101
|
Suno R, Sugita Y, Morimoto K, Takazaki H, Tsujimoto H, Hirose M, Suno-Ikeda C, Nomura N, Hino T, Inoue A, Iwasaki K, Kato T, Iwata S, Kobayashi T. Structural insights into the G protein selectivity revealed by the human EP3-G i signaling complex. Cell Rep 2022; 40:111323. [PMID: 36103815 DOI: 10.1016/j.celrep.2022.111323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/18/2022] [Accepted: 08/17/2022] [Indexed: 11/03/2022] Open
Abstract
Prostaglandin receptors have been implicated in a wide range of functions, including inflammation, immune response, reproduction, and cancer. Our group has previously determined the crystal structure of the active-like EP3 bound to its endogenous agonist, prostaglandin E2. Here, we present the single-particle cryoelectron microscopy (cryo-EM) structure of the human EP3-Gi signaling complex at a resolution of 3.4 Å. The structure reveals the binding mode of Gi to EP3 and the structural changes induced in EP3 by Gi binding. In addition, we compare the structure of the EP3-Gi complex with other subtypes of prostaglandin receptors (EP2 and EP4) bound to Gs that have been previously reported and examine the differences in amino acid composition at the receptor-G protein interface. Mutational analysis reveals that the selectivity of the G protein depends on specific amino acid residues in the second intracellular loop and TM5.
Collapse
Affiliation(s)
- Ryoji Suno
- Department of Medical Chemistry, Kansai Medical University, Hirakata 573-1010, Japan.
| | - Yukihiko Sugita
- Institute for Protein Research, Osaka University, Suita 565-0871, Japan; Hakubi Center for Advanced Research, Kyoto University, Kyoto 606-8501, Japan; Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Kazushi Morimoto
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hiroko Takazaki
- Institute for Protein Research, Osaka University, Suita 565-0871, Japan
| | - Hirokazu Tsujimoto
- Department of Cell Biology and Medical Chemistry, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan
| | - Mika Hirose
- Institute for Protein Research, Osaka University, Suita 565-0871, Japan
| | - Chiyo Suno-Ikeda
- Department of Medical Chemistry, Kansai Medical University, Hirakata 573-1010, Japan
| | - Norimichi Nomura
- Department of Cell Biology and Medical Chemistry, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tomoya Hino
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori, Japan; Center for Research on Green Sustainable Chemistry, Tottori University, Tottori, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-Ku, Sendai, Miyagi 980-8578, Japan
| | - Kenji Iwasaki
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Takayuki Kato
- Institute for Protein Research, Osaka University, Suita 565-0871, Japan
| | - So Iwata
- Department of Cell Biology and Medical Chemistry, Graduate School of Medicine, Kyoto University, Konoe-cho, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takuya Kobayashi
- Department of Medical Chemistry, Kansai Medical University, Hirakata 573-1010, Japan; Japan Agency for Medical Research and Development (AMED), Core Research for Evolutional Science and Technology (CREST), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
102
|
Ye L, Wang X, McFarland A, Madsen JJ. 19F NMR: A promising tool for dynamic conformational studies of G protein-coupled receptors. Structure 2022; 30:1372-1384. [PMID: 36130592 DOI: 10.1016/j.str.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 07/31/2022] [Accepted: 08/23/2022] [Indexed: 10/14/2022]
Abstract
Advances in X-ray crystallography and cryoelectron microscopy enabled unprecedented insights into the activation processes of G protein-coupled receptors (GPCRs). However, these static receptor structures provide limited information about dynamics and conformational transitions that play pivotal roles in mediating signaling diversity through the multifaceted interactions between ligands, receptors, and transducers. Developing NMR approaches to probe the dynamics of conformational transitions will push the frontier of receptor science toward a more comprehensive understanding of these signaling processes. Although much progress has been made during the last decades, it remains challenging to delineate receptor conformational states and interrogate the functions of the individual states at a quantitative level. Here we cover the progress of 19F NMR applications in GPCR conformational and dynamic studies during the past 20 years. Current challenges and limitations of 19F NMR for studying GPCR dynamics are also discussed, along with experimental strategies that will drive this field forward.
Collapse
Affiliation(s)
- Libin Ye
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA; H. Lee Moffitt Cancer Center & Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA.
| | - Xudong Wang
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Aidan McFarland
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Jesper J Madsen
- Global and Planetary Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
103
|
Welling MT, Deseo MA, Bacic A, Doblin MS. Biosynthetic origins of unusual cannabimimetic phytocannabinoids in Cannabis sativa L: A review. PHYTOCHEMISTRY 2022; 201:113282. [PMID: 35718133 DOI: 10.1016/j.phytochem.2022.113282] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/02/2022] [Accepted: 06/11/2022] [Indexed: 06/15/2023]
Abstract
Plants of Cannabis sativa L. (Cannabaceae) produce an array of more than 160 isoprenylated resorcinyl polyketides, commonly referred to as phytocannabinoids. These compounds represent molecules of therapeutic importance due to their modulation of the human endocannabinoid system (ECS). While understanding of the biosynthesis of the major phytocannabinoids Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD) has grown rapidly in recent years, the biosynthetic origin and genetic regulation of many potentially therapeutically relevant minor phytocannabinoids remain unknown, which limits the development of chemotypically elite varieties of C. sativa. This review provides an up-to-date inventory of unusual phytocannabinoids which exhibit cannabimimetic-like activities and proposes putative metabolic origins. Metabolic branch points exploitable for combinatorial biosynthesis and engineering of phytocannabinoids with augmented therapeutic activities are also described, as is the role of phytocannabinoid remodelling to accelerate the therapeutic portfolio expansion in C. sativa.
Collapse
Affiliation(s)
- Matthew T Welling
- La Trobe Institute for Agriculture & Food, AgriBio Building, Department of Animal, Plant and Soil Sciences, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia
| | - Myrna A Deseo
- La Trobe Institute for Agriculture & Food, AgriBio Building, Department of Animal, Plant and Soil Sciences, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia; Australian Research Council Research Hub for Medicinal Agriculture, AgriBio Building, La Trobe University, Bundoora, VIC 3086, Australia
| | - Antony Bacic
- La Trobe Institute for Agriculture & Food, AgriBio Building, Department of Animal, Plant and Soil Sciences, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia; Australian Research Council Research Hub for Medicinal Agriculture, AgriBio Building, La Trobe University, Bundoora, VIC 3086, Australia
| | - Monika S Doblin
- La Trobe Institute for Agriculture & Food, AgriBio Building, Department of Animal, Plant and Soil Sciences, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia; Australian Research Council Research Hub for Medicinal Agriculture, AgriBio Building, La Trobe University, Bundoora, VIC 3086, Australia.
| |
Collapse
|
104
|
Zhang LY, Kim AY, Cheer JF. Regulation of glutamate homeostasis in the nucleus accumbens by astrocytic CB1 receptors and its role in cocaine-motivated behaviors. ADDICTION NEUROSCIENCE 2022; 3:100022. [PMID: 36419922 PMCID: PMC9681119 DOI: 10.1016/j.addicn.2022.100022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cannabinoid type 1 receptors (CB1Rs) orchestrate brain reward circuitry and are prevalent neurobiological targets for endocannabinoids and cannabis in the mammalian brain. Decades of histological and electrophysiological studies have established CB1R as presynaptic G-protein coupled receptors (GPCRs) that inhibit neurotransmitter release through retrograde signaling mechanisms. Recent seminal work demonstrates CB1R expression on astrocytes and the pivotal function of glial cells in endocannabinoid-mediated modulation of neuron-astrocyte signaling. Here, we review key facets of CB1R-mediated astroglia regulation of synaptic glutamate transmission in the nucleus accumbens with a specific emphasis on cocaine-directed behaviors.
Collapse
Affiliation(s)
- Lan-Yuan Zhang
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Andrew Y. Kim
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Joseph F. Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States of America
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
105
|
Ferranti AS, Foster DJ. Cannabinoid type-2 receptors: An emerging target for regulating schizophrenia-relevant brain circuits. Front Neurosci 2022; 16:925792. [PMID: 36033626 PMCID: PMC9403189 DOI: 10.3389/fnins.2022.925792] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Although the cannabinoid type-2 receptor (CB2) is highly expressed in the immune system, emerging evidence points to CB2 playing a key role in regulating neuronal function in the central nervous system. Recent anatomical studies, combined with electrophysiological studies, indicate that CB2 receptors are expressed in specific dopaminergic and glutamatergic brain circuits that are hyperactive in schizophrenia patients. The ability of CB2 receptors to inhibit dopaminergic and hippocampal circuits, combined with the anti-inflammatory effects of CB2 receptor activation, make this receptor an intriguing target for treating schizophrenia, a disease where novel interventions that move beyond dopamine receptor antagonists are desperately needed. The development of new CB2-related pharmacological and genetic tools, including the first small molecule positive allosteric modulator of CB2 receptors, has greatly advanced our understanding of this receptor. While more work is needed to further elucidate the translational value of selectively targeting CB2 receptors with respect to schizophrenia, the studies discussed below could suggest that CB2 receptors are anatomically located in schizophrenia-relevant circuits, where the physiological consequence of CB2 receptor activation could correct circuit-based deficits commonly associated with positive and cognitive deficits.
Collapse
Affiliation(s)
- Anthony S. Ferranti
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Daniel J. Foster
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
106
|
Dafun AS, Marcoux J. Structural mass spectrometry of membrane proteins. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2022; 1870:140813. [PMID: 35750312 DOI: 10.1016/j.bbapap.2022.140813] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/10/2022] [Accepted: 06/17/2022] [Indexed: 06/15/2023]
Abstract
The analysis of proteins and protein complexes by mass spectrometry (MS) has come a long way since the invention of electrospray ionization (ESI) in the mid 80s. Originally used to characterize small soluble polypeptide chains, MS has progressively evolved over the past 3 decades towards the analysis of samples of ever increasing heterogeneity and complexity, while the instruments have become more and more sensitive and resolutive. The proofs of concepts and first examples of most structural MS methods appeared in the early 90s. However, their application to membrane proteins, key targets in the biopharma industry, is more recent. Nowadays, a wealth of information can be gathered from such MS-based methods, on all aspects of membrane protein structure: sequencing (and more precisely proteoform characterization), but also stoichiometry, non-covalent ligand binding (metals, drug, lipids, carbohydrates), conformations, dynamics and distance restraints for modelling. In this review, we present the concept and some historical and more recent applications on membrane proteins, for the major structural MS methods.
Collapse
Affiliation(s)
- Angelique Sanchez Dafun
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Julien Marcoux
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
107
|
Benzothiazole Derivatives Endowed with Antiproliferative Activity in Paraganglioma and Pancreatic Cancer Cells: Structure–Activity Relationship Studies and Target Prediction Analysis. Pharmaceuticals (Basel) 2022; 15:ph15080937. [PMID: 36015085 PMCID: PMC9412555 DOI: 10.3390/ph15080937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/14/2022] [Accepted: 07/25/2022] [Indexed: 12/04/2022] Open
Abstract
The antiproliferative effects played by benzothiazoles in different cancers have aroused the interest for these molecules as promising antitumor agents. In this work, a library of phenylacetamide derivatives containing the benzothiazole nucleus was synthesized and compounds were tested for their antiproliferative activity in paraganglioma and pancreatic cancer cell lines. The novel synthesized compounds induced a marked viability reduction at low micromolar concentrations both in paraganglioma and pancreatic cancer cells. Derivative 4l showed a greater antiproliferative effect and higher selectivity index against cancer cells, as compared to other compounds. Notably, combinations of derivative 4l with gemcitabine at low concentrations induced enhanced and synergistic effects on pancreatic cancer cell viability, thus supporting the relevance of compound 4l in the perspective of clinical translation. A target prediction analysis was also carried out on 4l by using multiple computational tools, identifying cannabinoid receptors and sentrin-specific proteases as putative targets contributing to the observed antiproliferative activity.
Collapse
|
108
|
Whiting ZM, Yin J, de la Harpe SM, Vernall AJ, Grimsey NL. Developing the Cannabinoid Receptor 2 (CB2) pharmacopoeia: past, present, and future. Trends Pharmacol Sci 2022; 43:754-771. [PMID: 35906103 DOI: 10.1016/j.tips.2022.06.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 12/28/2022]
Abstract
Cannabinoid Receptor 2 (CB2) is a G protein-coupled receptor (GPCR) with considerable, though as yet unrealised, therapeutic potential. Promising preclinical data supports the applicability of CB2 activation in autoimmune and inflammatory diseases, pain, neurodegeneration, and osteoporosis. A diverse pharmacopoeia of cannabinoid ligands is available, which has led to considerable advancements in the understanding of CB2 function and extensive preclinical evaluation. However, until recently, most CB2 ligands were highly lipophilic and as such not optimal for clinical application due to unfavourable physicochemical properties. A number of strategies have been applied to develop CB2 ligands to achieve closer to 'drug-like' properties and a few such compounds have now undergone clinical trial. We review the current state of CB2 ligand development and progress in optimising physicochemical properties, understanding advanced molecular pharmacology such as functional selectivity, and clinical evaluation of CB2-targeting compounds.
Collapse
Affiliation(s)
- Zak M Whiting
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jiazhen Yin
- Department of Chemistry, Division of Sciences, University of Otago, Dunedin, New Zealand
| | - Sara M de la Harpe
- Department of Chemistry, Division of Sciences, University of Otago, Dunedin, New Zealand
| | - Andrea J Vernall
- Department of Chemistry, Division of Sciences, University of Otago, Dunedin, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Natasha L Grimsey
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| |
Collapse
|
109
|
Gado F, Ferrisi R, Polini B, Mohamed KA, Ricardi C, Lucarini E, Carpi S, Domenichini F, Stevenson LA, Rapposelli S, Saccomanni G, Nieri P, Ortore G, Pertwee RG, Ghelardini C, Di Cesare Mannelli L, Chiellini G, Laprairie RB, Manera C. Design, Synthesis, and Biological Activity of New CB2 Receptor Ligands: from Orthosteric and Allosteric Modulators to Dualsteric/Bitopic Ligands. J Med Chem 2022; 65:9918-9938. [PMID: 35849804 PMCID: PMC10168668 DOI: 10.1021/acs.jmedchem.2c00582] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The design of dualsteric/bitopic agents as single chemical entities able to simultaneously interact with both the orthosteric and an allosteric binding site represents a novel approach in medicinal chemistry. Biased dualsteric/bitopic agents could enhance certain signaling pathways while diminishing the others that cause unwanted side effects. We have designed, synthesized, and functionally characterized the first CB2R heterobivalent bitopic ligands. In contrast to the parent orthosteric compound, our bitopic ligands selectively target CB2R versus CB1R and show a functional selectivity for the cAMP signaling pathway versus βarrestin2 recruitment. Moreover, the most promising bitopic ligand FD-22a displayed anti-inflammatory activity in a human microglial cell inflammatory model and antinociceptive activity in vivo in an experimental mouse model of neuropathic pain. Finally, computational studies clarified the binding mode of these compounds inside the CB2R, further confirming their bitopic nature.
Collapse
Affiliation(s)
- Francesca Gado
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy
| | - Rebecca Ferrisi
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy
| | - Beatrice Polini
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy.,Department of Pathology, University of Pisa, Pisa 56126, Italy
| | - Kawthar A Mohamed
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | | | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology and Toxicology, University of Florence, Florence 50139, Italy
| | - Sara Carpi
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy.,NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro, Pisa 56126, Italy
| | | | - Lesley A Stevenson
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, U.K
| | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy.,CISUP, Centre for Instrumentation Sharing Pisa University, Lungarno Pacinotti 43, Pisa 56126, Italy
| | | | - Paola Nieri
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy
| | | | - Roger G Pertwee
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, U.K
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology and Toxicology, University of Florence, Florence 50139, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology and Toxicology, University of Florence, Florence 50139, Italy
| | - Grazia Chiellini
- Department of Pathology, University of Pisa, Pisa 56126, Italy.,CISUP, Centre for Instrumentation Sharing Pisa University, Lungarno Pacinotti 43, Pisa 56126, Italy
| | - Robert B Laprairie
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada.,Department of Pharmacology, College of Medicine, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Clementina Manera
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy.,CISUP, Centre for Instrumentation Sharing Pisa University, Lungarno Pacinotti 43, Pisa 56126, Italy
| |
Collapse
|
110
|
Uba AI, Aluwala H, Liu H, Wu C. Elucidation of partial activation of cannabinoid receptor type 2 and identification of potential partial agonists: Molecular dynamics simulation and structure-based virtual screening. Comput Biol Chem 2022; 99:107723. [PMID: 35850049 DOI: 10.1016/j.compbiolchem.2022.107723] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/27/2022] [Accepted: 07/01/2022] [Indexed: 01/14/2023]
Abstract
Cannabinoid receptor type 2 (CB2R) is a member of the class A G protein-coupled receptor (GPCRs) family and a component of the endocannabinoid system that is modulated by the psychoactive chemical from Cannabis sativa, partial agonist Δ9-tetrahydrocannabinol (Δ9-THC). Selective activation of CB2R allows for the treatment of inflammatory and immune-related conditions without the psychotropic effects of CB1R. While CB2R-selective agonists are available, CB2R partial agonists are scarce. Hence, the pharmacological difference between CB2R full agonists and partial agonists remains to be deciphered, prompting the search for novel partial agonists. Here, using an induced-fit docking approach, we built a partial agonist Δ9-THC bound CB2R system from the inactive CB2R structure (PDB ID: 5ZTY) and performed microsecond molecular dynamics (MD) simulations. The simulations reveal an upward shift of the "toggle switch" W6.48(258) and minor outward movement of the transmembrane helix 6 (TM6). Dynamic network model identifies a possible communication path between the ligand and the toggle switch" W6.48(258). Furthermore, to identify potential CB2R partial agonists, we conducted structure-based virtual screening of ZINC15 "Druglike" library containing 17,900742 compounds against 3 conformations derived from MD simulation of CB2R complexed with partial agonist Δ9-THC using Glide virtual screening protocol comprising various filters with increasing accuracy. Nine diverse compounds predicted to have high MM-GBSA binding energy scores and good ADMET properties (including high gastrointestinal absorption and low toxicity) are proposed as potential CB2R partial agonists.
Collapse
Affiliation(s)
- Abdullahi Ibrahim Uba
- Complex Systems Division, Beijing Computational Science Research Center, Beijing 100193, China
| | - Harika Aluwala
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA
| | - Haiguang Liu
- Complex Systems Division, Beijing Computational Science Research Center, Beijing 100193, China.
| | - Chun Wu
- College of Science and Mathematics, Rowan University, Glassboro, NJ 08028, USA.
| |
Collapse
|
111
|
Yue Y, Liu L, Wu LJ, Wu Y, Wang L, Li F, Liu J, Han GW, Chen B, Lin X, Brouillette RL, Breault É, Longpré JM, Shi S, Lei H, Sarret P, Stevens RC, Hanson MA, Xu F. Structural insight into apelin receptor-G protein stoichiometry. Nat Struct Mol Biol 2022; 29:688-697. [PMID: 35817871 DOI: 10.1038/s41594-022-00797-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/26/2022] [Indexed: 11/09/2022]
Abstract
The technique of cryogenic-electron microscopy (cryo-EM) has revolutionized the field of membrane protein structure and function with a focus on the dominantly observed molecular species. This report describes the structural characterization of a fully active human apelin receptor (APJR) complexed with heterotrimeric G protein observed in both 2:1 and 1:1 stoichiometric ratios. We use cryo-EM single-particle analysis to determine the structural details of both species from the same sample preparation. Protein preparations, in the presence of the endogenous peptide ligand ELA or a synthetic small molecule, both demonstrate these mixed stoichiometric states. Structural differences in G protein engagement between dimeric and monomeric APJR suggest a role for the stoichiometry of G protein-coupled receptor- (GPCR-)G protein coupling on downstream signaling and receptor pharmacology. Furthermore, a small, hydrophobic dimer interface provides a starting framework for additional class A GPCR dimerization studies. Together, these findings uncover a mechanism of versatile regulation through oligomerization by which GPCRs can modulate their signaling.
Collapse
Affiliation(s)
- Yang Yue
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Lier Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Li-Jie Wu
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Yiran Wu
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Ling Wang
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Fei Li
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Junlin Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Gye-Won Han
- Departments of Biological Sciences and Chemistry, Bridge Institute, University of Southern California, Los Angeles, CA, USA
| | - Bo Chen
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Xi Lin
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Rebecca L Brouillette
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institute of Pharmacology at Sherbrooke, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Émile Breault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institute of Pharmacology at Sherbrooke, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jean-Michel Longpré
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institute of Pharmacology at Sherbrooke, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Songting Shi
- Structure Therapeutics, South San Francisco, CA, USA
| | - Hui Lei
- Structure Therapeutics, South San Francisco, CA, USA
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institute of Pharmacology at Sherbrooke, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Raymond C Stevens
- iHuman Institute, ShanghaiTech University, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,Structure Therapeutics, South San Francisco, CA, USA
| | | | - Fei Xu
- iHuman Institute, ShanghaiTech University, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
112
|
Bondar AN. Graphs of Hydrogen-Bond Networks to Dissect Protein Conformational Dynamics. J Phys Chem B 2022; 126:3973-3984. [PMID: 35639610 DOI: 10.1021/acs.jpcb.2c00200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Dynamic hydrogen bonds and hydrogen-bond networks are ubiquitous in proteins and protein complexes. Functional roles that have been assigned to hydrogen-bond networks include structural plasticity for protein function, allosteric conformational coupling, long-distance proton transfers, and transient storage of protons. Advances in structural biology provide invaluable insights into architectures of large proteins and protein complexes of direct interest to human physiology and disease, including G Protein Coupled Receptors (GPCRs) and the SARS-Covid-19 spike protein S, and give rise to the challenge of how to identify those interactions that are more likely to govern protein dynamics. This Perspective discusses applications of graph-based algorithms to dissect dynamical hydrogen-bond networks of protein complexes, with illustrations for GPCRs and spike protein S. H-bond graphs provide an overview of sites in GPCR structures where hydrogen-bond dynamics would be required to assemble longer-distance networks between functionally important motifs. In the case of spike protein S, graphs identify regions of the protein where hydrogen bonds rearrange during the reaction cycle and where local hydrogen-bond networks likely change in a virus variant of concern.
Collapse
Affiliation(s)
- Ana-Nicoleta Bondar
- University of Bucharest, Faculty of Physics, Str. Atomiştilor 405, 077125 Bucharest-Măgurele, Romania.,Institute for Neuroscience and Medicine and Institute for Advanced Simulations (IAS-5/INM-9), Computational Biomedicine, Forschungszentrum Jülich, 52425 Jülich, Germany
| |
Collapse
|
113
|
Huang S, Xu P, Shen DD, Simon IA, Mao C, Tan Y, Zhang H, Harpsøe K, Li H, Zhang Y, You C, Yu X, Jiang Y, Zhang Y, Gloriam DE, Xu HE. GPCRs steer G i and G s selectivity via TM5-TM6 switches as revealed by structures of serotonin receptors. Mol Cell 2022; 82:2681-2695.e6. [PMID: 35714614 DOI: 10.1016/j.molcel.2022.05.031] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 04/27/2022] [Accepted: 05/26/2022] [Indexed: 01/23/2023]
Abstract
Serotonin (or 5-hydroxytryptamine, 5-HT) is an important neurotransmitter that activates 12 different G protein-coupled receptors (GPCRs) through selective coupling of Gs, Gi, or Gq proteins. The structural basis for G protein subtype selectivity by these GPCRs remains elusive. Here, we report the structures of the serotonin receptors 5-HT4, 5-HT6, and 5-HT7 with Gs, and 5-HT4 with Gi1. The structures reveal that transmembrane helices TM5 and TM6 alternate lengths as a macro-switch to determine receptor's selectivity for Gs and Gi, respectively. We find that the macro-switch by the TM5-TM6 length is shared by class A GPCR-G protein structures. Furthermore, we discover specific residues within TM5 and TM6 that function as micro-switches to form specific interactions with Gs or Gi. Together, these results present a common mechanism of Gs versus Gi protein coupling selectivity or promiscuity by class A GPCRs and extend the basis of ligand recognition at serotonin receptors.
Collapse
Affiliation(s)
- Sijie Huang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Peiyu Xu
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dan-Dan Shen
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China; MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Icaro A Simon
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; SARomics Biostructures AB, Scheelevägen 2, 223 63 Lund, Sweden; Present address: Vrije Universiteit Amsterdam, Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands
| | - Chunyou Mao
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China; MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yangxia Tan
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Huibing Zhang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China; MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Kasper Harpsøe
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Huadong Li
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yumu Zhang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chongzhao You
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuekui Yu
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Cryo-Electron Microscopy Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yi Jiang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Zhang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China; MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China.
| | - David E Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| | - H Eric Xu
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
114
|
Structural insights into human brain-gut peptide cholecystokinin receptors. Cell Discov 2022; 8:55. [PMID: 35672283 PMCID: PMC9174195 DOI: 10.1038/s41421-022-00420-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 05/11/2022] [Indexed: 11/26/2022] Open
Abstract
The intestinal hormone and neuromodulator cholecystokinin (CCK) receptors CCK1R and CCK2R act as a signaling hub in brain–gut axis, mediating digestion, emotion, and memory regulation. CCK receptors exhibit distinct preferences for ligands in different posttranslational modification (PTM) states. CCK1R couples to Gs and Gq, whereas CCK2R primarily couples to Gq. Here we report the cryo-electron microscopy (cryo-EM) structures of CCK1R–Gs signaling complexes liganded either by sulfated cholecystokinin octapeptide (CCK-8) or a CCK1R-selective small-molecule SR146131, and CCK2R–Gq complexes stabilized by either sulfated CCK-8 or a CCK2R-selective ligand gastrin-17. Our structures reveal a location-conserved yet charge-distinct pocket discriminating the effects of ligand PTM states on receptor subtype preference, the unique pocket topology underlying selectivity of SR146131 and gastrin-17, the conformational changes in receptor activation, and key residues contributing to G protein subtype specificity, providing multiple structural templates for drug design targeting the brain–gut axis.
Collapse
|
115
|
Molecular mechanism of allosteric modulation for the cannabinoid receptor CB1. Nat Chem Biol 2022; 18:831-840. [DOI: 10.1038/s41589-022-01038-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 04/13/2022] [Indexed: 02/07/2023]
|
116
|
Liddle I, Glass M, Tyndall JDA, Vernall AJ. Covalent cannabinoid receptor ligands - structural insight and selectivity challenges. RSC Med Chem 2022; 13:497-510. [PMID: 35694688 PMCID: PMC9132230 DOI: 10.1039/d2md00006g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/31/2022] [Indexed: 11/21/2022] Open
Abstract
X-ray crystallography and cryogenic electronic microscopy have provided significant advancement in the knowledge of GPCR structure and have allowed the rational design of GPCR ligands. The class A GPCRs cannabinoid receptor type 1 and type 2 are implicated in many pathophysiological processes and thus rational design of drug and tool compounds is of great interest. Recent structural insight into cannabinoid receptors has already led to a greater understanding of ligand binding sites and receptor residues that likely contribute to ligand selectivity. Herein, classes of heterocyclic covalent cannabinoid receptor ligands are reviewed in light of the recent advances in structural knowledge of cannabinoid receptors, with particular discussion regarding covalent ligand selectivity and rationale design.
Collapse
Affiliation(s)
- Ian Liddle
- Department of Chemistry, University of Otago Dunedin New Zealand +64 3 479 5214
| | - Michelle Glass
- Department of Pharmacology and Toxicology, University of Otago Dunedin New Zealand
| | | | - Andrea J Vernall
- Department of Chemistry, University of Otago Dunedin New Zealand +64 3 479 5214
| |
Collapse
|
117
|
Xiao W, Chen Y. TRPV1 in male reproductive system: focus on sperm function. Mol Cell Biochem 2022; 477:2567-2579. [PMID: 35595954 DOI: 10.1007/s11010-022-04469-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 05/04/2022] [Indexed: 12/21/2022]
Abstract
The transient receptor potential vanilloid 1 (TRPV1) is a receptor used to perceive external noxious stimuli and participates in the regulation of various pathophysiological mechanisms in vivo by integrating multiple signals. The explosive growth in knowledge of TRPV1 stemmed from research on neuronal pain and heat sensation over the last decades and is being expanded tremendously in peripheral tissue research. The discovery that TRPV1 is functionally active in male animal and human reproductive tissues have attracted increasing attention in recent years. Indeed, many studies have indicated that TRPV1 is an endocannabinoid receptor that mediates Anandamide's regulation of sperm function. Other characteristics of the TRPV1 channel itself, such as calcium penetration and temperature sensitivity, have also been investigated, especially the possibility that TRPV1 could act as a mediator for sperm thermotaxis. In addition, some reproductive diseases appear to be related to the protective effects of TRPV1 on oxidative stress and heat stress. A better understanding of TRPV1 in these areas should provide strategies for tackling male infertility. This paper is the first to review the expression and mechanism of TRPV1 in the male reproductive system from molecular and cellular perspectives. A focus is given on sperm function, including calcium homeostasis, crosstalk with endocannabinoid system, participation in cholesterol-related sperm maturation, and thermotaxis, hoping to capture the current situation of this rapidly developing field.
Collapse
Affiliation(s)
- Wanglong Xiao
- Institute of Life Science and School of Life Science, Nanchang University, No. 999 Xuefu Avenue, Honggutan District, Nanchang, 330031, Jiangxi, People's Republic of China
| | - Ying Chen
- Institute of Life Science and School of Life Science, Nanchang University, No. 999 Xuefu Avenue, Honggutan District, Nanchang, 330031, Jiangxi, People's Republic of China.
- Key Laboratory of Reproductive Physiology and Pathology in Jiangxi Province, Nanchang, Jiangxi, People's Republic of China.
| |
Collapse
|
118
|
Gazzi T, Brennecke B, Atz K, Korn C, Sykes D, Forn-Cuni G, Pfaff P, Sarott RC, Westphal MV, Mostinski Y, Mach L, Wasinska-Kalwa M, Weise M, Hoare BL, Miljuš T, Mexi M, Roth N, Koers EJ, Guba W, Alker A, Rufer AC, Kusznir EA, Huber S, Raposo C, Zirwes EA, Osterwald A, Pavlovic A, Moes S, Beck J, Nettekoven M, Benito-Cuesta I, Grande T, Drawnel F, Widmer G, Holzer D, van der Wel T, Mandhair H, Honer M, Fingerle J, Scheffel J, Broichhagen J, Gawrisch K, Romero J, Hillard CJ, Varga ZV, van der Stelt M, Pacher P, Gertsch J, Ullmer C, McCormick PJ, Oddi S, Spaink HP, Maccarrone M, Veprintsev DB, Carreira EM, Grether U, Nazaré M. Detection of cannabinoid receptor type 2 in native cells and zebrafish with a highly potent, cell-permeable fluorescent probe. Chem Sci 2022; 13:5539-5545. [PMID: 35694350 PMCID: PMC9116301 DOI: 10.1039/d1sc06659e] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/22/2022] [Indexed: 12/16/2022] Open
Abstract
Despite its essential role in the (patho)physiology of several diseases, CB2R tissue expression profiles and signaling mechanisms are not yet fully understood. We report the development of a highly potent, fluorescent CB2R agonist probe employing structure-based reverse design. It commences with a highly potent, preclinically validated ligand, which is conjugated to a silicon-rhodamine fluorophore, enabling cell permeability. The probe is the first to preserve interspecies affinity and selectivity for both mouse and human CB2R. Extensive cross-validation (FACS, TR-FRET and confocal microscopy) set the stage for CB2R detection in endogenously expressing living cells along with zebrafish larvae. Together, these findings will benefit clinical translatability of CB2R based drugs. Detection and visualization of the cannabinoid receptor type 2 by a cell-permeable high affinity fluorescent probe platform enables tracing receptor trafficking in live cells and in zebrafish.![]()
Collapse
Affiliation(s)
- Thais Gazzi
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Benjamin Brennecke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Kenneth Atz
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Claudia Korn
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - David Sykes
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK.,United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | | | - Patrick Pfaff
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich Vladimir-Prelog-Weg 3 8093 Zürich Switzerland
| | - Roman C Sarott
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich Vladimir-Prelog-Weg 3 8093 Zürich Switzerland
| | - Matthias V Westphal
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich Vladimir-Prelog-Weg 3 8093 Zürich Switzerland
| | - Yelena Mostinski
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Leonard Mach
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Malgorzata Wasinska-Kalwa
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Marie Weise
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Bradley L Hoare
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK.,United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | - Tamara Miljuš
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK.,United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | - Maira Mexi
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK.,United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | - Nicolas Roth
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London London EC1M 6BQ England UK
| | - Eline J Koers
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK.,United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | - Wolfgang Guba
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - André Alker
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Arne C Rufer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Eric A Kusznir
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Sylwia Huber
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Catarina Raposo
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Elisabeth A Zirwes
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Anja Osterwald
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Anto Pavlovic
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Svenja Moes
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Jennifer Beck
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Matthias Nettekoven
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Irene Benito-Cuesta
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria Pozuelo de Alarcón 28223 Madrid Spain
| | - Teresa Grande
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria Pozuelo de Alarcón 28223 Madrid Spain
| | - Faye Drawnel
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Gabriella Widmer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Daniela Holzer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Tom van der Wel
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University 2333 CC Leiden the Netherlands
| | - Harpreet Mandhair
- Institute of Biochemistry and Molecular Medicine, University of Bern 3012 Bern Switzerland
| | - Michael Honer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Jürgen Fingerle
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Jörg Scheffel
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin Berlin Germany.,Allergology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP Berlin Germany
| | - Johannes Broichhagen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| | - Klaus Gawrisch
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health Rockville MD 20852 USA
| | - Julián Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria Pozuelo de Alarcón 28223 Madrid Spain
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology, Neuroscience Research Center, Medical College of Wisconsin Milwaukee WI 53226 USA
| | - Zoltan V Varga
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health Rockville MD 20852 USA.,HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University 1085 Budapest Hungary
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University 2333 CC Leiden the Netherlands
| | - Pal Pacher
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health Rockville MD 20852 USA
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, University of Bern 3012 Bern Switzerland
| | - Christoph Ullmer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Peter J McCormick
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London London EC1M 6BQ England UK
| | - Sergio Oddi
- Faculty of Veterinary Medicine, University of Teramo 64100 Teramo European Italy.,European Center for Brain Research (CERC), Santa Lucia Foundation 00179 Rome Italy
| | - Herman P Spaink
- Leiden University Einsteinweg 55 2333 CC Leiden the Netherlands
| | - Mauro Maccarrone
- European Center for Brain Research (CERC), Santa Lucia Foundation 00179 Rome Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila 67100 L'Aquila Italy
| | - Dmitry B Veprintsev
- Faculty of Medicine & Health Sciences, University of Nottingham Nottingham NG7 2UH England UK.,United Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham Midlands England UK
| | - Erick M Carreira
- Laboratorium für Organische Chemie, Eidgenössische Technische Hochschule Zürich Vladimir-Prelog-Weg 3 8093 Zürich Switzerland
| | - Uwe Grether
- Roche Pharma Research & Early Development, Roche Innovation Center Basel F. Hoffmann-La Roche Ltd. 4070 Basel Switzerland
| | - Marc Nazaré
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch 13125 Berlin Germany
| |
Collapse
|
119
|
Kevin RC, Mirlohi S, Manning JJ, Boyd R, Cairns EA, Ametovski A, Lai F, Luo JL, Jorgensen W, Ellison R, Gerona RR, Hibbs DE, McGregor IS, Glass M, Connor M, Bladen C, Zamponi GW, Banister SD. Putative Synthetic Cannabinoids MEPIRAPIM, 5F-BEPIRAPIM (NNL-2), and Their Analogues Are T-Type Calcium Channel (Ca V3) Inhibitors. ACS Chem Neurosci 2022; 13:1395-1409. [PMID: 35442021 DOI: 10.1021/acschemneuro.1c00822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Synthetic cannabinoid receptor agonists (SCRAs) are a large and growing class of new psychoactive substances (NPSs). Two recently identified compounds, MEPIRAPIM and 5F-BEPIRAPIM (NNL-2), have not been confirmed as agonists of either cannabinoid receptor subtype but share structural similarities with both SCRAs and a class of T-type calcium channel (CaV3) inhibitors under development as new treatments for epilepsy and pain. In this study, MEPIRAPIM and 5F-BEPIRAPIM and 10 systematic analogues were synthesized, analytically characterized, and pharmacologically evaluated using in vitro cannabinoid receptor and CaV3 assays. Several compounds showed micromolar affinities for CB1 and/or CB2, with several functioning as low potency agonists of CB1 and CB2 in a membrane potential assay. 5F-BEPIRAPIM and four other derivatives were identified as potential CaV3 inhibitors through a functional calcium flux assay (>70% inhibition), which was further confirmed using whole-cell patch-clamp electrophysiology. Additionally, MEPIRAPIM and 5F-BEPIRAPIM were evaluated in vivo using a cannabimimetic mouse model. Despite detections of MEPIRAPIM and 5F-BEPIRAPIM in the NPS market, only the highest MEPIRAPIM dose (30 mg/kg) elicited a mild hypothermic response in mice, with no hypothermia observed for 5F-BEPIRAPIM, suggesting minimal central CB1 receptor activity.
Collapse
Affiliation(s)
- Richard C. Kevin
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, NSW 2050, Australia
- School of Pharmacy, The University of Sydney, NSW 2006, Australia
| | - Somayeh Mirlohi
- Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW 2109, Australia
| | - Jamie J. Manning
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| | - Rochelle Boyd
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, NSW 2050, Australia
- School of Chemistry, The University of Sydney, NSW 2006, Australia
| | - Elizabeth A. Cairns
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, NSW 2050, Australia
- School of Psychology, The University of Sydney, NSW 2006, Australia
| | - Adam Ametovski
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, NSW 2050, Australia
- School of Chemistry, The University of Sydney, NSW 2006, Australia
| | - Felcia Lai
- School of Pharmacy, The University of Sydney, NSW 2006, Australia
| | - Jia Lin Luo
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, NSW 2050, Australia
- School of Psychology, The University of Sydney, NSW 2006, Australia
| | | | - Ross Ellison
- Clinical Toxicology and Environmental Biomonitoring Laboratory, University of California, San Francisco, California 94143, United States
| | - Roy R. Gerona
- Clinical Toxicology and Environmental Biomonitoring Laboratory, University of California, San Francisco, California 94143, United States
| | - David E. Hibbs
- School of Pharmacy, The University of Sydney, NSW 2006, Australia
| | - Iain S. McGregor
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, NSW 2050, Australia
- School of Psychology, The University of Sydney, NSW 2006, Australia
| | - Michelle Glass
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| | - Mark Connor
- Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW 2109, Australia
| | - Chris Bladen
- Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW 2109, Australia
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Gerald W. Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Samuel D. Banister
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, NSW 2050, Australia
- School of Chemistry, The University of Sydney, NSW 2006, Australia
| |
Collapse
|
120
|
De novo design of a transcription factor for a progesterone biosensor. Biosens Bioelectron 2022; 203:113897. [DOI: 10.1016/j.bios.2021.113897] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022]
|
121
|
Markham J, Sparkes E, Boyd R, Chen S, Manning JJ, Finlay D, Lai F, McGregor E, Maloney CJ, Gerona RR, Connor M, McGregor IS, Hibbs DE, Glass M, Kevin RC, Banister SD. Defining Steric Requirements at CB 1 and CB 2 Cannabinoid Receptors Using Synthetic Cannabinoid Receptor Agonists 5F-AB-PINACA, 5F-ADB-PINACA, PX-1, PX-2, NNL-1, and Their Analogues. ACS Chem Neurosci 2022; 13:1281-1295. [PMID: 35404067 DOI: 10.1021/acschemneuro.2c00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Synthetic cannabinoid receptor agonists (SCRAs) are a diverse class of new psychoactive substances (NPS). They commonly comprise N-alkylated indole, indazole, or 7-azaindole scaffolds with amide-linked pendant amino acid groups. To explore the contribution of the amino acid side chain to the cannabinoid pharmacology of SCRA NPS, a systematic library of side chain-modified SCRAs was prepared based on the recent detections of amino acid derivatives 17 (5F-AB-PINACA), 18 (5F-ADB-PINACA), 15 (PX-1), 19 (PX-2), and 20 (NNL-1). In vitro binding affinities and functional activities at cannabinoid type 1 and 2 receptors (CB1 and CB2, respectively) were determined for all the library members using radioligand competition experiments and a fluorescence-based membrane potential assay. Binding affinities and functional activities varied widely across compounds (Ki = 0.32 to >10 000 nM, EC50 = 0.24-1259 nM), with several clear structure-activity relationships (SARs) emerging. Affinity and potency at CB1 changed as a function of the heterocyclic core (indazole > indole > 7-azaindole) and the pendant amino acid side chain (tert-butyl > iso-propyl > iso-butyl > benzyl > ethyl > methyl > hydrogen). Ensemble docking at CB1 revealed a clear steric basis for observed SAR trends. Interestingly, although 15 (PX-1) and 19 (PX-2) have been detected in recreational drug markets, they failed to induce centrally CB1-mediated effects (e.g., hypothermia) in mice using radiobiotelemetry. Together, these data provide insights regarding structural contributions to the cannabimimetic profiles of 17 (5F-AB-PINACA), 18 (5F-ADB-PINACA), 15 (PX-1), 19 (PX-2), 20 (NNL-1), and other SCRA NPS.
Collapse
Affiliation(s)
- Jack Markham
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney 2050, New South Wales, Australia
- School of Chemistry, The University of Sydney, Sydney 2006, New South Wales, Australia
| | - Eric Sparkes
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney 2050, New South Wales, Australia
- School of Chemistry, The University of Sydney, Sydney 2006, New South Wales, Australia
| | - Rochelle Boyd
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney 2050, New South Wales, Australia
- School of Chemistry, The University of Sydney, Sydney 2006, New South Wales, Australia
| | - Shuli Chen
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| | - Jamie J. Manning
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| | - David Finlay
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| | - Felcia Lai
- School of Pharmacy, The University of Sydney, Sydney 2006, New South Wales, Australia
| | - Eila McGregor
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney 2050, New South Wales, Australia
- School of Psychology, The University of Sydney, Sydney 2005, New South Wales, Australia
| | - Callan J. Maloney
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney 2050, New South Wales, Australia
- School of Chemistry, The University of Sydney, Sydney 2006, New South Wales, Australia
| | - Roy R. Gerona
- Clinical Toxicology and Environmental Biomonitoring Laboratory, University of California, San Francisco, California 94143, United States
| | - Mark Connor
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney 2109, New South Wales, Australia
| | - Iain S. McGregor
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney 2050, New South Wales, Australia
- School of Psychology, The University of Sydney, Sydney 2005, New South Wales, Australia
| | - David E. Hibbs
- School of Pharmacy, The University of Sydney, Sydney 2006, New South Wales, Australia
| | - Michelle Glass
- Department of Pharmacology and Toxicology, University of Otago, Dunedin 9016, New Zealand
| | - Richard C. Kevin
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney 2050, New South Wales, Australia
- School of Pharmacy, The University of Sydney, Sydney 2006, New South Wales, Australia
| | - Samuel D. Banister
- The Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney 2050, New South Wales, Australia
- School of Chemistry, The University of Sydney, Sydney 2006, New South Wales, Australia
| |
Collapse
|
122
|
Bassani D, Pavan M, Federico S, Spalluto G, Sturlese M, Moro S. The Multifaceted Role of GPCRs in Amyotrophic Lateral Sclerosis: A New Therapeutic Perspective? Int J Mol Sci 2022; 23:4504. [PMID: 35562894 PMCID: PMC9106011 DOI: 10.3390/ijms23094504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/15/2022] [Accepted: 04/15/2022] [Indexed: 02/05/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a degenerating disease involving the motor neurons, which causes a progressive loss of movement ability, usually leading to death within 2 to 5 years from the diagnosis. Much effort has been put into research for an effective therapy for its eradication, but still, no cure is available. The only two drugs approved for this pathology, Riluzole and Edaravone, are onlyable to slow down the inevitable disease progression. As assessed in the literature, drug targets such as protein kinases have already been extensively examined as potential drug targets for ALS, with some molecules already in clinical trials. Here, we focus on the involvement of another very important and studied class of biological entities, G protein-coupled receptors (GPCRs), in the onset and progression of ALS. This workaimsto give an overview of what has been already discovered on the topic, providing useful information and insights that can be used by scientists all around the world who are putting efforts into the fight against this very important neurodegenerating disease.
Collapse
Affiliation(s)
- Davide Bassani
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| | - Matteo Pavan
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| | - Stephanie Federico
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy; (S.F.); (G.S.)
| | - Giampiero Spalluto
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy; (S.F.); (G.S.)
| | - Mattia Sturlese
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (D.B.); (M.P.); (M.S.)
| |
Collapse
|
123
|
Xiong X, Chen S, Shen J, You H, Yang H, Yan C, Fang Z, Zhang J, Cai X, Dong X, Kang T, Li W, Zhou P. Cannabis suppresses antitumor immunity by inhibiting JAK/STAT signaling in T cells through CNR2. Signal Transduct Target Ther 2022; 7:99. [PMID: 35383142 PMCID: PMC8983672 DOI: 10.1038/s41392-022-00918-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 01/15/2022] [Accepted: 01/25/2022] [Indexed: 11/09/2022] Open
Abstract
The combination of immune checkpoint blockade (ICB) with chemotherapy significantly improves clinical benefit of cancer treatment. Since chemotherapy is often associated with adverse events, concomitant treatment with drugs managing side effects of chemotherapy is frequently used in the combination therapy. However, whether these ancillary drugs could impede immunotherapy remains unknown. Here, we showed that ∆9-tetrahydrocannabinol (THC), the key ingredient of drugs approved for the treatment of chemotherapy-caused nausea, reduced the therapeutic effect of PD-1 blockade. The endogenous cannabinoid anandamide (AEA) also impeded antitumor immunity, indicating an immunosuppressive role of the endogenous cannabinoid system (ECS). Consistently, high levels of AEA in the sera were associated with poor overall survival in cancer patients. We further found that cannabinoids impaired the function of tumor-specific T cells through CNR2. Using a knock-in mouse model expressing a FLAG-tagged Cnr2 gene, we discovered that CNR2 binds to JAK1 and inhibits the downstream STAT signaling in T cells. Taken together, our results unveiled a novel mechanism of the ECS-mediated suppression on T-cell immunity against cancer, and suggest that cannabis and cannabinoid drugs should be avoided during immunotherapy.
Collapse
Affiliation(s)
- Xinxin Xiong
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510515, China
| | - Siyu Chen
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Key Laboratory of Laboratory Animals, Guangzhou, 510663, China
| | - Jianfei Shen
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, 317000, China
| | - Hua You
- Affiliated Cancer Hospital &Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Han Yang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, China
| | - Ziqian Fang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jianeng Zhang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiuyu Cai
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xingjun Dong
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Tiebang Kang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wende Li
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Key Laboratory of Laboratory Animals, Guangzhou, 510663, China.
| | - Penghui Zhou
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
124
|
Wu Y, Li X, Hua T, Liu ZJ, Liu H, Zhao S. MD Simulations Revealing Special Activation Mechanism of Cannabinoid Receptor 1. Front Mol Biosci 2022; 9:860035. [PMID: 35425811 PMCID: PMC9004671 DOI: 10.3389/fmolb.2022.860035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/28/2022] [Indexed: 11/24/2022] Open
Abstract
Cannabinoid receptor 1 (CB1) is a G protein-coupled receptor (GPCR) that is gaining much interest for its regulating role in the central nervous system and its value as a drug target. Structures of CB1 in inactive and active states have revealed conformational change details that are not common in other GPCRs. Here, we performed molecular dynamics simulations of CB1 in different ligand binding states and with mutations to reveal its activation mechanism. The conformational change of the “twin toggle switch” residues F2003.36 and W3566.48 that correlates with ligand efficacy is identified as a key barrier step in CB1 activation. Similar conformational change of residues 3.36/6.48 is also observed in melanocortin receptor 4, showing this “twin toggle switch” residue pair is crucial for the activation of multiple GPCR members.
Collapse
Affiliation(s)
- Yiran Wu
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Xuanxuan Li
- Complex Systems Division, Beijing Computational Science Research Center, Beijing, China
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhi-Jie Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Haiguang Liu
- Complex Systems Division, Beijing Computational Science Research Center, Beijing, China
- *Correspondence: Haiguang Liu, ; Suwen Zhao,
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- *Correspondence: Haiguang Liu, ; Suwen Zhao,
| |
Collapse
|
125
|
Creanza TM, Lamanna G, Delre P, Contino M, Corriero N, Saviano M, Mangiatordi GF, Ancona N. DeLA-Drug: A Deep Learning Algorithm for Automated Design of Druglike Analogues. J Chem Inf Model 2022; 62:1411-1424. [PMID: 35294184 DOI: 10.1021/acs.jcim.2c00205] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In this paper, we present a deep learning algorithm for automated design of druglike analogues (DeLA-Drug), a recurrent neural network (RNN) model composed of two long short-term memory (LSTM) layers and conceived for data-driven generation of similar-to-bioactive compounds. DeLA-Drug captures the syntax of SMILES strings of more than 1 million compounds belonging to the ChEMBL28 database and, by employing a new strategy called sampling with substitutions (SWS), generates molecules starting from a single user-defined query compound. Remarkably, the algorithm preserves druglikeness and synthetic accessibility of the known bioactive compounds present in the ChEMBL28 repository. The absence of any time-demanding fine-tuning procedure enables DeLA-Drug to perform a fast generation of focused libraries for further high-throughput screening and makes it a suitable tool for performing de novo design even in low-data regimes. To provide a concrete idea of its applicability, DeLA-Drug was applied to the cannabinoid receptor subtype 2 (CB2R), a known target involved in different pathological conditions such as cancer and neurodegeneration. DeLA-Drug, available as a free web platform (http://www.ba.ic.cnr.it/softwareic/deladrugportal/), can help medicinal chemists interested in generating analogues of compounds already available in their laboratories and, for this reason, good candidates for an easy and low-cost synthesis.
Collapse
Affiliation(s)
- Teresa Maria Creanza
- CNR─Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, Via Amendola 122/o, 70126 Bari, Italy
| | - Giuseppe Lamanna
- Chemistry Department, University of Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.,CNR─Institute of Crystallography, Via Amendola 122/o, 70126 Bari, Italy
| | - Pietro Delre
- Chemistry Department, University of Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy.,CNR─Institute of Crystallography, Via Amendola 122/o, 70126 Bari, Italy
| | - Marialessandra Contino
- Department of Pharmacy─Pharmaceutical Sciences, University of Bari "Aldo Moro", via E. Orabona, 4, I-70125 Bari, Italy
| | - Nicola Corriero
- CNR─Institute of Crystallography, Via Amendola 122/o, 70126 Bari, Italy
| | - Michele Saviano
- CNR─Institute of Crystallography, Via Amendola 122/o, 70126 Bari, Italy
| | | | - Nicola Ancona
- CNR─Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, Via Amendola 122/o, 70126 Bari, Italy
| |
Collapse
|
126
|
Lipiński PFJ, Matalińska J. Fentanyl Structure as a Scaffold for Opioid/Non-Opioid Multitarget Analgesics. Int J Mol Sci 2022; 23:ijms23052766. [PMID: 35269909 PMCID: PMC8910985 DOI: 10.3390/ijms23052766] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 11/16/2022] Open
Abstract
One of the strategies in the search for safe and effective analgesic drugs is the design of multitarget analgesics. Such compounds are intended to have high affinity and activity at more than one molecular target involved in pain modulation. In the present contribution we summarize the attempts in which fentanyl or its substructures were used as a μ-opioid receptor pharmacophoric fragment and a scaffold to which fragments related to non-opioid receptors were attached. The non-opioid ‘second’ targets included proteins as diverse as imidazoline I2 binding sites, CB1 cannabinoid receptor, NK1 tachykinin receptor, D2 dopamine receptor, cyclooxygenases, fatty acid amide hydrolase and monoacylglycerol lipase and σ1 receptor. Reviewing the individual attempts, we outline the chemistry, the obtained pharmacological properties and structure-activity relationships. Finally, we discuss the possible directions for future work.
Collapse
|
127
|
El-Atawneh S, Goldblum A. Candidate Therapeutics by Screening for Multitargeting Ligands: Combining the CB2 Receptor With CB1, PPARγ and 5-HT4 Receptors. Front Pharmacol 2022; 13:812745. [PMID: 35295337 PMCID: PMC8918518 DOI: 10.3389/fphar.2022.812745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/13/2022] [Indexed: 12/15/2022] Open
Abstract
In recent years, the cannabinoid type 2 receptor (CB2R) has become a major target for treating many disease conditions. The old therapeutic paradigm of “one disease-one target-one drug” is being transformed to “complex disease-many targets-one drug.” Multitargeting, therefore, attracts much attention as a promising approach. We thus focus on designing single multitargeting agents (MTAs), which have many advantages over combined therapies. Using our ligand-based approach, the “Iterative Stochastic Elimination” (ISE) algorithm, we produce activity models of agonists and antagonists for desired therapeutic targets and anti-targets. These models are used for sequential virtual screening and scoring large libraries of molecules in order to pick top-scored candidates for testing in vitro and in vivo. In this study, we built activity models for CB2R and other targets for combinations that could be used for several indications. Those additional targets are the cannabinoid 1 receptor (CB1R), peroxisome proliferator-activated receptor gamma (PPARγ), and 5-Hydroxytryptamine receptor 4 (5-HT4R). All these models have high statistical parameters and are reliable. Many more CB2R/CBIR agonists were found than combined CB2R agonists with CB1R antagonist activity (by 200 fold). CB2R agonism combined with PPARγ or 5-HT4R agonist activity may be used for treating Inflammatory Bowel Disease (IBD). Combining CB2R agonism with 5-HT4R generates more candidates (14,008) than combining CB2R agonism with agonists for the nuclear receptor PPARγ (374 candidates) from an initial set of ∼2.1 million molecules. Improved enrichment of true vs. false positives may be achieved by requiring a better ISE score cutoff or by performing docking. Those candidates can be purchased and tested experimentally to validate their activity. Further, we performed docking to CB2R structures and found lower statistical performance of the docking (“structure-based”) compared to ISE modeling (“ligand-based”). Therefore, ISE modeling may be a better starting point for molecular discovery than docking.
Collapse
|
128
|
Morales P, Muller C, Jagerovic N, Reggio PH. Targeting CB2 and TRPV1: Computational Approaches for the Identification of Dual Modulators. Front Mol Biosci 2022; 9:841190. [PMID: 35281260 PMCID: PMC8914543 DOI: 10.3389/fmolb.2022.841190] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
Both metabotropic (CBRs) and ionotropic cannabinoid receptors (ICRs) have implications in a range of neurological disorders. The metabotropic canonical CBRs CB1 and CB2 are highly implicated in these pathological events. However, selective targeting at CB2 versus CB1 offers optimized pharmacology due to the absence of psychoactive outcomes. The ICR transient receptor potential vanilloid type 1 (TRPV1) has also been reported to play a role in CNS disorders. Thus, activation of both targets, CB2 and TRPV1, offers a promising polypharmacological strategy for the treatment of neurological events including analgesia and neuroprotection. This brief research report aims to identify chemotypes with a potential dual CB2/TRPV1 profile. For this purpose, we have rationalized key structural features for activation and performed virtual screening at both targets using curated chemical libraries.
Collapse
Affiliation(s)
- Paula Morales
- Medicinal Chemistry Institute, Spanish National Research Council, Madrid, Spain
| | - Chanté Muller
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, United States
| | - Nadine Jagerovic
- Medicinal Chemistry Institute, Spanish National Research Council, Madrid, Spain
| | - Patricia H. Reggio
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, United States
| |
Collapse
|
129
|
Franco R, Morales P, Navarro G, Jagerovic N, Reyes-Resina I. The Binding Mode to Orthosteric Sites and/or Exosites Underlies the Therapeutic Potential of Drugs Targeting Cannabinoid CB2 Receptors. Front Pharmacol 2022; 13:852631. [PMID: 35250601 PMCID: PMC8889005 DOI: 10.3389/fphar.2022.852631] [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: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 12/02/2022] Open
Abstract
The classical terms agonists and antagonists for G protein coupled receptors (GPCRs) have often become misleading. Even the biased agonism concept does not describe all the possibilities already demonstrated for GPCRs. The cannabinoid CB2 receptor (CB2R) emerged as a promising target for a variety of diseases. Reasons for such huge potential are centered around the way drugs sit in the orthosteric and/or exosites of the receptor. On the one hand, a given drug in a specific CB2R conformation leads to a signaling cascade that differs qualitatively and/or quantitatively from that triggered by another drug. On the other hand, a given drug may lead to different signaling outputs in two different tissues (or cell contexts) in which the conformation of the receptor is affected by allosteric effects derived from interactions with other proteins or with membrane lipids. This highlights the pharmacological complexity of this receptor and the need to further unravel the binding mode of CB2R ligands in order to fine-tune signaling effects and therapeutic propositions.
Collapse
Affiliation(s)
- Rafael Franco
- CiberNed. Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biolomedicine, Universitat de Barcelona, Barcelona, Spain
- School of Chemistry, Universitat de Barcelona, Barcelona, Spain
- *Correspondence: Rafael Franco,
| | - Paula Morales
- Medicinal Chemistry Institute, Spanish National Research Council, Madrid, Spain
| | - Gemma Navarro
- CiberNed. Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
| | - Nadine Jagerovic
- Medicinal Chemistry Institute, Spanish National Research Council, Madrid, Spain
| | - Irene Reyes-Resina
- CiberNed. Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biolomedicine, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
130
|
Dutta S, Selvam B, Shukla D. Distinct Binding Mechanisms for Allosteric Sodium Ion in Cannabinoid Receptors. ACS Chem Neurosci 2022; 13:379-389. [PMID: 35019279 DOI: 10.1021/acschemneuro.1c00760] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The therapeutic potential of cannabinoid receptors is not fully explored due to psychoactive side effects and lack of selectivity associated with orthosteric ligands. Allosteric modulators have the potential to become selective therapeutics for cannabinoid receptors. Biochemical experiments have shown the effects of the allosteric Na+ binding on cannabinoid receptor activity. However, the Na+ coordination site and binding pathway are still unknown. Here, we perform molecular dynamic simulations to explore Na+ binding in the cannabinoid receptors, CB1 and CB2. Simulations reveal that Na+ binds to the primary binding site from different extracellular sites for CB1 and CB2. A distinct secondary Na+ coordination site is identified in CB1 that is not present in CB2. Furthermore, simulations also show that intracellular Na+ could bind to the Na+ binding site in CB1. Constructed Markov state models show that the standard free energy of Na+ binding is similar to the previously calculated free energy for other class A GPCRs.
Collapse
Affiliation(s)
- Soumajit Dutta
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Balaji Selvam
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Diwakar Shukla
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
- National Center for Supercomputing Applications, University of Illinois, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
- NIH Center for Macromolecular Modeling and Bioinformatics, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
131
|
Mechanistic Origin of Partial Agonism of Tetrahydrocannabinol for Cannabinoid Receptors. J Biol Chem 2022; 298:101764. [PMID: 35227761 PMCID: PMC8965160 DOI: 10.1016/j.jbc.2022.101764] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 01/14/2023] Open
Abstract
Cannabinoid receptor 1 (CB1) is a therapeutically relevant drug target for controlling pain, obesity, and other central nervous system disorders. However, full agonists and antagonists of CB1 have been reported to cause serious side effects in patients. Therefore, partial agonists have emerged as a viable alternative as they can mitigate overstimulation and side effects. One of the key bottlenecks in the design of partial agonists, however, is the lack of understanding of the molecular mechanism of partial agonism itself. In this study, we examine two mechanistic hypotheses for the origin of partial agonism in cannabinoid receptors and predict the mechanistic basis of partial agonism exhibited by Δ9-Tetrahydrocannabinol (THC) against CB1. In particular, we inspect whether partial agonism emerges from the ability of THC to bind in both agonist and antagonist-binding poses or from its ability to only partially activate the receptor. We used extensive molecular dynamics simulations and Markov state modeling to capture the THC binding in both antagonist and agonist-binding poses in the CB1 receptor. Furthermore, we predict that binding of THC in the agonist-binding pose leads to rotation of toggle switch residues and causes partial outward movement of intracellular transmembrane helix 6 (TM6). Our simulations also suggest that the alkyl side chain of THC plays a crucial role in determining partial agonism by stabilizing the ligand in the agonist and antagonist-like poses within the pocket. Taken together, this study provides important insights into the mechanistic origin of the partial agonism of THC.
Collapse
|
132
|
Improved cyclobutyl nabilone analogs as potent CB1 receptor agonists. Eur J Med Chem 2022; 230:114027. [DOI: 10.1016/j.ejmech.2021.114027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 11/18/2022]
|
133
|
Structural basis of the ligand binding and signaling mechanism of melatonin receptors. Nat Commun 2022; 13:454. [PMID: 35075127 PMCID: PMC8786939 DOI: 10.1038/s41467-022-28111-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/05/2022] [Indexed: 12/13/2022] Open
Abstract
Melatonin receptors (MT1 and MT2 in humans) are family A G protein-coupled receptors that respond to the neurohormone melatonin to regulate circadian rhythm and sleep. Numerous efforts have been made to develop drugs targeting melatonin receptors for the treatment of insomnia, circadian rhythm disorder, and cancer. However, designing subtype-selective melatonergic drugs remains challenging. Here, we report the cryo-EM structures of the MT1-Gi signaling complex with 2-iodomelatonin and ramelteon and the MT2-Gi signaling complex with ramelteon. These structures, together with the reported functional data, reveal that although MT1 and MT2 possess highly similar orthosteric ligand-binding pockets, they also display distinctive features that could be targeted to design subtype-selective drugs. The unique structural motifs in MT1 and MT2 mediate structural rearrangements with a particularly wide opening on the cytoplasmic side. Gi is engaged in the receptor core shared by MT1 and MT2 and presents a conformation deviating from those in other Gi complexes. Together, our results provide new clues for designing melatonergic drugs and further insights into understanding the G protein coupling mechanism.
Collapse
|
134
|
Yuan J, Jiang C, Wang J, Chen CJ, Hao Y, Zhao G, Feng Z, Xie XQ. In Silico Prediction and Validation of CB2 Allosteric Binding Sites to Aid the Design of Allosteric Modulators. Molecules 2022; 27:molecules27020453. [PMID: 35056767 PMCID: PMC8781014 DOI: 10.3390/molecules27020453] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/01/2022] [Accepted: 01/03/2022] [Indexed: 11/16/2022] Open
Abstract
Although the 3D structures of active and inactive cannabinoid receptors type 2 (CB2) are available, neither the X-ray crystal nor the cryo-EM structure of CB2-orthosteric ligand-modulator has been resolved, prohibiting the drug discovery and development of CB2 allosteric modulators (AMs). In the present work, we mainly focused on investigating the potential allosteric binding site(s) of CB2. We applied different algorithms or tools to predict the potential allosteric binding sites of CB2 with the existing agonists. Seven potential allosteric sites can be observed for either CB2-CP55940 or CB2-WIN 55,212-2 complex, among which sites B, C, G and K are supported by the reported 3D structures of Class A GPCRs coupled with AMs. Applying our novel algorithm toolset-MCCS, we docked three known AMs of CB2 including Ec2la (C-2), trans-β-caryophyllene (TBC) and cannabidiol (CBD) to each site for further comparisons and quantified the potential binding residues in each allosteric binding site. Sequentially, we selected the most promising binding pose of C-2 in five allosteric sites to conduct the molecular dynamics (MD) simulations. Based on the results of docking studies and MD simulations, we suggest that site H is the most promising allosteric binding site. We plan to conduct bio-assay validations in the future.
Collapse
Affiliation(s)
- Jiayi Yuan
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.Y.); (C.J.); (J.W.); (C.-J.C.); (Y.H.); (G.Z.)
- Department of Pharmaceutical Sciences and National Center of Excellence for Computational Drug Abuse Research, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Chen Jiang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.Y.); (C.J.); (J.W.); (C.-J.C.); (Y.H.); (G.Z.)
- Department of Pharmaceutical Sciences and National Center of Excellence for Computational Drug Abuse Research, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Junmei Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.Y.); (C.J.); (J.W.); (C.-J.C.); (Y.H.); (G.Z.)
- Department of Pharmaceutical Sciences and National Center of Excellence for Computational Drug Abuse Research, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Chih-Jung Chen
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.Y.); (C.J.); (J.W.); (C.-J.C.); (Y.H.); (G.Z.)
- Department of Pharmaceutical Sciences and National Center of Excellence for Computational Drug Abuse Research, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yixuan Hao
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.Y.); (C.J.); (J.W.); (C.-J.C.); (Y.H.); (G.Z.)
- Department of Pharmaceutical Sciences and National Center of Excellence for Computational Drug Abuse Research, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Guangyi Zhao
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.Y.); (C.J.); (J.W.); (C.-J.C.); (Y.H.); (G.Z.)
- Department of Pharmaceutical Sciences and National Center of Excellence for Computational Drug Abuse Research, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.Y.); (C.J.); (J.W.); (C.-J.C.); (Y.H.); (G.Z.)
- Department of Pharmaceutical Sciences and National Center of Excellence for Computational Drug Abuse Research, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence: (Z.F.); (X.-Q.X.)
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.Y.); (C.J.); (J.W.); (C.-J.C.); (Y.H.); (G.Z.)
- Department of Pharmaceutical Sciences and National Center of Excellence for Computational Drug Abuse Research, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence: (Z.F.); (X.-Q.X.)
| |
Collapse
|
135
|
Castro J, Garcia-Caraballo S, Maddern J, Schober G, Lumsden A, Harrington A, Schmiel S, Lindstrom B, Adams J, Brierley SM. Olorinab (APD371), a peripherally acting, highly selective, full agonist of the cannabinoid receptor 2, reduces colitis-induced acute and chronic visceral hypersensitivity in rodents. Pain 2022; 163:e72-e86. [PMID: 33863856 PMCID: PMC8675055 DOI: 10.1097/j.pain.0000000000002314] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/12/2021] [Accepted: 04/02/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Abdominal pain is a key symptom of inflammatory bowel disease and irritable bowel syndrome, for which there are inadequate therapeutic options. We tested whether olorinab-a highly selective, full agonist of the cannabinoid receptor 2 (CB2)-reduced visceral hypersensitivity in models of colitis and chronic visceral hypersensitivity (CVH). In rodents, colitis was induced by intrarectal administration of nitrobenzene sulfonic acid derivatives. Control or colitis animals were administered vehicle or olorinab (3 or 30 mg/kg) twice daily by oral gavage for 5 days, starting 1 day before colitis induction. Chronic visceral hypersensitivity mice were administered olorinab (1, 3, 10, or 30 mg/kg) twice daily by oral gavage for 5 days, starting 24 days after colitis induction. Visceral mechanosensitivity was assessed in vivo by quantifying visceromotor responses (VMRs) to colorectal distension. Ex vivo afferent recordings determined colonic nociceptor firing evoked by mechanical stimuli. Colitis and CVH animals displayed significantly elevated VMRs to colorectal distension and colonic nociceptor hypersensitivity. Olorinab treatment significantly reduced VMRs to control levels in colitis and CVH animals. In addition, olorinab reduced nociceptor hypersensitivity in colitis and CVH states in a concentration- and CB2-dependent manner. By contrast, olorinab did not alter VMRs nor nociceptor responsiveness in control animals. Cannabinoid receptor 2 mRNA was detected in colonic tissue, particularly within epithelial cells, and dorsal root ganglia, with no significant differences between healthy, colitis, and CVH states. These results demonstrate that olorinab reduces visceral hypersensitivity through CB2 agonism in animal models, suggesting that olorinab may provide a novel therapy for inflammatory bowel disease- and irritable bowel syndrome-associated abdominal pain.
Collapse
Affiliation(s)
- Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Sonia Garcia-Caraballo
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Jessica Maddern
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Gudrun Schober
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Amanda Lumsden
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Andrea Harrington
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| | - Shirdi Schmiel
- Arena Pharmaceuticals, Inc, San Diego, CA, United States
| | | | - John Adams
- Arena Pharmaceuticals, Inc, San Diego, CA, United States
| | - Stuart M. Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, North Terrace, Adelaide, South Australia, Australia
| |
Collapse
|
136
|
Krishna Deepak RNV, Verma RK, Hartono YD, Yew WS, Fan H. Recent Advances in Structure, Function, and Pharmacology of Class A Lipid GPCRs: Opportunities and Challenges for Drug Discovery. Pharmaceuticals (Basel) 2021; 15:12. [PMID: 35056070 PMCID: PMC8779880 DOI: 10.3390/ph15010012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/17/2021] [Accepted: 12/17/2021] [Indexed: 01/01/2023] Open
Abstract
Great progress has been made over the past decade in understanding the structural, functional, and pharmacological diversity of lipid GPCRs. From the first determination of the crystal structure of bovine rhodopsin in 2000, much progress has been made in the field of GPCR structural biology. The extraordinary progress in structural biology and pharmacology of GPCRs, coupled with rapid advances in computational approaches to study receptor dynamics and receptor-ligand interactions, has broadened our comprehension of the structural and functional facets of the receptor family members and has helped usher in a modern age of structure-based drug design and development. First, we provide a primer on lipid mediators and lipid GPCRs and their role in physiology and diseases as well as their value as drug targets. Second, we summarize the current advancements in the understanding of structural features of lipid GPCRs, such as the structural variation of their extracellular domains, diversity of their orthosteric and allosteric ligand binding sites, and molecular mechanisms of ligand binding. Third, we close by collating the emerging paradigms and opportunities in targeting lipid GPCRs, including a brief discussion on current strategies, challenges, and the future outlook.
Collapse
Affiliation(s)
- R. N. V. Krishna Deepak
- Bioinformatics Institute, A*STAR, 30 Biopolis Street, Matrix #07-01, Singapore 138671, Singapore; (R.K.V.); (Y.D.H.)
| | - Ravi Kumar Verma
- Bioinformatics Institute, A*STAR, 30 Biopolis Street, Matrix #07-01, Singapore 138671, Singapore; (R.K.V.); (Y.D.H.)
| | - Yossa Dwi Hartono
- Bioinformatics Institute, A*STAR, 30 Biopolis Street, Matrix #07-01, Singapore 138671, Singapore; (R.K.V.); (Y.D.H.)
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore;
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore
| | - Wen Shan Yew
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore;
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore
| | - Hao Fan
- Bioinformatics Institute, A*STAR, 30 Biopolis Street, Matrix #07-01, Singapore 138671, Singapore; (R.K.V.); (Y.D.H.)
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore;
| |
Collapse
|
137
|
Duan W, Sun Y, Wu M, Zhang Z, Zhang T, Wang H, Li F, Yang L, Xu Y, Liu ZJ, Hua T, Nie H, Cheng J. Carbon-silicon switch led to the discovery of novel synthetic cannabinoids with therapeutic effects in a mouse model of multiple sclerosis. Eur J Med Chem 2021; 226:113878. [PMID: 34634742 DOI: 10.1016/j.ejmech.2021.113878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/16/2021] [Accepted: 09/27/2021] [Indexed: 11/17/2022]
Abstract
Cannabinoids are widely studied as therapeutic agents for the treatment of various diseases. Among them, THC and CBD are two important phytocannabinoids which have served as structural templates for the design of synthetic analogs. In this study, we designed and synthesized a variety of novel cannabinoids based on the structural backbones of THC and CBD using the carbon-silicon switch strategy. A dimethyl silyl group was introduced as the tail group and two series of novel compounds were designed and synthesized, which showed a wide range of binding affinity for CB1 and CB2 receptors. Among them, compound 15b was identified as a non-selective CB1 and CB2 agonist and 38b as a selective agonist for the CB2 receptor. Preliminary screening showed that both compounds have improved metabolic stability than their carbon analogs and good in vivo pharmacokinetic profiles. Furthermore, both 15b and 38b significantly alleviated the phenotype of experimental autoimmune encephalomyelitis (EAE) in mice.
Collapse
Affiliation(s)
- Wenwen Duan
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
| | - Ying Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Meng Wu
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
| | - Zhiyuan Zhang
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
| | - Taotao Zhang
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, 650500, China
| | - Huan Wang
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
| | - Fei Li
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
| | - Lingyun Yang
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
| | - Yueming Xu
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
| | - Zhi-Jie Liu
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China; School of Life Sciences and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China; School of Life Sciences and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Hong Nie
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
138
|
Gado F, Ceni C, Ferrisi R, Sbrana G, Stevenson LA, Macchia M, Pertwee RG, Bertini S, Manera C, Ortore G. CB1 receptor binding sites for NAM and PAM: A first approach for studying, new n‑butyl‑diphenylcarboxamides as allosteric modulators. Eur J Pharm Sci 2021; 169:106088. [PMID: 34863873 DOI: 10.1016/j.ejps.2021.106088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 01/01/2023]
Abstract
The development of cannabinoid receptor type-1 (CB1R) modulators has been implicated in multiple pathophysiological events ranging from memory deficits to neurodegenerative disorders among others, even if their central psychiatric side effects such as depression, anxiety, and suicidal tendencies, have limited their clinical use. Thus, the identification of ligands which selectively act on peripheral CB1Rs, is becoming more interesting. A recent study reported a class of peripheral CB1R selective antagonists, characterized by a 5-aryl substituted nicotinamide core. These derivatives have structural similarities with the biphenyl compounds, endowed with CB2R antagonist activity, previously synthesized by our research group. In this work we combined the pharmacophoric portion of both classes, in order to obtain novel CBR antagonists. Among the synthesized compounds rather unexpectedly two compounds of this series, C7 and C10, did not show the radioligand ([3H]CP55940) displacement on CB1R but increased binding (∼ 150%), suggesting a possible allosteric behavior. Computational studies were performed to investigate the role of these compounds in CB1R modulation. The analysis of their binding poses in two different binding cavities of the CB1R surface, revealed a preferred interaction with the experimental binding site for negative allosteric modulators.
Collapse
Affiliation(s)
- Francesca Gado
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy
| | - Costanza Ceni
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy; Doctoral school in Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Rebecca Ferrisi
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy
| | - Giulia Sbrana
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy
| | - Lesley A Stevenson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, AB25 2ZD Aberdeen, Scotland, UK
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy
| | - Roger G Pertwee
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, AB25 2ZD Aberdeen, Scotland, UK
| | - Simone Bertini
- Department of Pharmacy, University of Pisa, 56126 Pisa Italy
| | | | | |
Collapse
|
139
|
Raïch I, Rivas-Santisteban R, Lillo A, Lillo J, Reyes-Resina I, Nadal X, Ferreiro-Vera C, de Medina VS, Majellaro M, Sotelo E, Navarro G, Franco R. Similarities and differences upon binding of naturally occurring Δ 9-tetrahydrocannabinol-derivatives to cannabinoid CB 1 and CB 2 receptors. Pharmacol Res 2021; 174:105970. [PMID: 34758399 DOI: 10.1016/j.phrs.2021.105970] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022]
Abstract
We have here assessed, using Δ9-tetrahydrocannabinol (Δ9-THC) for comparison, the effect of Δ9-tetrahydrocannabinolic acid (Δ9-THCA) and of Δ9-tetrahydrocannabivarin (Δ9-THCV) that is mediated by human versions of CB1, CB2, and CB1-CB2 receptor functional units, expressed in a heterologous system. Binding to the CB1 and CB2 receptors was addressed in living cells by means of a homogeneous assay. A biphasic competition curve for the binding to the CB2 receptor, was obtained for Δ9-THCV in cells expressing the two receptors. Signaling studies included cAMP level determination, activation of the mitogen-activated protein kinase pathway and ß-arrestin recruitment were performed. The signaling triggered by Δ9-THCA and Δ9-THCV via individual receptors or receptor heteromers disclosed differential bias, i.e. the bias observed using a given phytocannabinoid depended on the receptor (CB1, CB2 or CB1-CB2) and on the compound used as reference to calculate the bias factor (Δ9-THC, a selective agonist or a non-selective agonist). These results are consistent with different binding modes leading to differential functional selectivity depending on the agonist structure, and the state (monomeric or heteromeric) of the cannabinoid receptor. In addition, on studying Gi-coupling we showed that Δ9-THCV and Δ9-THCA and Δ9-THCV were able to revert the effect of a selective CB2 receptor agonist, but only Δ9-THCV, and not Δ9-THCA, reverted the effect of arachidonyl-2'-chloroethylamide (ACEA 100 nM) a selective agonist of the CB1 receptor. Overall, these results indicate that cannabinoids may have a variety of binding modes that results in qualitatively different effects depending on the signaling pathway that is engaged upon cannabinoid receptor activation.
Collapse
Affiliation(s)
- Iu Raïch
- Department of Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Spanish National Institute of Health, Carlos iii, 28034 Madrid, Spain
| | - Rafael Rivas-Santisteban
- Department of Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Spanish National Institute of Health, Carlos iii, 28034 Madrid, Spain
| | - Alejandro Lillo
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
| | - Jaume Lillo
- Department of Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Spanish National Institute of Health, Carlos iii, 28034 Madrid, Spain
| | - Irene Reyes-Resina
- Department of Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, 08028 Barcelona, Spain; RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Saxony-Anhalt 39118, Germany
| | - Xavier Nadal
- Ethnophytotech Research & Consulting S.L.U., Córdoba, Spain
| | | | | | - Maria Majellaro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Eddy Sotelo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Gemma Navarro
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain; Institut de Neurociències de la Universitat de Barcelona, Barcelona, Spain.
| | - Rafael Franco
- Department of Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Spanish National Institute of Health, Carlos iii, 28034 Madrid, Spain; School of Chemistry. University of Barcelona, Barcelona, Spain.
| |
Collapse
|
140
|
Yokoi S, Mitsutake A. Characteristic structural difference between inactive and active states of orexin 2 receptor determined using molecular dynamics simulations. Biophys Rev 2021; 14:221-231. [DOI: 10.1007/s12551-021-00862-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
|
141
|
Jannuzzi AT, Yıldız M, Bayrak N, Yıldırım H, Shilkar D, Jayaprakash V, TuYuN AF. Anticancer agents based on Plastoquinone analogs with N-phenylpiperazine: Structure-activity relationship and mechanism of action in breast cancer cells. Chem Biol Interact 2021; 349:109673. [PMID: 34560069 DOI: 10.1016/j.cbi.2021.109673] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/08/2021] [Accepted: 09/20/2021] [Indexed: 11/18/2022]
Abstract
2,3-Dimethyl-1,4-benzoquinones named as Plastoquinone (PQ) analogs have antiproliferative activity and are promising new members of molecules that can be used to cope with cancer. In an attempt to develop effective and potentially safe antiproliferative agents, previously reported twelve Plastoquinone analogs (PQ1-12) have been obtained to understand their antiproliferative profile. All PQ analogs have been selected by the National Cancer Institute (NCI) of Bethesda based on the NCI Developmental Therapeutics Program and tested against the panel of 60 cancer cell lines. Based on those studies, the cytotoxicity of the selected PQ analogs (PQ8, PQ9, PQ11, and PQ12) was determined using four breast cancer cell lines (MCF7, UACC-2087, MDA-MB-231, and MDA-MB-435) and a normal cell line (HaCaT). For better understanding, apoptosis induction, changes in cell proliferation, cell migration, and reactive oxygen species (ROS) generation were investigated for the selected PQ analog (PQ11) on MCF7 and UACC-2087 cell lines. According to the study results, PQ11 showed the most promising anticancer activity against MCF7 cell line through increased oxidative stress and apoptosis and suppression of cell proliferation. Based on the biological activity profile, we hypothesize that PQ11 could be a modulator of the cannabinoid 2 (CB2) receptor. Accordingly, we analyzed molecular level interaction of PQ11 with CB2 receptor through molecular docking simulation and it was also predicted to have a favorable ADMET profile. Overall, our findings suggest that integration of the N-phenylpiperazine moiety can be a good strategy for the structural optimization of PQ analogs as anticancer agents, especially in breast cancer.
Collapse
Affiliation(s)
- Ayse Tarbin Jannuzzi
- Pharmaceutical Toxicology Department, Pharmacy Faculty, Istanbul University, Beyazit, 34116, Istanbul, Turkey
| | - Mahmut Yıldız
- Chemistry Department, Gebze Technical University, Gebze, 41400, Kocaeli, Turkey
| | - Nilüfer Bayrak
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Avcılar, 34320, Istanbul, Turkey
| | - Hatice Yıldırım
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Avcılar, 34320, Istanbul, Turkey
| | - Deepak Shilkar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835 215, Jharkhand, India
| | - Venkatesan Jayaprakash
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835 215, Jharkhand, India
| | - Amaç Fatih TuYuN
- Department of Chemistry, Faculty of Science, Istanbul University, Fatih, Istanbul, Turkey.
| |
Collapse
|
142
|
Faúndez-Parraguez M, Alarcón-Miranda C, Cho YH, Pessoa-Mahana H, Gallardo-Garrido C, Chung H, Faúndez M, Pessoa-Mahana D. New Pyridone-Based Derivatives as Cannabinoid Receptor Type 2 Agonists. Int J Mol Sci 2021; 22:11212. [PMID: 34681877 PMCID: PMC8537746 DOI: 10.3390/ijms222011212] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
The activation of the human cannabinoid receptor type II (CB2R) is known to mediate analgesic and anti-inflammatory processes without the central adverse effects related to cannabinoid receptor type I (CB1R). In this work we describe the synthesis and evaluation of a novel series of N-aryl-2-pyridone-3-carboxamide derivatives tested as human cannabinoid receptor type II (CB2R) agonists. Different cycloalkanes linked to the N-aryl pyridone by an amide group displayed CB2R agonist activity as determined by intracellular [cAMP] levels. The most promising compound 8d exhibited a non-toxic profile and similar potency (EC50 = 112 nM) to endogenous agonists Anandamide (AEA) and 2-Arachidonoylglycerol (2-AG) providing new information for the development of small molecules activating CB2R. Molecular docking studies showed a binding pose consistent with two structurally different agonists WIN-55212-2 and AM12033 and suggested structural requirements on the pyridone substituents that can satisfy the orthosteric pocket and induce an agonist response. Our results provide additional evidence to support the 2-pyridone ring as a suitable scaffold for the design of CB2R agonists and represent a starting point for further optimization and development of novel compounds for the treatment of pain and inflammation.
Collapse
Affiliation(s)
- Manuel Faúndez-Parraguez
- Pharmacy Department, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago 7820436, Chile; (C.A.-M.); (Y.H.C.); (C.G.-G.); (H.C.); (M.F.)
| | - Carlos Alarcón-Miranda
- Pharmacy Department, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago 7820436, Chile; (C.A.-M.); (Y.H.C.); (C.G.-G.); (H.C.); (M.F.)
| | - Young Hwa Cho
- Pharmacy Department, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago 7820436, Chile; (C.A.-M.); (Y.H.C.); (C.G.-G.); (H.C.); (M.F.)
| | - Hernán Pessoa-Mahana
- Organic Chemistry and Physical Chemistry Department, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Olivos 1007, Santiago 7820436, Chile;
| | - Carlos Gallardo-Garrido
- Pharmacy Department, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago 7820436, Chile; (C.A.-M.); (Y.H.C.); (C.G.-G.); (H.C.); (M.F.)
| | - Hery Chung
- Pharmacy Department, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago 7820436, Chile; (C.A.-M.); (Y.H.C.); (C.G.-G.); (H.C.); (M.F.)
| | - Mario Faúndez
- Pharmacy Department, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago 7820436, Chile; (C.A.-M.); (Y.H.C.); (C.G.-G.); (H.C.); (M.F.)
| | - David Pessoa-Mahana
- Pharmacy Department, Faculty of Chemistry, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago 7820436, Chile; (C.A.-M.); (Y.H.C.); (C.G.-G.); (H.C.); (M.F.)
| |
Collapse
|
143
|
Wang X, Liu D, Shen L, Li F, Li Y, Yang L, Xu T, Tao H, Yao D, Wu L, Hirata K, Bohn LM, Makriyannis A, Liu X, Hua T, Liu ZJ, Wang J. A Genetically Encoded F-19 NMR Probe Reveals the Allosteric Modulation Mechanism of Cannabinoid Receptor 1. J Am Chem Soc 2021; 143:16320-16325. [PMID: 34596399 DOI: 10.1021/jacs.1c06847] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Due to the lack of genetically encoded probes for fluorine-19 nuclear magnetic resonance spectroscopy (19F NMR), its utility for probing eukaryotic membrane protein dynamics is limited. Here we report an efficient method for the genetic incorporation of an unnatural amino acid (UAA), 3'-trifluoromenthyl-phenylalanine (mtfF), into cannabinoid receptor 1 (CB1) in the Baculovirus Expression System. The probe can be inserted at any environmentally sensitive site, while causing minimal structural perturbation to the target protein. Using 19F NMR and X-ray crystallography methods, we discovered that the allosteric modulator Org27569 and agonists synergistically stabilize a previously unrecognized pre-active state. An allosteric modulation model is proposed to explain Org27569's distinct behavior. We demonstrate that our site-specific 19F NMR labeling method is a powerful tool in decoding the mechanism of GPCR allosteric modulation. This new method should be broadly applicable for uncovering conformational states for many important eukaryotic membrane proteins.
Collapse
Affiliation(s)
- Xiaoyan Wang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Dongsheng Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Ling Shen
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, Yunnan 650500, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai 200031, China
| | - Fahui Li
- Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Yongze Li
- Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Lingyun Yang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Tiandan Xu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Houchao Tao
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Deqiang Yao
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Lijie Wu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | | | - Laura M Bohn
- Departments of Molecular Medicine and Neuroscience, Scripps Research, Jupiter, Florida 33458, United States
| | - Alexandros Makriyannis
- Center for Drug Discovery, Department of Pharmaceutical Sciences, and Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Xiaohong Liu
- Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhi-Jie Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, Yunnan 650500, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jiangyun Wang
- Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| |
Collapse
|
144
|
Structures of signaling complexes of lipid receptors S1PR1 and S1PR5 reveal mechanisms of activation and drug recognition. Cell Res 2021; 31:1263-1274. [PMID: 34526663 PMCID: PMC8441948 DOI: 10.1038/s41422-021-00566-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/31/2021] [Indexed: 02/05/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is an important bioactive lipid molecule in cell membrane metabolism and binds to G protein-coupled S1P receptors (S1PRs) to regulate embryonic development, physiological homeostasis, and pathogenic processes in various organs. S1PRs are lipid-sensing receptors and are therapeutic targets for drug development, including potential treatment of COVID-19. Herein, we present five cryo-electron microscopy structures of S1PRs bound to diverse drug agonists and the heterotrimeric Gi protein. Our structural and functional assays demonstrate the different binding modes of chemically distinct agonists of S1PRs, reveal the mechanical switch that activates these receptors, and provide a framework for understanding ligand selectivity and G protein coupling.
Collapse
|
145
|
Magham SV, Thaggikuppe Krishnamurthy P, Shaji N, Mani L, Balasubramanian S. Cannabinoid receptor 2 selective agonists and Alzheimer's disease: An insight into the therapeutic potentials. J Neurosci Res 2021; 99:2888-2905. [PMID: 34486749 DOI: 10.1002/jnr.24933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/04/2021] [Accepted: 07/14/2021] [Indexed: 12/19/2022]
Abstract
Endocannabinoid system has been extensively studied in recent decades, particularly the cannabinoid receptors CB1 and CB2, due to their important role in neuroinflammation. Among these, CB2 has gained prominence due to its selective overexpression in glial cells during neuroinflammation. In contrast to CB1 agonists, CB2 agonists have no side effects such as ataxia, hypothermia, euphoria, psychological, or addiction liabilities. CB2 and its selective agonists' above-mentioned unique properties have become a research focus in neurodegenerative disorders such as Alzheimer's disease (AD). The review discusses the neuroprotective role of CB receptors, particularly CB2, in AD, as well as the significance and limitations of this research.
Collapse
Affiliation(s)
- Sai Varshini Magham
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | | | - Neenu Shaji
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | - Lalithkumar Mani
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | | |
Collapse
|
146
|
Leo LM, Abood ME. CB1 Cannabinoid Receptor Signaling and Biased Signaling. Molecules 2021; 26:molecules26175413. [PMID: 34500853 PMCID: PMC8433814 DOI: 10.3390/molecules26175413] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/30/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022] Open
Abstract
The CB1 cannabinoid receptor is a G-protein coupled receptor highly expressed throughout the central nervous system that is a promising target for the treatment of various disorders, including anxiety, pain, and neurodegeneration. Despite the wide therapeutic potential of CB1, the development of drug candidates is hindered by adverse effects, rapid tolerance development, and abuse potential. Ligands that produce biased signaling—the preferential activation of a signaling transducer in detriment of another—have been proposed as a strategy to dissociate therapeutic and adverse effects for a variety of G-protein coupled receptors. However, biased signaling at the CB1 receptor is poorly understood due to a lack of strongly biased agonists. Here, we review studies that have investigated the biased signaling profile of classical cannabinoid agonists and allosteric ligands, searching for a potential therapeutic advantage of CB1 biased signaling in different pathological states. Agonist and antagonist bound structures of CB1 and proposed mechanisms of action of biased allosteric modulators are used to discuss a putative molecular mechanism for CB1 receptor activation and biased signaling. Current studies suggest that allosteric binding sites on CB1 can be explored to yield biased ligands that favor or hinder conformational changes important for biased signaling.
Collapse
|
147
|
Zhang H, Chen LN, Yang D, Mao C, Shen Q, Feng W, Shen DD, Dai A, Xie S, Zhou Y, Qin J, Sun JP, Scharf DH, Hou T, Zhou T, Wang MW, Zhang Y. Structural insights into ligand recognition and activation of the melanocortin-4 receptor. Cell Res 2021; 31:1163-1175. [PMID: 34433901 DOI: 10.1038/s41422-021-00552-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Melanocortin-4 receptor (MC4R) plays a central role in the regulation of energy homeostasis. Its high sequence similarity to other MC receptor family members, low agonist selectivity and the lack of structural information concerning MC4R-specific activation have hampered the development of MC4R-seletive therapeutics to treat obesity. Here, we report four high-resolution structures of full-length MC4R in complex with the heterotrimeric Gs protein stimulated by the endogenous peptide ligand α-MSH, FDA-approved drugs afamelanotide (Scenesse™) and bremelanotide (Vyleesi™), and a selective small-molecule ligand THIQ, respectively. Together with pharmacological studies, our results reveal the conserved binding mode of peptidic agonists, the distinctive molecular details of small-molecule agonist recognition underlying receptor subtype selectivity, and a distinct activation mechanism for MC4R, thereby offering new insights into G protein coupling. Our work may facilitate the discovery of selective therapeutic agents targeting MC4R.
Collapse
Affiliation(s)
- Huibing Zhang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Li-Nan Chen
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Dehua Yang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chunyou Mao
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Qingya Shen
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wenbo Feng
- School of Pharmacy, Fudan University, Shanghai, China
| | - Dan-Dan Shen
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Antao Dai
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shanshan Xie
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of Gastroenterology, Zhejiang University, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, China; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Yan Zhou
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jiao Qin
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jin-Peng Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Daniel H Scharf
- Department of Microbiology and The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tingjun Hou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tianhua Zhou
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of Gastroenterology, Zhejiang University, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, China; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ming-Wei Wang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,School of Pharmacy, Fudan University, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China. .,Department of Pharmacology, Fudan University, Shanghai, China.
| | - Yan Zhang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. .,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China. .,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. .,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
148
|
Booth WT, Clodfelter JE, Leone-Kabler S, Hughes EK, Eldeeb K, Howlett AC, Lowther WT. Cannabinoid receptor interacting protein 1a interacts with myristoylated Gα i N terminus via a unique gapped β-barrel structure. J Biol Chem 2021; 297:101099. [PMID: 34418434 PMCID: PMC8446797 DOI: 10.1016/j.jbc.2021.101099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 11/15/2022] Open
Abstract
Cannabinoid receptor interacting protein 1a (CRIP1a) modulates CB1 cannabinoid receptor G-protein coupling in part by altering the selectivity for Gαi subtype activation, but the molecular basis for this function of CRIP1a is not known. We report herein the first structure of CRIP1a at a resolution of 1.55 Å. CRIP1a exhibits a 10-stranded and antiparallel β-barrel with an interior comprised of conserved hydrophobic residues and loops at the bottom and a short helical cap at the top to exclude solvent. The β-barrel has a gap between strands β8 and β10, which deviates from β-sandwich fatty acid–binding proteins that carry endocannabinoid compounds and the Rho-guanine nucleotide dissociation inhibitor predicted by computational threading algorithms. The structural homology search program DALI identified CRIP1a as homologous to a family of lipidated-protein carriers that includes phosphodiesterase 6 delta subunit and Unc119. Comparison with these proteins suggests that CRIP1a may carry two possible types of cargo: either (i) like phosphodiesterase 6 delta subunit, cargo with a farnesyl moiety that enters from the top of the β-barrel to occupy the hydrophobic interior or (ii) like Unc119, cargo with a palmitoyl or a myristoyl moiety that enters from the side where the missing β-strand creates an opening to the hydrophobic pocket. Fluorescence polarization analysis demonstrated CRIP1a binding of an N-terminally myristoylated 9-mer peptide mimicking the Gαi N terminus. However, CRIP1a could not bind the nonmyristolyated Gαi peptide or cargo of homologs. Thus, binding of CRIP1a to Gαi proteins represents a novel mechanism to regulate cell signaling initiated by the CB1 receptor.
Collapse
Affiliation(s)
- William T Booth
- Department of Biochemistry and Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jill E Clodfelter
- Department of Biochemistry and Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Sandra Leone-Kabler
- Department of Physiology and Pharmacology and Center for Research on Substance Use and Addiction, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Erin K Hughes
- Department of Biochemistry and Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA; Department of Physiology and Pharmacology and Center for Research on Substance Use and Addiction, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Khalil Eldeeb
- Department of Physiology and Pharmacology and Center for Research on Substance Use and Addiction, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Allyn C Howlett
- Department of Physiology and Pharmacology and Center for Research on Substance Use and Addiction, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA; Center for Molecular Signaling, Wake Forest University, Winston-Salem, North Carolina, USA.
| | - W Todd Lowther
- Department of Biochemistry and Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA; Center for Molecular Signaling, Wake Forest University, Winston-Salem, North Carolina, USA.
| |
Collapse
|
149
|
Cryo-EM structure of the human MT 1-G i signaling complex. Nat Struct Mol Biol 2021; 28:694-701. [PMID: 34354246 DOI: 10.1038/s41594-021-00634-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 06/30/2021] [Indexed: 11/08/2022]
Abstract
Melatonin receptors (MT1 and MT2) transduce inhibitory signaling by melatonin (N-acetyl-5-methoxytryptamine), which is associated with sleep induction and circadian rhythm modulation. Although recently reported crystal structures of ligand-bound MT1 and MT2 elucidated the basis of ligand entry and recognition, the ligand-induced MT1 rearrangement leading to Gi-coupling remains unclear. Here we report a cryo-EM structure of the human MT1-Gi signaling complex at 3.3 Å resolution, revealing melatonin-induced conformational changes propagated to the G-protein-coupling interface during activation. In contrast to other Gi-coupled receptors, MT1 exhibits a large outward movement of TM6, which is considered a specific feature of Gs-coupled receptors. Structural comparison of Gi and Gs complexes demonstrated conformational diversity of the C-terminal entry of the Gi protein, suggesting loose and variable interactions at the end of the α5 helix of Gi protein. These notions, together with our biochemical and computational analyses, highlight variable binding modes of Gαi and provide the basis for the selectivity of G-protein signaling.
Collapse
|
150
|
Yeliseev AA, Zoretich K, Hooper L, Teague W, Zoubak L, Hines KG, Gawrisch K. Site-selective labeling and electron paramagnetic resonance studies of human cannabinoid receptor CB 2. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2021; 1863:183621. [PMID: 33865808 PMCID: PMC8154700 DOI: 10.1016/j.bbamem.2021.183621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 11/19/2022]
Abstract
Integral membrane G protein-coupled receptors (GPCR) regulate multiple physiological processes by transmitting signals from extracellular milieu to intracellular proteins and are major targets of pharmaceutical drug development. Since GPCR are inherently flexible proteins, their conformational dynamics can be studied by spectroscopic techniques such as electron paramagnetic resonance (EPR) which requires selective chemical labeling of the protein. Here, we developed protocols for selective chemical labeling of the recombinant human cannabinoid receptor CB2 by judiciously replacing naturally occurring reactive cysteine residues and introducing a new single cysteine residue in selected positions. The majority of the 47 newly generated single cysteine constructs expressed well in E. coli cells, and more than half of them retained high functional activity. The reactivity of newly introduced cysteine residues was assessed by incorporating nitroxide spin label and EPR measurement. The conformational transition of the receptor between the inactive and activated form were studied by EPR of selectively labeled constructs in the presence of either a full agonist CP-55,940 or an inverse agonist SR-144,528. We observed evidence for higher mobility of labels in the center of internal loop 3 and a structural change between agonist vs. inverse agonist-bound CB2 in the extracellular tip of transmembrane helix 6. Our results demonstrate the utility of EPR for studies of conformational dynamics of CB2.
Collapse
Affiliation(s)
- Alexei A Yeliseev
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Kaeli Zoretich
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Levi Hooper
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Walter Teague
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lioudmila Zoubak
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kirk G Hines
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Klaus Gawrisch
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|