripheral nervous system. This biogenic monoamine acts by activating five types of dopamine receptors (D1-5 Rs), which belong to the G protein-coupled receptor family. Antagonists and partial agonists of D2 Rs are used to treat schizophrenia, Parkinson's disease, depression, and anxiety. The typical pharmacophore with high D2 R affinity comprises four main areas, namely aromatic moiety, cyclic amine, central linker and aromatic/heteroaromatic lipophilic fragment. From the literature reviewed herein, we can conclude that 4-(2,3-dichlorophenyl), 4-(2-methoxyphenyl)-, 4-(benzo[b]thiophen-4-yl)-1-substituted piperazine, and 4-(6-fluorobenzo[d]isoxazol-3-yl)piperidine moieties are critical for high D2 R affinity. Four to six atoms chains are optimal for D2 R affinity with 4-butoxyl as the most pronounced one. The bicyclic aromatic/heteroaromatic systems are most frequently occurring as lipophilic appendages to retain high D2 R affinity. In this review, we provide a thorough overview of the therapeutic potential of D2 R modulators in the treatment of the aforementioned disorders. In addition, this review summarizes current knowledge about these diseases, with a focus on the dopaminergic pathway underlying these pathologies. Major attention is paid to the structure, function, and pharmacology of novel D2 R ligands, which have been developed in the last decade (2010-2021), and belong to the 1,4-disubstituted aromatic cyclic amine group. Due to the abundance of data, allosteric D2 R ligands and D2 R modulators from patents are not discussed in this review.
Collapse
Affiliation(s)
- Radomir Juza
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Musilek
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Eva Mezeiova
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
11
|
Gregory KJ, Jörg M. Chemical biology-based approaches to study adenosine A 2A - dopamine D 2 receptor heteromers. Purinergic Signal 2022; 18:395-398. [PMID: 35348986 PMCID: PMC9832194 DOI: 10.1007/s11302-022-09860-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/15/2022] [Indexed: 01/14/2023] Open
Affiliation(s)
- Karen J. Gregory
- grid.1002.30000 0004 1936 7857Drug Discovery Biology Theme, ARC Centre for Cryo-Electron Microscopy of Membrane Proteins and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052 Australia
| | - Manuela Jörg
- Medicinal Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia. .,Chemistry-School of Natural & Environmental Sciences, Newcastle University Centre for Cancer, Newcastle University, Bedson Building, Newcastle Upon Tyne, NE1 7RU, UK.
| |
Collapse
|
12
|
Ceradini D, Cacivkins P, Ramos-Llorca A, Shubin K. Improved Synthesis of the Selected Serine Protease uPA Inhibitor UAMC-00050, a Lead Compound for the Treatment of Dry Eye Disease. Org Process Res Dev 2022; 26:2937-2946. [PMID: 36311379 PMCID: PMC9594321 DOI: 10.1021/acs.oprd.2c00244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Indexed: 11/29/2022]
Abstract
![]()
The α-aminophosphonate UAMC-00050, a newly developed
trypsin-like
serine protease inhibitor, is a lead compound for the treatment of
dry eye syndrome and ocular inflammation. The medicinal chemistry
route developed at the University of Antwerp possessed several problems
hampering the scale-up such as poor yields for some of the steps,
hazardous reagents, and environmental footprint. Herein, we report
an optimized route for the UAMC-00050, in which environmental unfriendly
solvents were excluded, hazardous reagents were replaced with safer
alternatives, and are more efficient in terms of atom economy. Every
reaction step was optimized to reach a higher yield, and design of
experiment was used to find the optimum conditions in the last step.
Furthermore, all the flash chromatography purifications of intermediates
were replaced with plug filtration, slurry purifications, or crystallization.
The overall yield was increased from 3% in the medicinal chemistry
route to 22% in the process development route.
Collapse
Affiliation(s)
- Davide Ceradini
- Latvian Institute of Organic Synthesis, Aizkraukle Iela 21, Riga LV-1006, Latvia
| | - Pavel Cacivkins
- Exponential Technologies Ltd., Dze̅rbenes iela 14, Riga LV-1006, Latvia
| | | | - Kirill Shubin
- Latvian Institute of Organic Synthesis, Aizkraukle Iela 21, Riga LV-1006, Latvia
| |
Collapse
|
13
|
Ferré S, Ciruela F, Dessauer CW, González-Maeso J, Hébert TE, Jockers R, Logothetis DE, Pardo L. G protein-coupled receptor-effector macromolecular membrane assemblies (GEMMAs). Pharmacol Ther 2022; 231:107977. [PMID: 34480967 PMCID: PMC9375844 DOI: 10.1016/j.pharmthera.2021.107977] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest group of receptors involved in cellular signaling across the plasma membrane and a major class of drug targets. The canonical model for GPCR signaling involves three components - the GPCR, a heterotrimeric G protein and a proximal plasma membrane effector - that have been generally thought to be freely mobile molecules able to interact by 'collision coupling'. Here, we synthesize evidence that supports the existence of GPCR-effector macromolecular membrane assemblies (GEMMAs) comprised of specific GPCRs, G proteins, plasma membrane effector molecules and other associated transmembrane proteins that are pre-assembled prior to receptor activation by agonists, which then leads to subsequent rearrangement of the GEMMA components. The GEMMA concept offers an alternative and complementary model to the canonical collision-coupling model, allowing more efficient interactions between specific signaling components, as well as the integration of the concept of GPCR oligomerization as well as GPCR interactions with orphan receptors, truncated GPCRs and other membrane-localized GPCR-associated proteins. Collision-coupling and pre-assembled mechanisms are not exclusive and likely both operate in the cell, providing a spectrum of signaling modalities which explains the differential properties of a multitude of GPCRs in their different cellular environments. Here, we explore the unique pharmacological characteristics of individual GEMMAs, which could provide new opportunities to therapeutically modulate GPCR signaling.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Addiction, Intramural Research Program, NIH, DHHS, Baltimore, MD, USA.
| | - Francisco Ciruela
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, IDIBELL, University of Barcelona, L’Hospitalet de Llobregat, Spain
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Javier González-Maeso
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Terence E. Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec
| | - Ralf Jockers
- University of Paris, Institute Cochin, INSERM, CNRS, Paris, France
| | - Diomedes E. Logothetis
- Laboratory of Electrophysiology, Departments of Pharmaceutical Sciences, Chemistry and Chemical Biology and Center for Drug Discovery, School of Pharmacy at the Bouvé College of Health Sciences and College of Science, Northeastern University, Boston, Massachusetts, USA
| | - Leonardo Pardo
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Autonomous University of Barcelona, Bellaterra, Spain
| |
Collapse
|
14
|
Pulido D, Casadó-Anguera V, Gómez-Autet M, Llopart N, Moreno E, Casajuana-Martin N, Ferré S, Pardo L, Casadó V, Royo M. Heterobivalent Ligand for the Adenosine A 2A-Dopamine D 2 Receptor Heteromer. J Med Chem 2022; 65:616-632. [PMID: 34982555 PMCID: PMC11915710 DOI: 10.1021/acs.jmedchem.1c01763] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A G protein-coupled receptor heteromer that fulfills the established criteria for its existence in vivo is the complex between adenosine A2A (A2AR) and dopamine D2 (D2R) receptors. Here, we have designed and synthesized heterobivalent ligands for the A2AR-D2R heteromer with various spacer lengths. The indispensable simultaneous binding of these ligands to the two different orthosteric sites of the heteromer has been evaluated by radioligand competition-binding assays in the absence and presence of specific peptides that disrupt the formation of the heteromer, label-free dynamic mass redistribution assays in living cells, and molecular dynamic simulations. This combination of techniques has permitted us to identify compound 26 [KDB1 (A2AR) = 2.1 nM, KDB1 (D2R) = 0.13 nM], with a spacer length of 43-atoms, as a true bivalent ligand that simultaneously binds to the two different orthosteric sites. Moreover, bioluminescence resonance energy transfer experiments indicate that 26 favors the stabilization of the A2AR-D2R heteromer.
Collapse
Affiliation(s)
- Daniel Pulido
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08034 Barcelona, Spain
- Department of Surfactants and Nanobiotechnology, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - Verònica Casadó-Anguera
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
| | - Marc Gómez-Autet
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Natàlia Llopart
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
| | - Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
| | - Nil Casajuana-Martin
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Leonardo Pardo
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
| | - Miriam Royo
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08034 Barcelona, Spain
- Department of Surfactants and Nanobiotechnology, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| |
Collapse
|
15
|
Ferraiolo M, Atik H, Ponthot R, Belo do Nascimento I, Beckers P, Stove C, Hermans E. Receptor density influences ligand-induced dopamine D 2L receptor homodimerization. Eur J Pharmacol 2021; 911:174557. [PMID: 34626593 DOI: 10.1016/j.ejphar.2021.174557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/03/2021] [Accepted: 09/10/2021] [Indexed: 01/23/2023]
Abstract
Chronic treatments with dopamine D2 receptor ligands induce fluctuations in D2 receptor density. Since D2 receptors tend to assemble as homodimers, we hypothesized that receptor density might influence constitutive and ligand-induced homodimerization. Using a nanoluciferase-based complementation assay to monitor dopamine D2L receptor homodimerization in a cellular model enabling the tetracycline-controlled expression of dopamine D2L receptors, we observed that increasing receptor density promoted constitutive dopamine D2L receptor homodimerization. Receptor full agonists promoted homodimerization, while antagonists and partial agonists disrupted dopamine D2L receptor homodimers. High receptor densities enhanced this inhibitory effect only for receptor antagonists. Taken together, our findings indicate that both receptor density and receptor ligands influence dopamine D2L receptor homodimerization, albeit excluding any strict correlation with ligands' intrinsic activity and highlighting further complexity to dopaminergic pharmacology.
Collapse
Affiliation(s)
- Mattia Ferraiolo
- Neuropharmacology Laboratory, Institute of Neuroscience, UCLouvain, Brussels, Belgium
| | - Hicham Atik
- Neuropharmacology Laboratory, Institute of Neuroscience, UCLouvain, Brussels, Belgium
| | - Romane Ponthot
- Neuropharmacology Laboratory, Institute of Neuroscience, UCLouvain, Brussels, Belgium
| | | | - Pauline Beckers
- Neuropharmacology Laboratory, Institute of Neuroscience, UCLouvain, Brussels, Belgium
| | - Christophe Stove
- Laboratory of Toxicology, Department of Bioanalysis, Ghent University, Ghent, Belgium
| | - Emmanuel Hermans
- Neuropharmacology Laboratory, Institute of Neuroscience, UCLouvain, Brussels, Belgium.
| |
Collapse
|
16
|
Rossino G, Rui M, Linciano P, Rossi D, Boiocchi M, Peviani M, Poggio E, Curti D, Schepmann D, Wünsch B, González-Avendaño M, Vergara-Jaque A, Caballero J, Collina S. Bitopic Sigma 1 Receptor Modulators to Shed Light on Molecular Mechanisms Underpinning Ligand Binding and Receptor Oligomerization. J Med Chem 2021; 64:14997-15016. [PMID: 34624193 DOI: 10.1021/acs.jmedchem.1c00886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The sigma 1 receptor (S1R) is an enigmatic ligand-operated chaperone involved in many important biological processes, and its functions are not fully understood yet. Herein, we developed a novel series of bitopic S1R ligands as versatile tools to investigate binding processes, allosteric modulation, and the oligomerization mechanism. These molecules have been prepared in the enantiopure form and subjected to a preliminary biological evaluation, while in silico investigations helped to rationalize the results. Compound 7 emerged as the first bitopic S1R ligand endowed with low nanomolar affinity (Ki = 2.6 nM) reported thus far. Computational analyses suggested that 7 may stabilize the open conformation of the S1R by simultaneously binding the occluded primary binding site and a peripheral site on the cytosol-exposed surface. These findings pave the way to new S1R ligands with enhanced activity and/or selectivity, which could also be used as probes for the identification of a potential allosteric site.
Collapse
Affiliation(s)
- Giacomo Rossino
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Marta Rui
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Pasquale Linciano
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Daniela Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Massimo Boiocchi
- Centro Grandi Strumenti, University of Pavia, via Bassi 21, 27100 Pavia, Italy
| | - Marco Peviani
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100 Pavia, Italy
| | - Elena Poggio
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100 Pavia, Italy
| | - Daniela Curti
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100 Pavia, Italy
| | - Dirk Schepmann
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Correnstraße 48, 48149 Münster, Germany
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Correnstraße 48, 48149 Münster, Germany
| | - Mariela González-Avendaño
- Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 1 Poniente, 1141 Talca, Chile
| | - Ariela Vergara-Jaque
- Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 1 Poniente, 1141 Talca, Chile
| | - Julio Caballero
- Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 1 Poniente, 1141 Talca, Chile
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| |
Collapse
|
17
|
Huang B, Wang H, Zheng Y, Li M, Kang G, Barreto-de-Souza V, Nassehi N, Knapp PE, Selley DE, Hauser KF, Zhang Y. Structure-Based Design and Development of Chemical Probes Targeting Putative MOR-CCR5 Heterodimers to Inhibit Opioid Exacerbated HIV-1 Infectivity. J Med Chem 2021; 64:7702-7723. [PMID: 34027668 PMCID: PMC10548452 DOI: 10.1021/acs.jmedchem.1c00408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Crystal structures of ligand-bound G-protein-coupled receptors provide tangible templates for rationally designing molecular probes. Herein, we report the structure-based design, chemical synthesis, and biological investigations of bivalent ligands targeting putative mu opioid receptor C-C motif chemokine ligand 5 (MOR-CCR5) heterodimers. The bivalent ligand VZMC013 possessed nanomolar level binding affinities for both the MOR and CCR5, inhibited CCL5-stimulated calcium mobilization, and remarkably improved anti-HIV-1BaL activity over previously reported bivalent ligands. VZMC013 inhibited viral infection in TZM-bl cells coexpressing CCR5 and MOR to a greater degree than cells expressing CCR5 alone. Furthermore, VZMC013 blocked human immunodeficiency virus (HIV)-1 entry in peripheral blood mononuclear cells (PBMC) cells in a concentration-dependent manner and inhibited opioid-accelerated HIV-1 entry more effectively in phytohemagglutinin-stimulated PBMC cells than in the absence of opioids. A three-dimensional molecular model of VZMC013 binding to the MOR-CCR5 heterodimer complex is constructed to elucidate its mechanism of action. VZMC013 is a potent chemical probe targeting MOR-CCR5 heterodimers and may serve as a pharmacological agent to inhibit opioid-exacerbated HIV-1 entry.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Anti-HIV Agents/chemistry
- Anti-HIV Agents/metabolism
- Anti-HIV Agents/pharmacology
- Binding Sites
- Dimerization
- Drug Design
- HIV-1/drug effects
- HIV-1/physiology
- Humans
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/virology
- Ligands
- Maraviroc/chemistry
- Molecular Docking Simulation
- Molecular Dynamics Simulation
- Naltrexone/chemistry
- Phytohemagglutinins/pharmacology
- Protein Binding
- Receptors, CCR5/chemistry
- Receptors, CCR5/metabolism
- Receptors, Opioid, mu/chemistry
- Receptors, Opioid, mu/metabolism
- Virus Internalization/drug effects
Collapse
Affiliation(s)
- Boshi Huang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, Virginia 23298, United States
| | - Huiqun Wang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, Virginia 23298, United States
| | - Yi Zheng
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, Virginia 23298, United States
| | - Mengchu Li
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, Virginia 23298, United States
| | - Guifeng Kang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, Virginia 23298, United States
| | - Victor Barreto-de-Souza
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, Virginia 23298, United States
| | - Nima Nassehi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, Virginia 23298, United States
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, Virginia 23298, United States
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, Virginia 23298, United States
| | - Dana E Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, Virginia 23298, United States
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, Virginia 23298, United States
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, 1101 E. Marshall Street, Richmond, Virginia 23298, United States
| | - Yan Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, Virginia 23298, United States
| |
Collapse
|
18
|
Gallo M, Moreno E, Defaus S, Ortega-Alvaro A, Gonzalez A, Robledo P, Cavaco M, Neves V, Castanho MARB, Casadó V, Pardo L, Maldonado R, Andreu D. Orally Active Peptide Vector Allows Using Cannabis to Fight Pain While Avoiding Side Effects. J Med Chem 2021; 64:6937-6948. [PMID: 33887904 PMCID: PMC8486167 DOI: 10.1021/acs.jmedchem.1c00484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
The
activation of cannabinoid CB1 receptors (CB1R) by Δ9-tetrahydrocannabinol (THC), the
main component of Cannabis sativa,
induces analgesia. CB1R activation, however, also causes
cognitive impairment via the serotonin 5HT2A receptor (5HT2AR), a component of a CB1R–5HT2AR heteromer, posing a serious drawback for cannabinoid therapeutic
use. We have shown that peptides reproducing CB1R transmembrane
(TM) helices 5 and 6, fused to a cell-penetrating sequence (CPP),
can alter the structure of the CB1R–5HT2AR heteromer and avert THC cognitive impairment while preserving analgesia.
Here, we report the optimization of these prototypes into drug-like
leads by (i) shortening the TM5, TM6, and CPP sequences, without losing
the ability to disturb the CB1R–5HT2AR heteromer, and (ii) extensive sequence remodeling to achieve protease
resistance and blood–brain barrier penetration. Our efforts
have culminated in the identification of an ideal candidate for cannabis-based
pain management, an orally active 16-residue peptide preserving THC-induced
analgesia.
Collapse
Affiliation(s)
- Maria Gallo
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Sira Defaus
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Antonio Ortega-Alvaro
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Angel Gonzalez
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Patricia Robledo
- Integrative Pharmacology and Systems Neuroscience, IMIM-Hospital del Mar Research Institute, 08003 Barcelona, Spain
| | - Marco Cavaco
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Vera Neves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Miguel A R B Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Leonardo Pardo
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Rafael Maldonado
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - David Andreu
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| |
Collapse
|
19
|
Ullmann T, Gienger M, Budzinski J, Hellmann J, Hübner H, Gmeiner P, Weikert D. Homobivalent Dopamine D 2 Receptor Ligands Modulate the Dynamic Equilibrium of D 2 Monomers and Homo- and Heterodimers. ACS Chem Biol 2021; 16:371-379. [PMID: 33435665 DOI: 10.1021/acschembio.0c00895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Dopamine D2 receptors (D2Rs) are major targets in the treatment of psychiatric and neurodegenerative diseases. As with many other G protein-coupled receptors (GPCRs), D2Rs interact within the cellular membrane, leading to a transient receptor homo- or heterodimerization. These interactions are known to alter ligand binding, signaling, and receptor trafficking. Bivalent ligands are ideally suited to target GPCR dimers and are composed of two pharmacophores connected by a spacer element. If properly designed, bivalent ligands are able to engange the two orthosteric binding sites of a GPCR dimer simultaneously. Taking advantage of previously developed ligands for heterodimers of D2R and the neurotensin receptor 1 (NTSR1), we synthesized homobivalent ligands targeting D2R. Employing bioluminescence resonance energy transfer, we found that the bivalent ligands 3b and 4b comprising a 92-atom spacer are able to foster D2R-homodimerization while simultaneously reducing interactions of D2R with NTSR1. Both receptors are coexpressed in the central nervous system and involved in important physiological processes. The newly developed bivalent ligands are excellent tools to further understand the pharmacological consequences of D2R homo- and heterodimerization. Not limited to the dopaminergic system, modifying class A GPCRs' dynamic equilibrium between monomers, homomers, and heteromers with bivalent ligands may represent a novel pharmacological concept paving the way toward innovative drugs.
Collapse
Affiliation(s)
- Tamara Ullmann
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Marie Gienger
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Julian Budzinski
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Jan Hellmann
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Dorothee Weikert
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| |
Collapse
|
20
|
Qian M, Ricarte A, Wouters E, Dalton JAR, Risseeuw MDP, Giraldo J, Van Calenbergh S. Discovery of a true bivalent dopamine D 2 receptor agonist. Eur J Med Chem 2021; 212:113151. [PMID: 33450620 DOI: 10.1016/j.ejmech.2020.113151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/06/2020] [Accepted: 12/28/2020] [Indexed: 10/22/2022]
Abstract
Employing two different alkyne-modified dopamine agonists to construct bivalent compounds via click chemistry resulted in the identification of a bivalent ligand (11c) for dopamine D2 receptor homodimer, which, compared to its parent monomeric alkyne, showed a 16-fold higher binding affinity for the dopamine D2 receptor and a 5-fold higher potency in a cAMP assay in HEK 293T cells stably expressing D2R. Molecular modeling revealed that 11c can indeed bridge the orthosteric binding sites of a D2R homodimer in a relaxed conformation via the TM5-TM6 interface and allows to largely rationalize the results of the receptor assays.
Collapse
Affiliation(s)
- Mingcheng Qian
- Department of Medicinal Chemistry, School of Pharmacy, Changzhou University, Changzhou, 213164, Jiangsu, China; Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000, Ghent, Belgium
| | - Adrián Ricarte
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigaciói InnovacióParc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Elise Wouters
- Laboratory of Toxicology, Ghent University, Ottergemsesteenweg 460, B-9000, Ghent, Belgium
| | - James A R Dalton
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigaciói InnovacióParc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Martijn D P Risseeuw
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000, Ghent, Belgium
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigaciói InnovacióParc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry, Ghent University, Ottergemsesteenweg 460, B-9000, Ghent, Belgium.
| |
Collapse
|
21
|
Morales P, Navarro G, Gómez‐Autet M, Redondo L, Fernández‐Ruiz J, Pérez‐Benito L, Cordomí A, Pardo L, Franco R, Jagerovic N. Discovery of Homobivalent Bitopic Ligands of the Cannabinoid CB 2 Receptor*. Chemistry 2020; 26:15839-15842. [PMID: 32794211 PMCID: PMC7756656 DOI: 10.1002/chem.202003389] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/12/2020] [Indexed: 12/21/2022]
Abstract
Single chemical entities with potential to simultaneously interact with two binding sites are emerging strategies in medicinal chemistry. We have designed, synthesized and functionally characterized the first bitopic ligands for the CB2 receptor. These compounds selectively target CB2 versus CB1 receptors. Their binding mode was studied by molecular dynamic simulations and site-directed mutagenesis.
Collapse
Affiliation(s)
- Paula Morales
- Medicinal Chemistry InstituteSpanish Research CouncilMadridSpain
| | - Gemma Navarro
- Department of Biochemistry and Physiology, CIBERNEDFaculty of Pharmacy and Food SciencesUniversitat de BarcelonaBarcelonaSpain
| | - Marc Gómez‐Autet
- Laboratory of Computational Medicine, Biostatistics UnitFaculty of MedicineUniversitat Autónoma de BarcelonaBarcelonaSpain
| | - Laura Redondo
- Medicinal Chemistry InstituteSpanish Research CouncilMadridSpain
| | - Javier Fernández‐Ruiz
- Department of Biochemistry and Molecular Biology, CIBERNED, IRYCISFaculty of MedicineUniversidad Complutense de MadridMadridSpain
| | - Laura Pérez‐Benito
- Laboratory of Computational Medicine, Biostatistics UnitFaculty of MedicineUniversitat Autónoma de BarcelonaBarcelonaSpain
- Present address: Computational ChemistryJanssen Research & Development, Janssen Pharmaceutica N.V.Belgium
| | - Arnau Cordomí
- Laboratory of Computational Medicine, Biostatistics UnitFaculty of MedicineUniversitat Autónoma de BarcelonaBarcelonaSpain
| | - Leonardo Pardo
- Laboratory of Computational Medicine, Biostatistics UnitFaculty of MedicineUniversitat Autónoma de BarcelonaBarcelonaSpain
| | - Rafael Franco
- Department of Biochemistry and Physiology, CIBERNEDFaculty of Pharmacy and Food SciencesUniversitat de BarcelonaBarcelonaSpain
- Department of Biochemistry and Molecular Biology, CIBERNEDSchool of ChemistryUniversitat de BarcelonaBarcelonaSpain
| | - Nadine Jagerovic
- Medicinal Chemistry InstituteSpanish Research CouncilMadridSpain
| |
Collapse
|
22
|
Huang B, St Onge CM, Ma H, Zhang Y. Design of bivalent ligands targeting putative GPCR dimers. Drug Discov Today 2020; 26:189-199. [PMID: 33075471 DOI: 10.1016/j.drudis.2020.10.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/15/2020] [Accepted: 10/09/2020] [Indexed: 12/17/2022]
Abstract
G protein-coupled receptors (GPCRs) have been exploited as primary targets for drug discovery, and GPCR dimerization offers opportunities for drug design and disease treatment. An important strategy for targeting putative GPCR dimers is the use of bivalent ligands, which are single molecules that contain two pharmacophores connected through a spacer. Here, we discuss the selection of pharmacophores, the optimal length and chemical composition of the spacer, and the choice of spacer attachment points to the pharmacophores. Furthermore, we review the most recent advances (from 2018 to the present) in the design, discovery and development of bivalent ligands. We aim to reveal the state-of-the-art design strategy for bivalent ligands and provide insights into future opportunities in this promising field of drug discovery.
Collapse
Affiliation(s)
- Boshi Huang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, VA 23298, USA
| | - Celsey M St Onge
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, VA 23298, USA
| | - Hongguang Ma
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, VA 23298, USA
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E. Leigh Street, Richmond, VA 23298, USA.
| |
Collapse
|
23
|
Martel JC, Gatti McArthur S. Dopamine Receptor Subtypes, Physiology and Pharmacology: New Ligands and Concepts in Schizophrenia. Front Pharmacol 2020; 11:1003. [PMID: 32765257 PMCID: PMC7379027 DOI: 10.3389/fphar.2020.01003] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Dopamine receptors are widely distributed within the brain where they play critical modulator roles on motor functions, motivation and drive, as well as cognition. The identification of five genes coding for different dopamine receptor subtypes, pharmacologically grouped as D1- (D1 and D5) or D2-like (D2S, D2L, D3, and D4) has allowed the demonstration of differential receptor function in specific neurocircuits. Recent observation on dopamine receptor signaling point at dopamine-glutamate-NMDA neurobiology as the most relevant in schizophrenia and for the development of new therapies. Progress in the chemistry of D1- and D2-like receptor ligands (agonists, antagonists, and partial agonists) has provided more selective compounds possibly able to target the dopamine receptors homo and heterodimers and address different schizophrenia symptoms. Moreover, an extensive evaluation of the functional effect of these agents on dopamine receptor coupling and intracellular signaling highlights important differences that could also result in highly differentiated clinical pharmacology. The review summarizes the recent advances in the field, addressing the relevance of emerging new targets in schizophrenia in particular in relation to the dopamine - glutamate NMDA systems interactions.
Collapse
|
24
|
Shchepinova MM, Hanyaloglu AC, Frost GS, Tate EW. Chemical biology of noncanonical G protein-coupled receptor signaling: Toward advanced therapeutics. Curr Opin Chem Biol 2020; 56:98-110. [PMID: 32446179 DOI: 10.1016/j.cbpa.2020.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/20/2022]
Abstract
G protein-coupled receptors (GPCRs), the largest family of signaling membrane proteins, are the target of more than 30% of the drugs on the market. Recently, it has become clear that GPCR functions are far more multidimensional than previously thought, with multiple noncanonical aspects coming to light, including biased, oligomeric, and compartmentalized signaling. These additional layers of functional selectivity greatly expand opportunities for advanced therapeutic interventions, but the development of new chemical biology tools is absolutely required to improve our understanding of noncanonical GPCR regulation and pave the way for future drugs. In this opinion, we highlight the most notable examples of chemical and chemogenetic tools addressing new paradigms in GPCR signaling, discuss their promises and limitations, and explore future directions.
Collapse
Affiliation(s)
- Maria M Shchepinova
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 80 Wood Lane, London, W12 0BZ, UK.
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Dept. Surgery and Cancer, Imperial College, London, UK
| | - Gary S Frost
- Department of Medicine, Faculty of Medicine, Nutrition and Dietetic Research Group, Imperial College, London, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 80 Wood Lane, London, W12 0BZ, UK.
| |
Collapse
|
25
|
Newman AH, Battiti FO, Bonifazi A. 2016 Philip S. Portoghese Medicinal Chemistry Lectureship: Designing Bivalent or Bitopic Molecules for G-Protein Coupled Receptors. The Whole Is Greater Than the Sum of Its Parts. J Med Chem 2020; 63:1779-1797. [PMID: 31499001 PMCID: PMC8281448 DOI: 10.1021/acs.jmedchem.9b01105] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The genesis of designing bivalent or bitopic molecules that engender unique pharmacological properties began with Portoghese's work directed toward opioid receptors, in the early 1980s. This strategy has evolved as an attractive way to engineer highly selective compounds for targeted G-protein coupled receptors (GPCRs) with optimized efficacies and/or signaling bias. The emergence of X-ray crystal structures of many GPCRs and the identification of both orthosteric and allosteric binding sites have provided further guidance to ligand drug design that includes a primary pharmacophore (PP), a secondary pharmacophore (SP), and a linker between them. It is critical to note the synergistic relationship among all three of these components as they contribute to the overall interaction of these molecules with their receptor proteins and that strategically designed combinations have and will continue to provide the GPCR molecular tools of the future.
Collapse
Affiliation(s)
- Amy Hauck Newman
- Corresponding author: Amy H. Newman: Phone: (443)-740-2887. Fax: (443)-740-2111.
| | - Francisco O. Battiti
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| |
Collapse
|
26
|
Solbak SMØ, Zang J, Narayanan D, Høj LJ, Bucciarelli S, Softley C, Meier S, Langkilde AE, Gotfredsen CH, Sattler M, Bach A. Developing Inhibitors of the p47phox-p22phox Protein-Protein Interaction by Fragment-Based Drug Discovery. J Med Chem 2020; 63:1156-1177. [PMID: 31922756 DOI: 10.1021/acs.jmedchem.9b01492] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nicotinamide adenine dinucleotide phosphate oxidase isoform 2 is an enzyme complex, which generates reactive oxygen species and contributes to oxidative stress. The p47phox-p22phox interaction is critical for the activation of the catalytical NOX2 domain, and p47phox is a potential target for therapeutic intervention. By screening 2500 fragments using fluorescence polarization and a thermal shift assay and validation by surface plasmon resonance, we found eight hits toward the tandem SH3 domain of p47phox (p47phoxSH3A-B) with KD values of 400-600 μM. Structural studies revealed that fragments 1 and 2 bound two separate binding sites in the elongated conformation of p47phoxSH3A-B and these competed with p22phox for binding to p47phoxSH3A-B. Chemical optimization led to a dimeric compound with the ability to potently inhibit the p47phoxSH3A-B-p22phox interaction (Ki of 20 μM). Thereby, we reveal a new way of targeting p47phox and present the first report of drug-like molecules with the ability to bind p47phox and inhibit its interaction with p22phox.
Collapse
Affiliation(s)
- Sara Marie Øie Solbak
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , 2100 Copenhagen , Denmark
| | - Jie Zang
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , 2100 Copenhagen , Denmark
| | - Dilip Narayanan
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , 2100 Copenhagen , Denmark
| | - Lars Jakobsen Høj
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , 2100 Copenhagen , Denmark
| | - Saskia Bucciarelli
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , 2100 Copenhagen , Denmark
| | - Charlotte Softley
- Institute of Structural Biology , Helmholtz Zentrum München , 85764 Neuherberg , Germany.,Biomolecular NMR and Center for Integrated Protein Science Munich at Department of Chemistry , Technical University of Munich , 85747 Garching , Germany
| | - Sebastian Meier
- Department of Chemistry , Technical University of Denmark , Kemitorvet , 2800 Kgs Lyngby , Denmark
| | - Annette Eva Langkilde
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , 2100 Copenhagen , Denmark
| | | | - Michael Sattler
- Institute of Structural Biology , Helmholtz Zentrum München , 85764 Neuherberg , Germany.,Biomolecular NMR and Center for Integrated Protein Science Munich at Department of Chemistry , Technical University of Munich , 85747 Garching , Germany
| | - Anders Bach
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences , University of Copenhagen , Universitetsparken 2 , 2100 Copenhagen , Denmark
| |
Collapse
|
27
|
Pérez-Benito L, Llinas del Torrent C, Pardo L, Tresadern G. The computational modeling of allosteric modulation of metabotropic glutamate receptors. FROM STRUCTURE TO CLINICAL DEVELOPMENT: ALLOSTERIC MODULATION OF G PROTEIN-COUPLED RECEPTORS 2020; 88:1-33. [DOI: 10.1016/bs.apha.2020.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
28
|
Ferré S, Ciruela F, Casadó V, Pardo L. Oligomerization of G protein-coupled receptors: Still doubted? PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 169:297-321. [DOI: 10.1016/bs.pmbts.2019.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
29
|
Casadó-Anguera V, Cortés A, Casadó V, Moreno E. Targeting the receptor-based interactome of the dopamine D1 receptor: looking for heteromer-selective drugs. Expert Opin Drug Discov 2019; 14:1297-1312. [DOI: 10.1080/17460441.2019.1664469] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Verònica Casadó-Anguera
- Laboratory of Molecular Neurobiology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, (IBUB), Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Antoni Cortés
- Laboratory of Molecular Neurobiology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, (IBUB), Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Vicent Casadó
- Laboratory of Molecular Neurobiology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, (IBUB), Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Estefanía Moreno
- Laboratory of Molecular Neurobiology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, (IBUB), Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| |
Collapse
|
30
|
Montaruli M, Alberga D, Ciriaco F, Trisciuzzi D, Tondo AR, Mangiatordi GF, Nicolotti O. Accelerating Drug Discovery by Early Protein Drug Target Prediction Based on a Multi-Fingerprint Similarity Search. Molecules 2019; 24:molecules24122233. [PMID: 31207991 PMCID: PMC6631269 DOI: 10.3390/molecules24122233] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/08/2019] [Accepted: 06/12/2019] [Indexed: 01/06/2023] Open
Abstract
In this continuing work, we have updated our recently proposed Multi-fingerprint Similarity Search algorithm (MuSSel) by enabling the generation of dominant ionized species at a physiological pH and the exploration of a larger data domain, which included more than half a million high-quality small molecules extracted from the latest release of ChEMBL (version 24.1, at the time of writing). Provided with a high biological assay confidence score, these selected compounds explored up to 2822 protein drug targets. To improve the data accuracy, samples marked as prodrugs or with equivocal biological annotations were not considered. Notably, MuSSel performances were overall improved by using an object-relational database management system based on PostgreSQL. In order to challenge the real effectiveness of MuSSel in predicting relevant therapeutic drug targets, we analyzed a pool of 36 external bioactive compounds published in the Journal of Medicinal Chemistry from October to December 2018. This study demonstrates that the use of highly curated chemical and biological experimental data on one side, and a powerful multi-fingerprint search algorithm on the other, can be of the utmost importance in addressing the fate of newly conceived small molecules, by strongly reducing the attrition of early phases of drug discovery programs.
Collapse
Affiliation(s)
- Michele Montaruli
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| | - Domenico Alberga
- Cineca, Via Magnanelli 6/3, 40033 Casalecchio di Reno, Bologna, Italy.
| | - Fulvio Ciriaco
- Dipartimento di Chimica, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| | - Daniela Trisciuzzi
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| | - Anna Rita Tondo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via la Masa 19, 20156 Milano, Italy.
| | | | - Orazio Nicolotti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.
| |
Collapse
|
31
|
Moreno E, Cavic M, Krivokuca A, Casadó V, Canela E. The Endocannabinoid System as a Target in Cancer Diseases: Are We There Yet? Front Pharmacol 2019; 10:339. [PMID: 31024307 PMCID: PMC6459931 DOI: 10.3389/fphar.2019.00339] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 03/19/2019] [Indexed: 12/15/2022] Open
Abstract
The endocannabinoid system (ECS) has been placed in the anti-cancer spotlight in the last decade. The immense data load published on its dual role in both tumorigenesis and inhibition of tumor growth and metastatic spread has transformed the cannabinoid receptors CB1 (CB1R) and CB2 (CB2R), and other members of the endocannabinoid-like system, into attractive new targets for the treatment of various cancer subtypes. Although the clinical use of cannabinoids has been extensively documented in the palliative setting, clinical trials on their application as anti-cancer drugs are still ongoing. As drug repurposing is significantly faster and more economical than de novo introduction of a new drug into the clinic, there is hope that the existing pharmacokinetic and safety data on the ECS ligands will contribute to their successful translation into oncological healthcare. CB1R and CB2R are members of a large family of membrane proteins called G protein-coupled receptors (GPCR). GPCRs can form homodimers, heterodimers and higher order oligomers with other GPCRs or non-GPCRs. Currently, several CB1R and CB2R-containing heteromers have been reported and, in cancer cells, CB2R form heteromers with the G protein-coupled chemokine receptor CXCR4, the G protein-coupled receptor 55 (GPR55) and the tyrosine kinase receptor (TKR) human V-Erb-B2 Avian Erythroblastic Leukemia Viral Oncogene Homolog 2 (HER2). These protein complexes possess unique pharmacological and signaling properties, and their modulation might affect the antitumoral activity of the ECS. This review will explore the potential of the endocannabinoid network in the anti-cancer setting as well as the clinical and ethical pitfalls behind it, and will develop on the value of cannabinoid receptor heteromers as potential new targets for anti-cancer therapies and as prognostic biomarkers.
Collapse
Affiliation(s)
- Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Milena Cavic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Ana Krivokuca
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Enric Canela
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
32
|
Wouters E, Marín AR, Dalton JAR, Giraldo J, Stove C. Distinct Dopamine D₂ Receptor Antagonists Differentially Impact D₂ Receptor Oligomerization. Int J Mol Sci 2019; 20:ijms20071686. [PMID: 30987329 PMCID: PMC6480712 DOI: 10.3390/ijms20071686] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 12/21/2022] Open
Abstract
Dopamine D2 receptors (D2R) are known to form transient homodimer complexes, of which the increased formation has already been associated with development of schizophrenia. Pharmacological targeting and modulation of the equilibrium of these receptor homodimers might lead to a better understanding of the critical role played by these complexes in physiological and pathological conditions. Whereas agonist addition has shown to prolong the D2R dimer lifetime and increase the level of dimer formation, the possible influence of D2R antagonists on dimerization has remained rather unexplored. Here, using a live-cell reporter assay based on the functional complementation of a split Nanoluciferase, a panel of six D2R antagonists were screened for their ability to modulate the level of D2LR dimer formation. Incubation with the D2R antagonist spiperone decreased the level of D2LR dimer formation significantly by 40–60% in real-time and after long-term (≥16 h) incubations. The fact that dimer formation of the well-studied A2a–D2LR dimer was not altered following incubation with spiperone supports the specificity of this observation. Other D2R antagonists, such as clozapine, risperidone, and droperidol did not significantly evoke this dissociation event. Furthermore, molecular modeling reveals that spiperone presents specific Tyr1995.48 and Phe3906.52 conformations, compared to clozapine, which may determine D2R homodimerization.
Collapse
Affiliation(s)
- Elise Wouters
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Adrián Ricarte Marín
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | - James Andrew Rupert Dalton
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| | - Christophe Stove
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
33
|
Current status of multiscale simulations on GPCRs. Curr Opin Struct Biol 2019; 55:93-103. [DOI: 10.1016/j.sbi.2019.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/19/2019] [Accepted: 02/27/2019] [Indexed: 01/14/2023]
|
34
|
Cortés A, Casadó-Anguera V, Moreno E, Casadó V. The heterotetrameric structure of the adenosine A 1-dopamine D 1 receptor complex: Pharmacological implication for restless legs syndrome. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2019; 84:37-78. [PMID: 31229177 DOI: 10.1016/bs.apha.2019.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Dopaminergic and purinergic signaling play a pivotal role in neurological diseases associated with motor symptoms, including Parkinson's disease (PD), multiple sclerosis, amyotrophic lateral sclerosis, Huntington disease, Restless Legs Syndrome (RLS), spinal cord injury (SCI), and ataxias. Extracellular dopamine and adenosine exert their functions interacting with specific dopamine (DR) or adenosine (AR) receptors, respectively, expressed on the surface of target cells. These receptors are members of the family A of G protein-coupled receptors (GPCRs), which is the largest protein superfamily in mammalian genomes. GPCRs are target of about 40% of all current marketed drugs, highlighting their importance in clinical medicine. The striatum receives the densest dopamine innervations and contains the highest density of dopamine receptors. The modulatory role of adenosine on dopaminergic transmission depends largely on the existence of antagonistic interactions mediated by specific subtypes of DRs and ARs, the so-called A2AR-D2R and A1R-D1R interactions. Due to the dopamine/adenosine antagonism in the CNS, it was proposed that ARs and DRs could form heteromers in the neuronal cell surface. Therefore, adenosine can affect dopaminergic signaling through receptor-receptor interactions and by modulations in their shared intracellular pathways in the striatum and spinal cord. In this work we describe the allosteric modulations between GPCR protomers, focusing in those of adenosine and dopamine within the A1R-D1R heteromeric complex, which is involved in RLS. We also propose that the knowledge about the intricate allosteric interactions within the A1R-D1R heterotetramer, may facilitate the treatment of motor alterations, not only when the dopamine pathway is hyperactivated (RLS, chorea, etc.) but also when motor function is decreased (SCI, aging, PD, etc.).
Collapse
Affiliation(s)
- Antoni Cortés
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Verònica Casadó-Anguera
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Estefanía Moreno
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Vicent Casadó
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
35
|
Casadó-Anguera V, Moreno E, Mallol J, Ferré S, Canela EI, Cortés A, Casadó V. Reinterpreting anomalous competitive binding experiments within G protein-coupled receptor homodimers using a dimer receptor model. Pharmacol Res 2018; 139:337-347. [PMID: 30472462 DOI: 10.1016/j.phrs.2018.11.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/14/2018] [Accepted: 11/21/2018] [Indexed: 12/01/2022]
Abstract
An increasing number of G protein-coupled receptors (GPCRs) have been reported to be expressed in the plasma membrane as dimers. Since most ligand binding data are currently fitted by classical equations developed only for monomeric receptors, the interpretation of data could be misleading in the presence of GPCR dimers. On the other hand, the equations developed from dimer receptor models assuming the existence of two orthosteric binding sites within the dimeric molecule offer the possibility to directly calculate macroscopic equilibrium dissociation constants for the two sites, an index of cooperativity (DC) that reflects the molecular communication within the dimer and, importantly, a constant of radioligand-competitor allosteric interaction (KDAB) in competitive assays. Here, we provide a practical way to fit competitive binding data that allows the interpretation of apparently anomalous results, such as competition curves that could be either bell-shaped, monophasic or biphasic depending on the assay conditions. The consideration of a radioligand-competitor allosteric interaction allows fitting these curve patterns both under simulation conditions and in real radioligand binding experiments, obtaining competitor affinity parameters closer to the actual values. Our approach is the first that, assuming the formation of receptor homodimers, is able to explain several experimental results previously considered erroneous due to their impossibility to be fitted. We also deduce the radioligand concentration responsible for the conversion of biphasic to monophasic or to bell-shaped curves in competitive radioligand binding assays. In conclusion, bell-shaped curves in competitive binding experiments constitute evidence for GPCR homodimerization.
Collapse
Affiliation(s)
- Verònica Casadó-Anguera
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.
| | - Estefanía Moreno
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.
| | - Josefa Mallol
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.
| | - Sergi Ferré
- National Institute on Drug Abuse, I.R.P., N.I.H., D.H.H.S., Baltimore, MD, 21224, USA.
| | - Enric I Canela
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.
| | - Antoni Cortés
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.
| | - Vicent Casadó
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.
| |
Collapse
|