1
|
Barekatain M, Johansson LC, Lam JH, Chang H, Sadybekov AV, Han GW, Russo J, Bliesath J, Brice N, Carlton MBL, Saikatendu KS, Sun H, Murphy ST, Monenschein H, Schiffer HH, Popov P, Lutomski CA, Robinson CV, Liu ZJ, Hua T, Katritch V, Cherezov V. Structural insights into the high basal activity and inverse agonism of the orphan receptor GPR6 implicated in Parkinson's disease. Sci Signal 2024; 17:eado8741. [PMID: 39626010 PMCID: PMC11850111 DOI: 10.1126/scisignal.ado8741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 11/07/2024] [Indexed: 02/06/2025]
Abstract
GPR6 is an orphan G protein-coupled receptor with high constitutive activity found in D2-type dopamine receptor-expressing medium spiny neurons of the striatopallidal pathway, which is aberrantly hyperactivated in Parkinson's disease. Here, we solved crystal structures of GPR6 without the addition of a ligand (a pseudo-apo state) and in complex with two inverse agonists, including CVN424, which improved motor symptoms in patients with Parkinson's disease in clinical trials. In addition, we obtained a cryo-electron microscopy structure of the signaling complex between GPR6 and its cognate Gs heterotrimer. The pseudo-apo structure revealed a strong density in the orthosteric pocket of GPR6 corresponding to a lipid-like endogenous ligand. A combination of site-directed mutagenesis, native mass spectrometry, and computer modeling suggested potential mechanisms for high constitutive activity and inverse agonism in GPR6 and identified a series of lipids and ions bound to the receptor. The structures and results obtained in this study could guide the rational design of drugs that modulate GPR6 signaling.
Collapse
Affiliation(s)
- Mahta Barekatain
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Linda C. Johansson
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Jordy H. Lam
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Hao Chang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Anastasiia V. Sadybekov
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Gye Won Han
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Joseph Russo
- Takeda Development Center Americas, Inc, San Diego, CA 92121, USA
| | - Joshua Bliesath
- Takeda Development Center Americas, Inc, San Diego, CA 92121, USA
| | | | | | | | - Hukai Sun
- Takeda Development Center Americas, Inc, San Diego, CA 92121, USA
| | - Sean T. Murphy
- Takeda Development Center Americas, Inc, San Diego, CA 92121, USA
| | | | - Hans H. Schiffer
- Takeda Development Center Americas, Inc, San Diego, CA 92121, USA
| | - Petr Popov
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Corinne A. Lutomski
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford OX1 3QU, UK
| | - Carol V. Robinson
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford OX1 3QU, UK
| | - Zhi-Jie Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Vsevolod Katritch
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Vadim Cherezov
- Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
2
|
Lu Y, Hatzipantelis CJ, Langmead CJ, Stewart GD. Molecular insights into orphan G protein-coupled receptors relevant to schizophrenia. Br J Pharmacol 2024; 181:2095-2113. [PMID: 37605621 DOI: 10.1111/bph.16221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/25/2023] [Accepted: 07/23/2023] [Indexed: 08/23/2023] Open
Abstract
Schizophrenia remains a sizable socio-economic burden that continues to be treated with therapeutics based on 70-year old science. All currently approved therapeutics primarily target the dopamine D2 receptor to achieve their efficacy. Whilst dopaminergic dysregulation is a key feature in this disorder, the targeting of dopaminergic machinery has yielded limited efficacy and an appreciable side effect burden. Over the recent decades, numerous drugs that engage non-dopaminergic G protein-coupled receptors (GPCRs) have yielded a promise of efficacy without the deleterious side effect profile, yet none have successfully completed clinical studies and progressed to the market. More recently, there has been increased attention around non-dopaminergic GPCR-targeting drugs, which demonstrated efficacy in some schizophrenia symptom domains. This provides renewed hope that effective schizophrenia treatment may lie outside of the dopaminergic space. Despite the potential for muscarinic receptor- (and other well-characterised GPCR families) targeting drugs to treat schizophrenia, they are often plagued with complications such as lack of receptor subtype selectivity and peripheral on-target side effects. Orphan GPCR studies have opened a new avenue of exploration with many demonstrating schizophrenia-relevant mechanisms and a favourable expression profile, thus offering potential for novel drug development. This review discusses centrally expressed orphan GPCRs: GPR3, GPR6, GPR12, GPR52, GPR85, GPR88 and GPR139 and their relationship to schizophrenia. We review their expression, signalling mechanisms and cellular function, in conjunction with small molecule development and structural insights. We seek to provide a snapshot of the growing evidence and development potential of new classes of schizophrenia therapeutics. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Yao Lu
- Drug Discovery Biology and Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | | | - Christopher J Langmead
- Drug Discovery Biology and Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Australia
- Phrenix Therapeutics, Parkville, Australia
| | - Gregory D Stewart
- Drug Discovery Biology and Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Australia
- Phrenix Therapeutics, Parkville, Australia
| |
Collapse
|
3
|
Öz-Arslan D, Yavuz M, Kan B. Exploring orphan GPCRs in neurodegenerative diseases. Front Pharmacol 2024; 15:1394516. [PMID: 38895631 PMCID: PMC11183337 DOI: 10.3389/fphar.2024.1394516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
Neurodegenerative disorders represent a significant and growing health burden worldwide. Unfortunately, limited therapeutic options are currently available despite ongoing efforts. Over the past decades, research efforts have increasingly focused on understanding the molecular mechanisms underlying these devastating conditions. Orphan receptors, a class of receptors with no known endogenous ligands, emerge as promising druggable targets for diverse diseases. This review aims to direct attention to a subgroup of orphan GPCRs, in particular class A orphans that have roles in neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and Multiple sclerosis. We highlight the diverse roles orphan receptors play in regulating critical cellular processes such as synaptic transmission, neuronal survival and neuro-inflammation. Moreover, we discuss the therapeutic potential of targeting orphan receptors for the treatment of neurodegenerative disorders, emphasizing recent advances in drug discovery and preclinical studies. Finally, we outline future directions and challenges in orphan receptor research.
Collapse
Affiliation(s)
- Devrim Öz-Arslan
- Department of Biophysics, Acibadem MAA University, School of Medicine, Istanbul, Türkiye
- Department of Neurosciences, Acibadem MAA University, Institute of Health Sciences, İstanbul, Türkiye
| | - Melis Yavuz
- Department of Neurosciences, Acibadem MAA University, Institute of Health Sciences, İstanbul, Türkiye
- Department of Pharmacology, Acibadem MAA University, School of Pharmacy, Istanbul, Türkiye
| | - Beki Kan
- Department of Biophysics, Acibadem MAA University, School of Medicine, Istanbul, Türkiye
- Department of Neurosciences, Acibadem MAA University, Institute of Health Sciences, İstanbul, Türkiye
| |
Collapse
|
4
|
Anversa RG, Maddern XJ, Lawrence AJ, Walker LC. Orphan peptide and G protein-coupled receptor signalling in alcohol use disorder. Br J Pharmacol 2024; 181:595-609. [PMID: 38073127 PMCID: PMC10953447 DOI: 10.1111/bph.16301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 01/10/2024] Open
Abstract
Neuropeptides and G protein-coupled receptors (GPCRs) have long been, and continue to be, one of the most popular target classes for drug discovery in CNS disorders, including alcohol use disorder (AUD). Yet, orphaned neuropeptide systems and receptors (oGPCR), which have no known cognate receptor or ligand, remain understudied in drug discovery and development. Orphan neuropeptides and oGPCRs are abundantly expressed within the brain and represent an unprecedented opportunity to address brain function and may hold potential as novel treatments for disease. Here, we describe the current literature regarding orphaned neuropeptides and oGPCRs implicated in AUD. Specifically, in this review, we focus on the orphaned neuropeptide cocaine- and amphetamine-regulated transcript (CART), and several oGPCRs that have been directly implicated in AUD (GPR6, GPR26, GPR88, GPR139, GPR158) and discuss their potential and pitfalls as novel treatments, and progress in identifying their cognate receptors or ligands.
Collapse
Affiliation(s)
- Roberta Goncalves Anversa
- Florey Institute of Neuroscience and Mental HealthMelbourneVICAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVICAustralia
| | - Xavier J. Maddern
- Florey Institute of Neuroscience and Mental HealthMelbourneVICAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVICAustralia
| | - Andrew J. Lawrence
- Florey Institute of Neuroscience and Mental HealthMelbourneVICAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVICAustralia
| | - Leigh C. Walker
- Florey Institute of Neuroscience and Mental HealthMelbourneVICAustralia
- Florey Department of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVICAustralia
| |
Collapse
|
5
|
Gay EA, Harris DL, Wilson JW, Blough BE. The development of diphenyleneiodonium analogs as GPR3 agonists. Bioorg Med Chem Lett 2023; 94:129427. [PMID: 37541631 PMCID: PMC10631289 DOI: 10.1016/j.bmcl.2023.129427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/20/2023] [Accepted: 07/29/2023] [Indexed: 08/06/2023]
Abstract
G protein-coupled receptor 3 (GPR3) is an orphan receptor potentially involved in many important physiological processes such as drug abuse, neuropathic pain, and anxiety and depression related disorders. Pharmacological studies of GPR3 have been limited due to the restricted number of known agonists and inverse agonists for this constitutively active receptor. In this medicinal chemistry study, we report the discovery of GPR3 agonists based off the diphenyleneiodonium (DPI) scaffold. The most potent full agonist was the 3-trifluoromethoxy analog (32) with an EC50 of 260 nM and 90% efficacy compared to DPI. Investigation of a homology model of GPR3 from multiple sequence alignment resulted in the finding of a binding site rich in potential π-π and π-cation interactions stabilizing DPI-scaffold agonists. MMGBSA free energy analysis showed a good correlation with trends in observed EC50s. DPI analogs retained the same high receptor selectivity for GPR3 over GPR6 and GPR12 as observed with DPI. Collectively, the DPI analog series shows that order of magnitude improvements in potency with the scaffold were attainable; however, attempts to replace the iodonium ion to make the scaffold more druggable failed.
Collapse
Affiliation(s)
- Elaine A Gay
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, USA.
| | - Danni L Harris
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, USA
| | - Joseph W Wilson
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, USA
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, USA
| |
Collapse
|
6
|
Aguayo-Cerón KA, Sánchez-Muñoz F, Gutierrez-Rojas RA, Acevedo-Villavicencio LN, Flores-Zarate AV, Huang F, Giacoman-Martinez A, Villafaña S, Romero-Nava R. Glycine: The Smallest Anti-Inflammatory Micronutrient. Int J Mol Sci 2023; 24:11236. [PMID: 37510995 PMCID: PMC10379184 DOI: 10.3390/ijms241411236] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Glycine is a non-essential amino acid with many functions and effects. Glycine can bind to specific receptors and transporters that are expressed in many types of cells throughout an organism to exert its effects. There have been many studies focused on the anti-inflammatory effects of glycine, including its abilities to decrease pro-inflammatory cytokines and the concentration of free fatty acids, to improve the insulin response, and to mediate other changes. However, the mechanism through which glycine acts is not clear. In this review, we emphasize that glycine exerts its anti-inflammatory effects throughout the modulation of the expression of nuclear factor kappa B (NF-κB) in many cells. Although glycine is a non-essential amino acid, we highlight how dietary glycine supplementation is important in avoiding the development of chronic inflammation.
Collapse
Affiliation(s)
- Karla Aidee Aguayo-Cerón
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología "Ignacio Chávez", Ciudad de Mexico 14080, Mexico
| | | | | | - Aurora Vanessa Flores-Zarate
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| | - Fengyang Huang
- Laboratorio de Investigación en Obesidad y Asma, Hospital Infantil de México Federico Gómez, Ciudad de Mexico 06720, Mexico
| | - Abraham Giacoman-Martinez
- Laboratorio de Framacología, Departamaneto de Ciencias de la Salud, DCBS, Universidad Autónoma Mteropolitana-Iztapalapa (UAM-I), Ciudad de Mexico 09340, Mexico
| | - Santiago Villafaña
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| | - Rodrigo Romero-Nava
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Sección de Estudios de Posgrado e Investigación, Ciudad de Mexico 11340, Mexico
| |
Collapse
|
7
|
Targeting G Protein-Coupled Receptors in the Treatment of Parkinson's Disease. J Mol Biol 2022:167927. [PMID: 36563742 DOI: 10.1016/j.jmb.2022.167927] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease characterized in part by the deterioration of dopaminergic neurons which leads to motor impairment. Although there is no cure for PD, the motor symptoms can be treated using dopamine replacement therapies including the dopamine precursor L-DOPA, which has been in use since the 1960s. However, neurodegeneration in PD is not limited to dopaminergic neurons, and many patients experience non-motor symptoms including cognitive impairment or neuropsychiatric disturbances, for which there are limited treatment options. Moreover, there are currently no treatments able to alter the progression of neurodegeneration. There are many therapeutic strategies being investigated for PD, including alternatives to L-DOPA for the treatment of motor impairment, symptomatic treatments for non-motor symptoms, and neuroprotective or disease-modifying agents. G protein-coupled receptors (GPCRs), which include the dopamine receptors, are highly druggable cell surface proteins which can regulate numerous intracellular signaling pathways and thereby modulate the function of neuronal circuits affected by PD. This review will describe the treatment strategies being investigated for PD that target GPCRs and their downstream signaling mechanisms. First, we discuss new developments in dopaminergic agents for alleviating PD motor impairment, the role of dopamine receptors in L-DOPA induced dyskinesia, as well as agents targeting non-dopamine GPCRs which could augment or replace traditional dopaminergic treatments. We then discuss GPCRs as prospective treatments for neuropsychiatric and cognitive symptoms in PD. Finally, we discuss the evidence pertaining to ghrelin receptors, β-adrenergic receptors, angiotensin receptors and glucagon-like peptide 1 receptors, which have been proposed as disease modifying targets with potential neuroprotective effects in PD.
Collapse
|
8
|
Bharathi, Roy KK. Structural basis for the binding of a selective inverse agonist AF64394 with the human G-protein coupled receptor 3 (GPR3). J Biomol Struct Dyn 2022; 40:10181-10190. [PMID: 34157950 DOI: 10.1080/07391102.2021.1940282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The orphan class A G-protein coupled receptor 3 (GPR3) is highly expressed in brain and linked with various neuronal functions, and therefore, expected to play a vital role in the progression of Alzheimer's disease. In view of the lack of its experimental structure, we describe herein the three-dimensional structure and conformational dynamics of GPR3 complexed with the inverse agonist AF64394. The GPR3 model was predicted using the Iterative Threading ASSEmbly Refinement (I-TASSER) method. The Induced Fit Docking predicted two unique poses, Pose 1 and Pose 2, for AF64394, and then, molecular dynamics (MD) simulations followed by binding free-energy calculation revealed the Pose 1 as a very stable pose with the least fluctuation during the MD simulation while the Pose 2 underwent a significant fluctuation. The [1,2,4]triazolo[1,5-a]pyrimidine core was engaged in multiple hydrogen bonds (H-bonds), such as a water-mediated H-bond between the triazole nitrogen and T31, two direct H-bonds between the protonated triazole-ring nitrogen and V186 and T279, a direct H-bond between the secondary amine and V187. The phenyl substituent of AF64394 exhibited aromatic π-π stacking interactions with F97, F101, W43 and Y280. AF64394 showed a direct interaction with E28 and polar interactions with H96, T31 and T279. Throughout the MD simulation, the toggle switch residues, F120 and W260, remained in close contact, indicating that the GPR3 conformation represented an inactive state. The 4-(3-chloro-5-isopropoxyphenethyl) group resided near to the toggle switch residues. The insights gained here are expected to be useful in the structure-based design of new ligands targeting GPR3 modulation. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Bharathi
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Kolkata, India
| | - Kuldeep K Roy
- Department of Pharmaceutical Technology, School of Medical Sciences, Adamas University, Kolkata, India
| |
Collapse
|
9
|
Biringer RG. Endocannabinoid signaling pathways: beyond CB1R and CB2R. J Cell Commun Signal 2021; 15:335-360. [PMID: 33978927 PMCID: PMC8222499 DOI: 10.1007/s12079-021-00622-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022] Open
Abstract
The search for cannabinoid receptors other than CB1R and CB2R has been ongoing for over a decade. A number of orphan receptors have been proposed as potential cannabinoid receptors primarily based on phylogenic arguments and reactivity towards known endocannabinoids and phytocannabinoids. Seven putative cannabinoid receptors are described and discussed, and evidence for and against their inclusion in this category are presented.
Collapse
Affiliation(s)
- Roger Gregory Biringer
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
10
|
Brice NL, Schiffer HH, Monenschein H, Mulligan VJ, Page K, Powell J, Xu X, Cheung T, Burley JR, Sun H, Dickson L, Murphy ST, Kaushal N, Sheardown S, Lawrence J, Chen Y, Bartkowski D, Kanta A, Russo J, Hosea N, Dawson LA, Hitchcock SH, Carlton MB. Development of CVN424: A Selective and Novel GPR6 Inverse Agonist Effective in Models of Parkinson Disease. J Pharmacol Exp Ther 2021; 377:407-416. [PMID: 33795395 DOI: 10.1124/jpet.120.000438] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/29/2021] [Indexed: 07/25/2024] Open
Abstract
GPR6 is an orphan G-protein-coupled receptor that has enriched expression in the striatopallidal, indirect pathway and medium spiny neurons of the striatum. This pathway is greatly impacted by the loss of the nigro-striatal dopaminergic neurons in Parkinson disease, and modulating this neurocircuitry can be therapeutically beneficial. In this study, we describe the in vitro and in vivo pharmacological characterization of (R)-1-(2-(4-(2,4-difluorophenoxy)piperidin-1-yl)-3-((tetrahydrofuran-3-yl)amino)-7,8-dihydropyrido[3,4-b]pyrazin-6(5H)-yl)ethan-1-one (CVN424), a highly potent and selective small-molecule inverse agonist for GPR6 that is currently undergoing clinical evaluation. CVN424 is brain-penetrant and shows dose-dependent receptor occupancy that attained brain 50% of receptor occupancy at plasma concentrations of 6.0 and 7.4 ng/ml in mice and rats, respectively. Oral administration of CVN424 dose-dependently increases locomotor activity and reverses haloperidol-induced catalepsy. Furthermore, CVN424 restored mobility in bilateral 6-hydroxydopamine lesion model of Parkinson disease. The presence and localization of GPR6 in medium spiny neurons of striatum postmortem samples from both nondemented control and patients with Parkinson disease were confirmed at the level of both RNA (using Nuclear Enriched Transcript Sort sequencing) and protein. This body of work demonstrates that CVN424 is a potent, orally active, and brain-penetrant GPR6 inverse agonist that is effective in preclinical models and is a potential therapeutic for improving motor function in patients with Parkinson disease. SIGNIFICANCE STATEMENT: CVN424 represents a nondopaminergic novel drug for potential use in patients with Parkinson disease.
Collapse
Affiliation(s)
- Nicola L Brice
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Hans H Schiffer
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Holger Monenschein
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Victoria J Mulligan
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Keith Page
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Justin Powell
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Xiao Xu
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Toni Cheung
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - J Russell Burley
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Huikai Sun
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Louise Dickson
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Sean T Murphy
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Nidhi Kaushal
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Steve Sheardown
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Jason Lawrence
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Yun Chen
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Darian Bartkowski
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Anne Kanta
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Joseph Russo
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Natalie Hosea
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Lee A Dawson
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Stephen H Hitchcock
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| | - Mark B Carlton
- Cerevance Ltd, Cambridge, United Kingdom (N.L.B., V.J.M., K.P., J.P., X.X., T.C., J.R.B., L.D., S.S., J.L., L.A.D., M.B.C.); and Takeda California, San Diego, California (H.H.S., H.M., H.S., S.T.M., N.K., Y.C., D.B., A.K., J.R., N.H., S.H.H.)
| |
Collapse
|
11
|
Sun H, Monenschein H, Schiffer HH, Reichard HA, Kikuchi S, Hopkins M, Macklin TK, Hitchcock S, Adams M, Green J, Brown J, Murphy ST, Kaushal N, Collia DR, Moore S, Ray WJ, English NM, Carlton MBL, Brice NL. First-Time Disclosure of CVN424, a Potent and Selective GPR6 Inverse Agonist for the Treatment of Parkinson's Disease: Discovery, Pharmacological Validation, and Identification of a Clinical Candidate. J Med Chem 2021; 64:9875-9890. [PMID: 33861086 DOI: 10.1021/acs.jmedchem.0c02081] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Parkinson's disease (PD) is a chronic and progressive movement disorder with the urgent unmet need for efficient symptomatic therapies with fewer side effects. GPR6 is an orphan G-protein coupled receptor (GPCR) with highly restricted expression in dopamine receptor D2-type medium spiny neurons (MSNs) of the indirect pathway, a striatal brain circuit which shows aberrant hyperactivity in PD patients. Potent and selective GPR6 inverse agonists (IAG) were developed starting from a low-potency screening hit (EC50 = 43 μM). Herein, we describe the multiple parameter optimization that led to the discovery of multiple nanomolar potent and selective GPR6 IAG, including our clinical compound CVN424. GPR6 IAG reversed haloperidol-induced catalepsy in rats and restored mobility in the bilateral 6-OHDA-lesioned rat PD model demonstrating that inhibition of GPR6 activity in vivo normalizes activity in basal ganglia circuitry and motor behavior. CVN424 is currently in clinical development to treat motor symptoms in Parkinson's disease.
Collapse
Affiliation(s)
- Huikai Sun
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Holger Monenschein
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Hans H Schiffer
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Holly A Reichard
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Shota Kikuchi
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Maria Hopkins
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Todd K Macklin
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Stephen Hitchcock
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Mark Adams
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Jason Green
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Jason Brown
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Sean T Murphy
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Nidhi Kaushal
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Deanna R Collia
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Steve Moore
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - William J Ray
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Nicole Marion English
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | | | - Nicola L Brice
- Cerevance Ltd, 418 Cambridge Science Park, Cambridge, U.K
| |
Collapse
|
12
|
Characterization of Four Orphan Receptors (GPR3, GPR6, GPR12 and GPR12L) in Chickens and Ducks and Regulation of GPR12 Expression in Ovarian Granulosa Cells by Progesterone. Genes (Basel) 2021; 12:genes12040489. [PMID: 33801713 PMCID: PMC8065388 DOI: 10.3390/genes12040489] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 11/29/2022] Open
Abstract
The three structurally related orphan G protein-coupled receptors, GRP3, GPR6, and GPR12, are reported to be constitutively active and likely involved in the regulation of many physiological/pathological processes, such as neuronal outgrowth and oocyte meiotic arrest in mammals. However, the information regarding these orphan receptors in nonmammalian vertebrates is extremely limited. Here, we reported the structure, constitutive activity, and tissue expression of these receptors in two representative avian models: chickens and ducks. The cloned duck GPR3 and duck/chicken GPR6 and GPR12 are intron-less and encode receptors that show high amino acid (a.a.) sequence identities (66–88%) with their respective mammalian orthologs. Interestingly, a novel GPR12-like receptor (named GPR12L) sharing 66% a.a. identity to that in vertebrates was reported in the present study. Using dual-luciferase reporter assay and Western blot, we demonstrated that GPR3, GPR6, GPR12, and GPR12L are constitutively active and capable of stimulating the cAMP/PKA signaling pathway without ligand stimulation in birds (and zebrafish), indicating their conserved signaling property across vertebrates. RNA-seq data/qRT-PCR assays revealed that GPR6 and GPR12L expression is mainly restricted to the chicken brain, while GPR12 is highly expressed in chicken ovarian granulosa cells (GCs) and oocytes of 6 mm growing follicles and its expression in cultured GCs is upregulated by progesterone. Taken together, our data reveal the structure, function, and expression of GPR3, GPR6, GPR12, and GPR12L in birds, thus providing the first piece of evidence that GPR12 expression is upregulated by gonadal steroid (i.e., progesterone) in vertebrates.
Collapse
|
13
|
Mizuno H, Kihara Y. Druggable Lipid GPCRs: Past, Present, and Prospects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1274:223-258. [PMID: 32894513 DOI: 10.1007/978-3-030-50621-6_10] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptors (GPCRs) have seven transmembrane spanning domains and comprise the largest superfamily with ~800 receptors in humans. GPCRs are attractive targets for drug discovery because they transduce intracellular signaling in response to endogenous ligands via heterotrimeric G proteins or arrestins, resulting in a wide variety of physiological and pathophysiological responses. The endogenous ligands for GPCRs are highly chemically diverse and include ions, biogenic amines, nucleotides, peptides, and lipids. In this review, we follow the KonMari method to better understand druggable lipid GPCRs. First, we have a comprehensive tidying up of lipid GPCRs including receptors for prostanoids, leukotrienes, specialized pro-resolving mediators (SPMs), lysophospholipids, sphingosine 1-phosphate (S1P), cannabinoids, platelet-activating factor (PAF), free fatty acids (FFAs), and sterols. This tidying up consolidates 46 lipid GPCRs and declutters several perplexing lipid GPCRs. Then, we further tidy up the lipid GPCR-directed drugs from the literature and databases, which identified 24 clinical drugs targeting 16 unique lipid GPCRs available in the market and 44 drugs under evaluation in more than 100 clinical trials as of 2019. Finally, we introduce drug designs for GPCRs that spark joy, such as positive or negative allosteric modulators (PAM or NAM), biased agonism, functional antagonism like fingolimod, and monoclonal antibodies (MAbs). These strategic drug designs may increase the efficacy and specificity of drugs and reduce side effects. Technological advances will help to discover more endogenous lipid ligands from the vast number of remaining orphan GPCRs and will also lead to the development novel lipid GPCR drugs to treat various diseases.
Collapse
Affiliation(s)
| | - Yasuyuki Kihara
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
14
|
Isawi IH, Morales P, Sotudeh N, Hurst DP, Lynch DL, Reggio PH. GPR6 Structural Insights: Homology Model Construction and Docking Studies. Molecules 2020; 25:molecules25030725. [PMID: 32046081 PMCID: PMC7037797 DOI: 10.3390/molecules25030725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/01/2020] [Accepted: 02/04/2020] [Indexed: 01/14/2023] Open
Abstract
GPR6 is an orphan G protein-coupled receptor that has been associated with the cannabinoid family because of its recognition of a sub-set of cannabinoid ligands. The high abundance of GPR6 in the central nervous system, along with high constitutive activity and a link to several neurodegenerative diseases make GPR6 a promising biological target. In fact, diverse research groups have demonstrated that GPR6 represents a possible target for the treatment of neurodegenerative disorders such as Parkinson's disease, Alzheimer's disease, and Huntington's disease. Several patents have claimed the use of a wide range of pyrazine derivatives as GPR6 inverse agonists for the treatment of Parkinson's disease symptoms and other dyskinesia syndromes. However, the full pharmacological importance of GPR6 has not yet been fully explored due to the lack of high potency, readily available ligands targeting GPR6. The long-term goal of the present study is to develop such ligands. In this paper, we describe our initial steps towards this goal. A human GPR6 homology model was constructed using a suite of computational techniques. This model permitted the identification of unique GPR6 structural features and the exploration of the GPR6 binding crevice. A subset of patented pyrazine analogs were docked in the resultant GPR6 inactive state model to validate the model, rationalize the structure-activity relationships from the reported patents and identify the key residues in the binding crevice for ligand recognition. We will take this structural knowledge into the next phase of GPR6 project, in which scaffold hopping will be used to design new GPR6 ligands.
Collapse
Affiliation(s)
- Israa H. Isawi
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (I.H.I.); (D.P.H.); (D.L.L.)
| | - Paula Morales
- Instituto de Química Medica (IQM-CSIC), C/Juan de la Cierva 3, 28006 Madrid, Spain;
| | - Noori Sotudeh
- Department of Physiology and Biophysics, The State University of New York at Buffalo, Buffalo, NY 14260, USA;
| | - Dow P. Hurst
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (I.H.I.); (D.P.H.); (D.L.L.)
| | - Diane L. Lynch
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (I.H.I.); (D.P.H.); (D.L.L.)
| | - Patricia H. Reggio
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (I.H.I.); (D.P.H.); (D.L.L.)
- Correspondence:
| |
Collapse
|
15
|
Shrader SH, Song ZH. Discovery of endogenous inverse agonists for G protein-coupled receptor 6. Biochem Biophys Res Commun 2019; 522:1041-1045. [PMID: 31818461 DOI: 10.1016/j.bbrc.2019.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/01/2019] [Indexed: 01/01/2023]
Abstract
The orphan G protein-coupled receptor 6 (GPR6) is highly expressed in the striatum and has been linked to multiple striatal pathologies. The identification of endogenous ligands and their mechanisms of action at GPR6 will help to elucidate the physiological and pathological roles of the receptor. In the current study, we tested the concentration-dependent effects of a variety of endocannabinoid-like N-acylamides on GPR6 signaling. Here, we demonstrate for the first time that N-arachidonoyl dopamine, N-docosahexaenoyl dopamine, N-oleoyl dopamine and N-palmitoyl dopamine exert inverse agonism at GPR6. This effect was concentration-dependent, with potencies in the micromolar range, and functionally selective for β-arrestin2 recruitment. Structure-activity relationship studies demonstrate that both the N-acyl side chain and the dopamine head group are important for these ligands to act on GPR6. Our discovery of these N-acyl dopamines as endogenous inverse agonists for GPR6 moves us one step further in understanding the roles GPR6 play in neurodegenerative and neuropsychiatric disorders related to striatal dysfunction.
Collapse
Affiliation(s)
- Sarah H Shrader
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, United States
| | - Zhao-Hui Song
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, United States.
| |
Collapse
|
16
|
Laun AS, Shrader SH, Brown KJ, Song ZH. GPR3, GPR6, and GPR12 as novel molecular targets: their biological functions and interaction with cannabidiol. Acta Pharmacol Sin 2019; 40:300-308. [PMID: 29941868 PMCID: PMC6460361 DOI: 10.1038/s41401-018-0031-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/17/2018] [Indexed: 01/08/2023]
Abstract
The G protein-coupled receptors 3, 6, and 12 (GPR3, GPR6, and GPR12) comprise a family of closely related orphan receptors with no confirmed endogenous ligands. These receptors are constitutively active and capable of signaling through G protein-mediated and non-G protein-mediated mechanisms. These orphan receptors have previously been reported to play important roles in many normal physiological functions and to be involved in a variety of pathological conditions. Although they are orphans, GPR3, GPR6, and GPR12 are phylogenetically most closely related to the cannabinoid receptors. Using β-arrestin2 recruitment and cAMP accumulation assays, we recently found that the nonpsychoactive phytocannabinoid cannabidiol (CBD) is an inverse agonist for GPR3, GPR6, and GPR12. This discovery highlights these orphan receptors as potential new molecular targets for CBD, provides novel mechanisms of action, and suggests new therapeutic uses of CBD for illnesses such as Alzheimer's disease, Parkinson's disease, cancer, and infertility. Furthermore, identification of CBD as a new inverse agonist for GPR3, GPR6, and GPR12 provides the initial chemical scaffolds upon which potent and efficacious agents acting on these receptors can be developed, with the goal of developing chemical tools for studying these orphan receptors and ultimately new therapeutic agents.
Collapse
Affiliation(s)
- Alyssa S Laun
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Sarah H Shrader
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Kevin J Brown
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Zhao-Hui Song
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.
| |
Collapse
|
17
|
Laun AS, Shrader SH, Song ZH. Novel inverse agonists for the orphan G protein-coupled receptor 6. Heliyon 2018; 4:e00933. [PMID: 30480157 PMCID: PMC6240797 DOI: 10.1016/j.heliyon.2018.e00933] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/06/2018] [Accepted: 11/08/2018] [Indexed: 12/11/2022] Open
Abstract
The orphan G protein-coupled receptor 6 (GPR6) displays unique promise as a therapeutic target for the treatment of neuropsychiatric disorders due to its high expression in the striatopallidal neurons of the basal ganglia. GPR6, along with closely related orphan receptors GPR3 and GPR12, are phylogenetically related to CB1 and CB2 cannabinoid receptors. In the current study, we performed concentration-response studies on the effects of three different classes of cannabinoids: endogenous, phyto-, and synthetic, on both GPR6-mediated cAMP accumulation and β-arrestin2 recruitment. In addition, structure-activity relationship studies were conducted on cannabidiol (CBD), a recently discovered inverse agonist for GPR6. We have identified four additional cannabinoids, cannabidavarin (CBDV), WIN55212-2, SR141716A and SR144528, that exert inverse agonism on GPR6. Furthermore, we have discovered that these cannabinoids exhibit functional selectivity toward the β-arrestin2 recruitment pathway. These novel, functionally selective inverse agonists for GPR6 can be used as research tools and potentially developed into therapeutic agents.
Collapse
Affiliation(s)
- Alyssa S Laun
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, United States
| | - Sarah H Shrader
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, United States
| | - Zhao-Hui Song
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, United States
| |
Collapse
|
18
|
Morales P, Isawi I, Reggio PH. Towards a better understanding of the cannabinoid-related orphan receptors GPR3, GPR6, and GPR12. Drug Metab Rev 2018; 50:74-93. [PMID: 29390908 DOI: 10.1080/03602532.2018.1428616] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
GPR3, GPR6, and GPR12 are three orphan receptors that belong to the Class A family of G-protein-coupled receptors (GPCRs). These GPCRs share over 60% of sequence similarity among them. Because of their close phylogenetic relationship, GPR3, GPR6, and GPR12 share a high percentage of homology with other lipid receptors such as the lysophospholipid and the cannabinoid receptors. On the basis of sequence similarities at key structural motifs, these orphan receptors have been related to the cannabinoid family. However, further experimental data are required to confirm this association. GPR3, GPR6, and GPR12 are predominantly expressed in mammalian brain. Their high constitutive activation of adenylyl cyclase triggers increases in cAMP levels similar in amplitude to fully activated GPCRs. This feature defines their physiological role under certain pathological conditions. In this review, we aim to summarize the knowledge attained so far on the understanding of these receptors. Expression patterns, pharmacology, physiopathological relevance, and molecules targeting GPR3, GPR6, and GPR12 will be analyzed herein. Interestingly, certain cannabinoid ligands have been reported to modulate these orphan receptors. The current debate about sphingolipids as putative endogenous ligands will also be addressed. A special focus will be on their potential role in the brain, particularly under neurological conditions such as Parkinson or Alzheimer's disease. Reported physiological roles outside the central nervous system will also be covered. This critical overview may contribute to a further comprehension of the physiopathological role of these orphan GPCRs, hopefully attracting more research towards a future therapeutic exploitation of these promising targets.
Collapse
Affiliation(s)
- Paula Morales
- a Department of Chemistry and Biochemistry , University of North Carolina at Greensboro , Greensboro , NC , USA
| | - Israa Isawi
- a Department of Chemistry and Biochemistry , University of North Carolina at Greensboro , Greensboro , NC , USA
| | - Patricia H Reggio
- a Department of Chemistry and Biochemistry , University of North Carolina at Greensboro , Greensboro , NC , USA
| |
Collapse
|
19
|
Alavi MS, Shamsizadeh A, Azhdari-Zarmehri H, Roohbakhsh A. Orphan G protein-coupled receptors: The role in CNS disorders. Biomed Pharmacother 2017; 98:222-232. [PMID: 29268243 DOI: 10.1016/j.biopha.2017.12.056] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022] Open
Abstract
There are various types of receptors in the central nervous system (CNS). G protein-coupled receptors (GPCRs) have the highest expression with a wide range of physiological functions. A newer sub group of these receptors namely orphan GPCRs have been discovered. GPR3, GPR6, GPR17, GPR26, GPR37, GPR39, GPR40, GPR50, GPR52, GPR54, GPR55, GPR85, GPR88, GPR103, and GPR139 are the selected orphan GPCRs for this article. Their roles in the central nervous system have not been understood well so far. However, recent studies show that they may have very important functions in the CNS. Hence, in the present study, we reviewed most recent findings regarding the physiological roles of the selected orphan GPCRs in the CNS. After a brief presentation of each receptor, considering the results from genetic and pharmacological manipulation of the receptors, their roles in the pathophysiology of different diseases and disorders including anxiety, depression, schizophrenia, epilepsy, Alzheimer's disease, Parkinson's disease, and substance abuse will be discussed. At present, our knowledge regarding the role of GPCRs in the brain is very limited. However, previous limited studies show that orphan GPCRs have an important place in psychopharmacology and these receptors are potential new targets for the treatment of major CNS diseases.
Collapse
Affiliation(s)
- Mohaddeseh Sadat Alavi
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Shamsizadeh
- Physiology-Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hassan Azhdari-Zarmehri
- Department of Basic Medical Sciences and Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Morales P, Reggio PH. An Update on Non-CB 1, Non-CB 2 Cannabinoid Related G-Protein-Coupled Receptors. Cannabis Cannabinoid Res 2017; 2:265-273. [PMID: 29098189 PMCID: PMC5665501 DOI: 10.1089/can.2017.0036] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The endocannabinoid system (ECS) has been shown to be of great importance in the regulation of numerous physiological and pathological processes. To date, two Class A G-protein-coupled receptors (GPCRs) have been discovered and validated as the main therapeutic targets of this system: the cannabinoid receptor type 1 (CB1), which is the most abundant neuromodulatory receptor in the brain, and the cannabinoid receptor type 2 (CB2), predominantly found in the immune system among other organs and tissues. Endogenous cannabinoid receptor ligands (endocannabinoids) and the enzymes involved in their synthesis, cell uptake, and degradation have also been identified as part of the ECS. However, its complex pharmacology suggests that other GPCRs may also play physiologically relevant roles in this therapeutically promising system. In the last years, GPCRs such as GPR18 and GPR55 have emerged as possible missing members of the cannabinoid family. This categorization still stimulates strong debate due to the lack of pharmacological tools to validate it. Because of their close phylogenetic relationship, the Class A orphan GPCRs, GPR3, GPR6, and GPR12, have also been associated with the cannabinoids. Moreover, certain endo-, phyto-, and synthetic cannabinoid ligands have displayed activity at other well-established GPCRs, including the opioid, adenosine, serotonin, and dopamine receptor families. In addition, the cannabinoid receptors have also been shown to form dimers with other GPCRs triggering cross-talk signaling under specific conditions. In this mini review, we aim to provide insight into the non-CB1, non-CB2 cannabinoid-related GPCRs that have been reported thus far. We consider the physiological relevance of these molecular targets in modulating the ECS.
Collapse
Affiliation(s)
- Paula Morales
- Chemistry and Biochemistry Department, UNC Greensboro, Greensboro, North Carolina
| | - Patricia H. Reggio
- Chemistry and Biochemistry Department, UNC Greensboro, Greensboro, North Carolina
| |
Collapse
|
21
|
Laun AS, Song ZH. GPR3 and GPR6, novel molecular targets for cannabidiol. Biochem Biophys Res Commun 2017; 490:17-21. [PMID: 28571738 DOI: 10.1016/j.bbrc.2017.05.165] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/28/2017] [Indexed: 10/19/2022]
Abstract
GPR3 and GPR6 are members of a family of constitutively active, Gs protein-coupled receptors. Previously, it has been reported that GPR3 is involved in Alzheimer's disease whereas GPR6 plays potential roles in Parkinson's disease. GPR3 and GPR6 are considered orphan receptors because there are no confirmed endogenous agonists for them. However, GPR3 and GPR6 are phylogenetically related to the cannabinoid receptors. In this study, the activities of endocannabinoids and phytocannabinoids were tested on GPR3 and GPR6 using a β-arrestin2 recruitment assay. Among the variety of cannabinoids tested, cannabidiol (CBD), the major non-psychoactive component of marijuana, significantly reduced β-arrestin2 recruitment to both GPR3 and GPR6. In addition, the inhibitory effects of CBD on β-arrestin2 recruitment were concentration-dependent for both GPR3 and GPR6, with a higher potency for GPR6. These data show that CBD acts as an inverse agonist at both GPR3 and GPR6 receptors. These results demonstrate for the first time that both GPR3 and GPR6 are novel molecular targets for CBD. Our discovery that CBD acts as a novel inverse agonist on both GPR3 and GPR6 indicates that some of the potential therapeutic effects of CBD (e.g. treatment of Alzheimer's disease and Parkinson's disease) may be mediated through these important receptors.
Collapse
Affiliation(s)
- Alyssa S Laun
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, United States
| | - Zhao-Hui Song
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, United States.
| |
Collapse
|
22
|
Grondin Y, Bortoni ME, Sepulveda R, Ghelfi E, Bartos A, Cotanche D, Clifford RE, Rogers RA. Genetic Polymorphisms Associated with Hearing Threshold Shift in Subjects during First Encounter with Occupational Impulse Noise. PLoS One 2015; 10:e0130827. [PMID: 26121033 PMCID: PMC4488244 DOI: 10.1371/journal.pone.0130827] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 05/26/2015] [Indexed: 12/20/2022] Open
Abstract
Noise-induced hearing loss (NIHL) is the most significant occupational health issue worldwide. We conducted a genome-wide association study to identify single-nucleotide polymorphisms (SNPs) associated with hearing threshold shift in young males undergoing their first encounter with occupational impulse noise. We report a significant association of SNP rs7598759 (p < 5 x 10(-7); p = 0.01 after permutation and correction; Odds Ratio = 12.75) in the gene coding for nucleolin, a multifunctional phosphoprotein involved in the control of senescence and protection against apoptosis. Interestingly, nucleolin has been shown to mediate the anti-apoptotic effect of HSP70, a protein found to prevent ototoxicity and whose polymorphisms have been associated with susceptibility to NIHL. Increase in nucleolin expression has also been associated with the prevention of apoptosis in cells undergoing oxidative stress, a well-known metabolic sequela of noise exposure. To assess the potential role of nucleolin in hearing loss, we tested down-regulation of nucleolin in cochlear sensory cells HEI-OC1 under oxidative stress conditions and report increased sensitivity to cisplatin, a chemotherapeutic drug with ototoxic side effects. Additional SNPs were found with suggestive association (p < 5 x 10(-4)), of which 7 SNPs were located in genes previously reported to be related to NIHL and 43 of them were observed in 36 other genes previously not reported to be associated with NIHL. Taken together, our GWAS data and in vitro studies reported herein suggest that nucleolin is a potential candidate associated with NIHL in this population.
Collapse
Affiliation(s)
- Yohann Grondin
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard School of Public Health, 665 Huntington Ave, Boston, MA, 02115, United States of America
| | - Magda E. Bortoni
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard School of Public Health, 665 Huntington Ave, Boston, MA, 02115, United States of America
| | - Rosalinda Sepulveda
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard School of Public Health, 665 Huntington Ave, Boston, MA, 02115, United States of America
| | - Elisa Ghelfi
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard School of Public Health, 665 Huntington Ave, Boston, MA, 02115, United States of America
| | - Adam Bartos
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard School of Public Health, 665 Huntington Ave, Boston, MA, 02115, United States of America
| | - Douglas Cotanche
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard School of Public Health, 665 Huntington Ave, Boston, MA, 02115, United States of America
| | - Royce E. Clifford
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard School of Public Health, 665 Huntington Ave, Boston, MA, 02115, United States of America
- Department of Otolaryngology-Head and Neck Surgery, 34800 Bob Wilson Dr., Suite 200, Naval Medical Center, San Diego, CA, 92134, United States of America
| | - Rick A. Rogers
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard School of Public Health, 665 Huntington Ave, Boston, MA, 02115, United States of America
| |
Collapse
|
23
|
Ye C, Zhang Z, Wang Z, Hua Q, Zhang R, Xie X. Identification of a novel small-molecule agonist for human G protein-coupled receptor 3. J Pharmacol Exp Ther 2014; 349:437-43. [PMID: 24633425 DOI: 10.1124/jpet.114.213082] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
G protein-coupled receptor 3 (GPR3) is an orphan G protein-coupled receptor (GPCR) predominantly expressed in mammalian brain and oocytes. GPR3 plays important roles in these two organs and is known as a Gαs-coupled receptor-activated constitutively in cells. However, the signal transduction pathway and pharmacological function of GPR3 remain unclear because of the lack of a specific ligand. By use of a human embryonic kidney 293 cell line stably expressing FLAG-GPR3-green fluorescent protein, a chemical screening for GPR3 ligands was performed using homogeneous time-resolved fluorescence cAMP assay. Diphenyleneiodonium chloride (DPI) was identified as a novel agonist of GPR3 with weak or no cross-reactivity with other GPCRs. DPI was further characterized to activate several GPR3-mediated signal transduction pathways, including Ca(2+) mobilization, cAMP accumulation, membrane recruitment of β-arrestin2, and receptor desensitization. Parallel studies revealed that the activity of DPI is much more pronounced than sphingosine 1-phosphate, a previously reported GPR3 agonist. Our study identified a novel and specific agonist of GPR3, which provides a useful tool for further study of this orphan GPCR.
Collapse
Affiliation(s)
- Chenli Ye
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, School of Life Sciences and Technology, Tongji University, Shanghai, China (C.Y., Z.Z., Q.H., R.Z., X.X.); and CAS Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (Z.W., X.X.)
| | | | | | | | | | | |
Collapse
|
24
|
Weißenborn C, Ignatov T, Ochel HJ, Costa SD, Zenclussen AC, Ignatova Z, Ignatov A. GPER functions as a tumor suppressor in triple-negative breast cancer cells. J Cancer Res Clin Oncol 2014; 140:713-23. [PMID: 24553912 DOI: 10.1007/s00432-014-1620-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 02/10/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND The orphan, membrane-bound estrogen receptor (GPER) is expressed at high levels in a large fraction of breast cancer patients and its expression is favorable for patients' survival. METHODS We investigated the role of GPER as a potential tumor suppressor in triple-negative breast cancer cells MDA-MB-231 and MDA-MB-468 using cell cycle analysis and apoptosis assay. The constitutive activity of GPER was investigated. RESULTS GPER-specific activation with G-1 agonist inhibited breast cancer cell growth in concentration-dependent manner via induction of the cell cycle arrest in G2/M phase, enhanced phosphorylation of histone H3 and caspase-3-mediated apoptosis. Analysis of the methylation status of the GPER promoter in the triple-negative breast cancer cells and in tissues derived from breast cancer patients revealed that GPER amount is regulated by epigenetic mechanisms and GPER expression is inactivated by promoter methylation. Furthermore, GPER expression was induced by stress factors, such as radiation, and GPER amount inversely correlated with the p53 expression level. CONCLUSIONS Overall, our results establish the protective role in breast cancer tumorigenesis, and the cell surface expression of GPER makes it an excellent potential therapeutic target for triple-negative breast cancer.
Collapse
Affiliation(s)
- Christine Weißenborn
- Department of Obstetrics and Gynecology, University of Magdeburg, Gerhart-Hauptmann Str 35, Magdeburg, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
G-protein coupled receptor 6 deficiency alters striatal dopamine and cAMP concentrations and reduces dyskinesia in a mouse model of Parkinson's disease. Exp Neurol 2014; 257:1-9. [PMID: 24747358 DOI: 10.1016/j.expneurol.2014.04.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 04/07/2014] [Accepted: 04/10/2014] [Indexed: 11/21/2022]
Abstract
The orphan G-protein coupled receptor 6 (GPR6) is a constitutively active receptor which is positively coupled to the formation of cyclic adenosine-3',5'-monophosphate (cAMP). GPR6 is predominantly expressed in striatopallidal neurons. Here, we investigated neurochemical and behavioural effects of Gpr6 deficiency in mice. Gpr6 depletion decreased in vivo cAMP tissue concentrations (20%) in the striatum. An increase of striatal tissue dopamine concentrations (10%) was found in Gpr6(-/-) mice, whereas basal extracellular dopamine levels were not changed compared with Gpr6(+/+) mice, as shown by in vivo microdialysis. Western blot analyses revealed no alteration in the expression and subcellular localisation of the dopamine D2 receptor in the striatum of Gpr6(-/-) mice, and the number of tyrosine hydroxylase positive neurons in the substantia nigra was unchanged. DARPP-32 (dopamine and cAMP-regulated phosphoprotein of 32kDa) expression in the striatum of Gpr6(-/-) mice was not altered, however, a twofold increase in the phosphorylation of DARPP-32 at Thr34 was detected in Gpr6(-/-) compared with Gpr6(+/+) mice. Gpr6(-/-) mice showed higher locomotor activity in the open field, which persisted after treatment with the dopamine D2 receptor antagonist haloperidol. They also displayed reduced abnormal involuntary movements after apomorphine and quinpirole treatment in the mouse dyskinesia model of Parkinson's disease. In conclusion, the depletion of Gpr6 reduces cAMP concentrations in the striatum and alters the striatal dopaminergic system. Gpr6 deficiency causes an interesting behavioural phenotype in the form of enhanced motor activity combined with reduced abnormal involuntary movements. These findings could offer an opportunity for the treatment of Parkinson's disease beyond dopamine replacement.
Collapse
|
26
|
Weißenborn C, Ignatov T, Poehlmann A, Wege AK, Costa SD, Zenclussen AC, Ignatov A. GPER functions as a tumor suppressor in MCF-7 and SK-BR-3 breast cancer cells. J Cancer Res Clin Oncol 2014; 140:663-71. [PMID: 24515910 DOI: 10.1007/s00432-014-1598-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 01/24/2014] [Indexed: 12/17/2022]
Abstract
PURPOSE The orphan, membrane-bound estrogen receptor (GPER) is expressed at high levels in a large fraction of breast cancer patients, and its expression is favorable for patients' survival. We investigated the role of GPER as a potential tumor suppressor in MCF-7 and SK-BR-3 breast cancer cells. METHODS The effect of GPER agonist G-1 in cell culture was used to determine whether GPER inhibit cell growth. The methylation status of GPER promoter was investigated by methylation-specific PCR. RESULTS GPER-specific agonist G-1 inhibited breast cancer cell proliferation in concentration-dependent manner via induction of the cell cycle arrest in M-phase, enhanced phosphorylation of histone 3 and cell apoptosis. Analysis of the methylation status of the GPER promoter in MCF-7 and SK-BR-3 cells revealed that GPER expression is regulated by epigenetic mechanisms and GPER expression is inactivated by promoter methylation. Overall, our results are consistent with our recent findings in triple-negative breast cancer cells, and the cell surface expression of GPER makes it an excellent potential therapeutic target for non-triple-negative breast cancer.
Collapse
MESH Headings
- Apoptosis/drug effects
- Blotting, Western
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Cycle Checkpoints/drug effects
- Cell Proliferation/drug effects
- Cyclopentanes/pharmacology
- DNA Methylation/drug effects
- Female
- Humans
- Immunoenzyme Techniques
- Promoter Regions, Genetic/genetics
- Quinolines/pharmacology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptors, Estrogen/agonists
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Christine Weißenborn
- Department of Obstetrics and Gynecology, University Clinic Magdeburg, Gerhart-Hauptmann Str. 35, 39108, Magdeburg, Germany
| | | | | | | | | | | | | |
Collapse
|
27
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol 2013; 170:1459-581. [PMID: 24517644 PMCID: PMC3892287 DOI: 10.1111/bph.12445] [Citation(s) in RCA: 509] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. G protein-coupled receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
28
|
Engelstoft MS, Park WM, Sakata I, Kristensen LV, Husted AS, Osborne-Lawrence S, Piper PK, Walker AK, Pedersen MH, Nøhr MK, Pan J, Sinz CJ, Carrington PE, Akiyama TE, Jones RM, Tang C, Ahmed K, Offermanns S, Egerod KL, Zigman JM, Schwartz TW. Seven transmembrane G protein-coupled receptor repertoire of gastric ghrelin cells. Mol Metab 2013; 2:376-92. [PMID: 24327954 DOI: 10.1016/j.molmet.2013.08.006] [Citation(s) in RCA: 249] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 08/26/2013] [Indexed: 12/18/2022] Open
Abstract
The molecular mechanisms regulating secretion of the orexigenic-glucoregulatory hormone ghrelin remain unclear. Based on qPCR analysis of FACS-purified gastric ghrelin cells, highly expressed and enriched 7TM receptors were comprehensively identified and functionally characterized using in vitro, ex vivo and in vivo methods. Five Gαs-coupled receptors efficiently stimulated ghrelin secretion: as expected the β1-adrenergic, the GIP and the secretin receptors but surprisingly also the composite receptor for the sensory neuropeptide CGRP and the melanocortin 4 receptor. A number of Gαi/o-coupled receptors inhibited ghrelin secretion including somatostatin receptors SSTR1, SSTR2 and SSTR3 and unexpectedly the highly enriched lactate receptor, GPR81. Three other metabolite receptors known to be both Gαi/o- and Gαq/11-coupled all inhibited ghrelin secretion through a pertussis toxin-sensitive Gαi/o pathway: FFAR2 (short chain fatty acid receptor; GPR43), FFAR4 (long chain fatty acid receptor; GPR120) and CasR (calcium sensing receptor). In addition to the common Gα subunits three non-common Gαi/o subunits were highly enriched in ghrelin cells: GαoA, GαoB and Gαz. Inhibition of Gαi/o signaling via ghrelin cell-selective pertussis toxin expression markedly enhanced circulating ghrelin. These 7TM receptors and associated Gα subunits constitute a major part of the molecular machinery directly mediating neuronal and endocrine stimulation versus metabolite and somatostatin inhibition of ghrelin secretion including a series of novel receptor targets not previously identified on the ghrelin cell.
Collapse
Key Words
- 7TM, seven transmembrane segment
- BAC, bacterial artificial chromosome
- CCK, cholecystokinin
- CFMB, (S)-2-(4-chlorophenyl)-3,3-dimethyl-N-(5-phenylthiazol-2-yl)butamide
- CGRP, calcitonin gene-related peptide
- CHBA, 3-chloro-5-hydroxybenzoic acid
- Enteroendocrine
- G protein signaling
- GIP, glucose-dependent insulinotropic polypeptide
- GLP-1, glucagon-like peptide 1
- GPCR
- Ghrelin
- Metabolites
- PTx, Bordetella pertussis toxin
- PYY, peptide YY
- Secretion
- hrGFP, humanized Renilla reniformis green fluorescent protein
Collapse
Affiliation(s)
- Maja S Engelstoft
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark ; Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zhang BL, Li Y, Ding JH, Dong FL, Hou YJ, Jiang BC, Shi FX, Xu YX. Sphingosine 1-phosphate acts as an activator for the porcine Gpr3 of constitutively active G protein-coupled receptors. J Zhejiang Univ Sci B 2012; 13:555-66. [PMID: 22761247 DOI: 10.1631/jzus.b1100353] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We cloned the complete coding sequences of porcine Gpr3, Gpr6, and Gpr12 genes. Further, on the basis of their high levels of sequence similarity, these genes are identified as a subfamily of G protein-coupled receptors. These putative protein sequences also showed high sequence identity with other mammalian orthologs, including several highly conserved motifs. A wide expression of the Gpr3 gene in pigs was observed through tissue distribution analysis by reverse transcriptase-polymerase chain reaction (RT-PCR) and real-time PCR, specially in the brain, pituitary, fat, liver and oocyte, where its strong expression was observed. The Gpr3 gene was found to be located on chromosome 6 and a single exon coded for the entire open-reading frame. Expression of porcine Gpr3 in HEK293 cells resulted in constitutive activation of adenylate cyclase (AC) similar in amplitude to that produced by fully stimulated G(s)-coupled receptors. Moreover, sphingosine 1-phosphate (S1P) could increase AC activation via the constitutively active Gpr3 receptor. When a Gpr3-green fluorescent protein (GFP) construct was expressed in HEK293 cells, GFP-labeled Gpr3 protein was shown to be localized in the plasmalemma and subcellular membranes. After S1P treatment, agonist-mediated internalization could be visualized by confocal microscopy. In short, our findings suggest the porcine Gpr3, Gpr6, and Gpr12 genes as a subfamily of G protein-coupled receptors, and porcine Gpr3 was a constitutively active G protein-coupled receptor. Constitutive activation of AC and agonist-mediated internalization of Gpr3 receptor could be modulated by the S1P, suggesting that S1P might act as an activator for porcine Gpr3 receptor.
Collapse
Affiliation(s)
- Bao-le Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, China
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Lu X, Zhang N, Meng B, Dong S, Hu Y. Involvement of GPR12 in the regulation of cell proliferation and survival. Mol Cell Biochem 2012; 366:101-10. [PMID: 22430950 DOI: 10.1007/s11010-012-1287-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 03/02/2012] [Indexed: 01/27/2023]
Abstract
GPR12, a member of the orphan G-protein-coupled receptor family, constitutively activates the Gs protein and increases intracellular cyclic AMP concentrations. GPR12 can be activated by its known ligand-sphingosylphosphorylcholine, which regulates cellular physiological activities, including proliferation, neurite extension, cell clustering, and maintenance of meiotic arrest. However, signaling pathways involved in the GPR12-mediated physiological and biochemical changes are still not clearly illustrated. In the present study, heterologous GPR12 expression was demonstrated to promote proliferation and survival in human embryonic kidney 293 cells. Immunochemical analysis showed that Ki67, a prototypic cell cycle-related nuclear protein, might participate in the regulation of GPR12-mediated cell proliferation. Activation of extracellular signal-regulated protein kinase signaling and increased total Erk1/2 and B-cell lymphoma/leukemia-2 expression were also observed in HEK293 cells overexpressing human GPR12. In addition, we found that GPR12 promoted cell survival under serum deprivation, indicating that GPR12 may play a role in cell proliferation and survival.
Collapse
Affiliation(s)
- Xiaoming Lu
- Institutes for Advanced Interdisciplinary Research, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | | | | | | | | |
Collapse
|
31
|
Sphingolipid long chain base phosphates can regulate apoptotic-like programmed cell death in plants. Biochem Biophys Res Commun 2011; 410:574-80. [PMID: 21683064 DOI: 10.1016/j.bbrc.2011.06.028] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Accepted: 06/02/2011] [Indexed: 01/02/2023]
Abstract
Sphingolipids are ubiquitous components of eukaryotic cells and sphingolipid metabolites, such as the long chain base phosphate (LCB-P), sphingosine 1 phosphate (S1P) and ceramide (Cer) are important regulators of apoptosis in animal cells. This study evaluated the role of LCB-Ps in regulating apoptotic-like programmed cell death (AL-PCD) in plant cells using commercially available S1P as a tool. Arabidopsis cell cultures were exposed to a diverse array of cell death-inducing treatments (including Cer) in the presence of S1P. Rates of AL-PCD and cell survival were recorded using vital stains and morphological markers of AL-PCD. Internal LCB-P levels were altered in suspension cultured cells using inhibitors of sphingosine kinase and changes in rates of death in response to heat stress were evaluated. S1P reduced AL-PCD and promoted cell survival in cells subjected to a range of stresses. Treatments with inhibitors of sphingosine kinase lowered the temperature which induced maximal AL-PCD in cell cultures. The data supports the existence of a sphingolipid rheostat involved in controlling cell fate in Arabidopsis cells and that sphingolipid regulation of cell death may be a shared feature of both animal apoptosis and plant AL-PCD.
Collapse
|
32
|
Jee BC, Jo JW, Suh CS, Kim SH. Dose-Dependent Effect of Sphingosine-1-Phosphate in Mouse Oocyte Maturation Medium on Subsequent Embryo Development. Gynecol Obstet Invest 2011; 72:32-6. [DOI: 10.1159/000322222] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 10/18/2010] [Indexed: 12/13/2022]
|
33
|
Ignatov T, Eggemann H, Semczuk A, Smith B, Bischoff J, Roessner A, Costa SD, Kalinski T, Ignatov A. Role of GPR30 in endometrial pathology after tamoxifen for breast cancer. Am J Obstet Gynecol 2010; 203:595.e9-16. [PMID: 20965484 DOI: 10.1016/j.ajog.2010.07.034] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 06/19/2010] [Accepted: 07/21/2010] [Indexed: 11/17/2022]
Abstract
OBJECTIVE This study was undertaken to evaluate the potential role of G-protein-coupled estrogen receptor in endometrial pathology associated with tamoxifen treatment of breast cancer patients. STUDY DESIGN We investigated whether G-protein-coupled estrogen receptor plays a role in mediating proliferating effect of tamoxifen in endometrial carcinoma cells. These results were compared with the G-protein-coupled estrogen receptor expression pattern in endometrial tissue from a cohort of 95 breast cancer patients, who received tamoxifen or another adjuvant therapy. RESULTS In vitro tamoxifen significantly stimulated the mitogen-activated protein kinase phosphorylation and cell proliferation of endometrial cell lines via G-protein-coupled estrogen receptor. In vivo, there was a significant correlation between G-protein-coupled estrogen receptor expression and the tamoxifen-induced endometrial pathology (P = .006). Moreover, G-protein-coupled estrogen receptor positivity was predictive of an earlier development of symptoms, such as bleeding or suspect endometrial thickness, induced by tamoxifen therapy (P = .019). CONCLUSION G-protein-coupled estrogen receptor plays an important role in tamoxifen-induced endometrial abnormalities.
Collapse
Affiliation(s)
- Tanja Ignatov
- Department of Obstetrics and Gynecology, Otto-von-Guericke University, Magdeburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Pertwee RG, Howlett AC, Abood ME, Alexander SPH, Di Marzo V, Elphick MR, Greasley PJ, Hansen HS, Kunos G, Mackie K, Mechoulam R, Ross RA. International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid receptors and their ligands: beyond CB₁ and CB₂. Pharmacol Rev 2010; 62:588-631. [PMID: 21079038 PMCID: PMC2993256 DOI: 10.1124/pr.110.003004] [Citation(s) in RCA: 1233] [Impact Index Per Article: 82.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There are at least two types of cannabinoid receptors (CB(1) and CB(2)). Ligands activating these G protein-coupled receptors (GPCRs) include the phytocannabinoid Δ(9)-tetrahydrocannabinol, numerous synthetic compounds, and endogenous compounds known as endocannabinoids. Cannabinoid receptor antagonists have also been developed. Some of these ligands activate or block one type of cannabinoid receptor more potently than the other type. This review summarizes current data indicating the extent to which cannabinoid receptor ligands undergo orthosteric or allosteric interactions with non-CB(1), non-CB(2) established GPCRs, deorphanized receptors such as GPR55, ligand-gated ion channels, transient receptor potential (TRP) channels, and other ion channels or peroxisome proliferator-activated nuclear receptors. From these data, it is clear that some ligands that interact similarly with CB(1) and/or CB(2) receptors are likely to display significantly different pharmacological profiles. The review also lists some criteria that any novel "CB(3)" cannabinoid receptor or channel should fulfil and concludes that these criteria are not currently met by any non-CB(1), non-CB(2) pharmacological receptor or channel. However, it does identify certain pharmacological targets that should be investigated further as potential CB(3) receptors or channels. These include TRP vanilloid 1, which possibly functions as an ionotropic cannabinoid receptor under physiological and/or pathological conditions, and some deorphanized GPCRs. Also discussed are 1) the ability of CB(1) receptors to form heteromeric complexes with certain other GPCRs, 2) phylogenetic relationships that exist between CB(1)/CB(2) receptors and other GPCRs, 3) evidence for the existence of several as-yet-uncharacterized non-CB(1), non-CB(2) cannabinoid receptors; and 4) current cannabinoid receptor nomenclature.
Collapse
MESH Headings
- Cannabinoid Receptor Agonists
- Cannabinoid Receptor Antagonists
- Cannabinoid Receptor Modulators/metabolism
- Cannabinoids/metabolism
- Humans
- Ligands
- Phylogeny
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/antagonists & inhibitors
- Receptor, Cannabinoid, CB2/metabolism
- Receptors, Cannabinoid/metabolism
- Terminology as Topic
Collapse
Affiliation(s)
- R G Pertwee
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Prasad BM, Hollins B, Lambert NA. Methods to detect cell surface expression and constitutive activity of GPR6. Methods Enzymol 2010; 484:179-95. [PMID: 21036233 DOI: 10.1016/b978-0-12-381298-8.00010-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
GPR6 is a constitutively active Gs-coupled receptor that can signal from intracellular compartments. We present different methods used to study cell surface expression of receptors and other membrane proteins. A comparison of these methods shows that methods based on susceptibility to proteolytic enzymes are more efficient at providing estimates of cell surface expression than the commonly used cell surface biotinylation method. We also present different methods that can be used to detect constitutive activity of Gs-coupled receptors. Imaging-based assays to detect intracellular cyclic AMP accumulation are well suited to study signaling at a single cell level. These assays are particularly useful when the cells of interest form a small fraction of the culture such as primary cultures with low transfection efficiency.
Collapse
Affiliation(s)
- Balakrishna M Prasad
- Department of Clinical Investigation, Eisenhower Army Medical Center, Georgia, USA
| | | | | |
Collapse
|
36
|
Ignatov A, Ignatov T, Roessner A, Costa SD, Kalinski T. Role of GPR30 in the mechanisms of tamoxifen resistance in breast cancer MCF-7 cells. Breast Cancer Res Treat 2009; 123:87-96. [PMID: 19911269 DOI: 10.1007/s10549-009-0624-6] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Accepted: 10/28/2009] [Indexed: 01/01/2023]
Abstract
Tamoxifen is the most frequently used anti-hormonal drug for treatment of women with hormone-dependent breast cancer. The aim of this study is to investigate the mechanism of tamoxifen resistance and the impact of the new estrogen G-protein coupled receptor (GPR30). MCF-7 cells were continuously exposed to tamoxifen for 6 months to induce resistance to the inhibitory effect of tamoxifen. These tamoxifen-resistant cells (TAM-R) exhibited enhanced sensitivity to 17-ss-estradiol and GPR30 agonist, G1, when compared to the parental cells. In TAM-R cells, tamoxifen was able to stimulate the cell growth and MAPK phosphorylation. These effects were abolished by EGFR inhibitor AG1478, GPR30 anti-sense oligonucleotide, and the selective c-Src inhibitor PP2. Only EGFR basal expression was slightly elevated in the TAM-R cells, whereas GPR30 expression and the basal phosphorylation of Akt and MAPK remained unchanged when compared to the parental cells. Interestingly, estrogen treatment significantly increased GPR30 translocation to the cell surface, which was stronger in TAM-R cells. Continuous treatment of MCF-7 cells with GPR30 agonist G1 mimics the long-term treatment with tamoxifen and increases drastically its agonistic activity. This data suggests the important role of GPR30/EGFR receptor signaling in the development of tamoxifen resistance. The inhibition of this pathway is a valid option to improve anti-hormone response in breast cancer.
Collapse
Affiliation(s)
- Atanas Ignatov
- Department of Obstetrics and Gynecology, Otto-von-Guericke University, G-Hauptmann Str 35, 39108 Magdeburg, Germany.
| | | | | | | | | |
Collapse
|
37
|
Im DS. New intercellular lipid mediators and their GPCRs: An update. Prostaglandins Other Lipid Mediat 2009; 89:53-6. [DOI: 10.1016/j.prostaglandins.2009.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Revised: 01/24/2009] [Accepted: 01/25/2009] [Indexed: 01/08/2023]
|
38
|
Yin H, Chu A, Li W, Wang B, Shelton F, Otero F, Nguyen DG, Caldwell JS, Chen YA. Lipid G protein-coupled receptor ligand identification using beta-arrestin PathHunter assay. J Biol Chem 2009; 284:12328-38. [PMID: 19286662 DOI: 10.1074/jbc.m806516200] [Citation(s) in RCA: 248] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A growing number of orphan G-protein-coupled receptors (GPCRs) have been reported to be activated by lipid ligands, such as lysophosphatidic acid, sphingosine 1-phosphate (S1P), and cannabinoids, for which there are already well established receptors. These new ligand claims are controversial due to either lack of independent confirmations or conflicting reports. We used the beta-arrestin PathHunter assay system, a newly developed, generic GPCR assay format that measures beta-arrestin binding to GPCRs, to evaluate lipid receptor and ligand pairing. This assay eliminates interference from endogenous receptors on the parental cells because it measures a signal that is specifically generated by the tagged receptor and is immediately downstream of receptor activation. We screened a large number of newly "deorphaned" receptors (GPR23, GPR92, GPR55, G2A, GPR18, GPR3, GPR6, GPR12, and GPR63) and control receptors against a collection of approximately 400 lipid molecules to try to identify the receptor ligand in an unbiased fashion. GPR92 was confirmed to be a lysophosphatidic acid receptor with weaker responses to farnesyl pyrophosphate and geranylgeranyl diphosphate. The putative cannabinoid receptor GPR55 responded strongly to AM251, rimonabant, and lysophosphatidylinositol but only very weakly to endocannabinoids. G2A receptor was confirmed to be an oxidized free fatty acid receptor. In addition, we discovered that 3,3'-diindolylmethane, a dietary molecule from cruciferous vegetables, which has known anti-cancer properties, to be a CB(2) receptor partial agonist, with binding affinity around 1 microm. The anti-inflammatory effect of 3,3'-diindolylmethane in RAW264.7 cells was shown to be partially mediated by CB(2).
Collapse
Affiliation(s)
- Hong Yin
- GPCR Platform, Genomics Institute of the Novartis Research Foundation, San Diego, California 92121, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Molecular profiling of striatonigral and striatopallidal medium spiny neurons past, present, and future. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2009; 89:1-35. [PMID: 19900613 DOI: 10.1016/s0074-7742(09)89001-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Defining distinct molecular properties of the two striatal medium spiny neurons (MSNs) has been a challenging task for basal ganglia (BG) neuroscientists. Identifying differential molecular components in each MSN subtype is crucial for BG researchers to understand functional properties of these two neurons. The two MSN populations are morphologically identical except in their projections through the direct verses indirect BG pathways and they are heterogeneously dispersed throughout the dorsal striatum (dStr) and nucleus accumbens (NAc). These characteristics have made it difficult for researchers to distinguish and isolate these two neuronal populations thereby hindering progress toward a more comprehensive understanding of their differential molecular properties. Researchers began to investigate molecular differences in the striatonigral and striatopallidal neurons using in situ hybridization (ISH) techniques and single cell reverse transcription-polymerase chain reaction (scRT-PCR). Currently the field is utilizing more advanced techniques for large-scale gene expression studies including fluorescence activated cell sorting (FACS) of MSNs, from which RNA is purified, from fluorescent reporter transgenic mice or use of transgenic mice in which ribosomes from each MSN are tagged and can be immunoprecipitated followed by RNA isolation, a technique termed translating ribosomal affinity purification (TRAP). Additionally, the availability of fluorescent reporter mice for each MSN subtype is allowing, scientists to perform more accurate histology studies evaluating differential protein expression and signaling changes in each cell subtype. Finally, researchers are able to evaluate the role of specific genes in vivo by utilizing cell type-specific mouse models including Cre driver lines that can be crossed with conditional overexpression or knockout systems. This is a very exciting time in the BG field because researchers are well equipped with the most progressive tools to comprehensively evaluate molecular components in the two MSNs and their consequence on BG functional output in the normal, diseased, and developing brain.
Collapse
|
40
|
Heiman M, Schaefer A, Gong S, Peterson JD, Day M, Ramsey KE, Suárez-Fariñas M, Schwarz C, Stephan DA, Surmeier DJ, Greengard P, Heintz N. A translational profiling approach for the molecular characterization of CNS cell types. Cell 2008; 135:738-48. [PMID: 19013281 DOI: 10.1016/j.cell.2008.10.028] [Citation(s) in RCA: 904] [Impact Index Per Article: 53.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 07/18/2008] [Accepted: 10/28/2008] [Indexed: 01/01/2023]
Abstract
The cellular heterogeneity of the brain confounds efforts to elucidate the biological properties of distinct neuronal populations. Using bacterial artificial chromosome (BAC) transgenic mice that express EGFP-tagged ribosomal protein L10a in defined cell populations, we have developed a methodology for affinity purification of polysomal mRNAs from genetically defined cell populations in the brain. The utility of this approach is illustrated by the comparative analysis of four types of neurons, revealing hundreds of genes that distinguish these four cell populations. We find that even two morphologically indistinguishable, intermixed subclasses of medium spiny neurons display vastly different translational profiles and present examples of the physiological significance of such differences. This genetically targeted translating ribosome affinity purification (TRAP) methodology is a generalizable method useful for the identification of molecular changes in any genetically defined cell type in response to genetic alterations, disease, or pharmacological perturbations.
Collapse
Affiliation(s)
- Myriam Heiman
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Padmanabhan S, Myers AG, Prasad BM. Constitutively active GPR6 is located in the intracellular compartments. FEBS Lett 2008; 583:107-12. [DOI: 10.1016/j.febslet.2008.11.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 11/18/2008] [Indexed: 10/21/2022]
|
42
|
Borges S, Lindstrom S, Walters C, Warrier A, Wilson M. Discrete influx events refill depleted Ca2+ stores in a chick retinal neuron. J Physiol 2007; 586:605-26. [PMID: 18033816 DOI: 10.1113/jphysiol.2007.143339] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The depletion of ER Ca2+ stores, following the release of Ca2+ during intracellular signalling, triggers the Ca2+ entry across the plasma membrane known as store-operated calcium entry (SOCE). We show here that brief, local [Ca2+]i increases (motes) in the thin dendrites of cultured retinal amacrine cells derived from chick embryos represent the Ca2+ entry events of SOCE and are initiated by sphingosine-1-phosphate (S1P), a sphingolipid with multiple cellular signalling roles. Externally applied S1P elicits motes but not through a G protein-coupled membrane receptor. The endogenous precursor to S1P, sphingosine, also elicits motes but its action is suppressed by dimethylsphingosine (DMS), an inhibitor of sphingosine phosphorylation. DMS also suppresses motes induced by store depletion and retards the refilling of depleted stores. These effects are reversed by exogenously applied S1P. In these neurons formation of S1P is a step in the SOCE pathway that promotes Ca2+ entry in the form of motes.
Collapse
|
43
|
Lobo MK, Cui Y, Ostlund SB, Balleine BW, Yang XW. Genetic control of instrumental conditioning by striatopallidal neuron-specific S1P receptor Gpr6. Nat Neurosci 2007; 10:1395-7. [PMID: 17934457 DOI: 10.1038/nn1987] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Accepted: 08/30/2007] [Indexed: 11/09/2022]
Abstract
Instrumental conditioning allows animals to learn about the consequences of their own actions, but the underpinning molecular mechanisms remain elusive. Here we show that the sphingosine-1-phosphate (S1P) receptor Gpr6 is selectively expressed in the striatopallidal neurons in the striatum. Gpr6-deficient mice showed reduced striatal cyclic AMP production in vitro and selective alterations in instrumental conditioning in vivo. Thus, Gpr6 is the first striatopallidal neuron-specific genetic regulator of instrumental conditioning in a mammal.
Collapse
Affiliation(s)
- Mary Kay Lobo
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience & Human Behavior, University of California at Los Angeles, 695 Charles Young Drive South, Los Angeles, California 90095, USA
| | | | | | | | | |
Collapse
|
44
|
Kusner DJ, Thompson CR, Melrose NA, Pitson SM, Obeid LM, Iyer SS. The Localization and Activity of Sphingosine Kinase 1 Are Coordinately Regulated with Actin Cytoskeletal Dynamics in Macrophages. J Biol Chem 2007; 282:23147-62. [PMID: 17519232 DOI: 10.1074/jbc.m700193200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The physiologic and pathologic functions of sphingosine kinase (SK) require translocation to specific membrane compartments. We tested the hypothesis that interactions with actin filaments regulate the localization of SK1 to membrane surfaces, including the plasma membrane and phagosome. Macrophage activation is accompanied by a marked increase in association of SK1 with actin filaments. Catalytically-inactive (CI)- and phosphorylation-defective (PD)-SK1 mutants exhibited reductions in plasma membrane translocation, colocalization with cortical actin filaments, membrane ruffling, and lamellipodia formation, compared with wild-type (WT)-SK1. However, translocation of CI- and PD-SK1 to phagosomes were equivalent to WT-SK1. SK1 exhibited constitutive- and stimulus-enhanced association with actin filaments and F-actin-enriched membrane fractions in both intact macrophages and a novel in vitro assay. In contrast, SK1 bound G-actin only under stimulated conditions. Actin inhibitors disrupted SK1 localization and modulated its activity. Conversely, reduction of SK1 levels or activity via RNA interference or specific chemical inhibition resulted in dysregulation of actin filaments. Thus, the localization and activity of SK1 are coordinately regulated with actin dynamics during macrophage activation.
Collapse
Affiliation(s)
- David J Kusner
- Inflammation Program, Division of Infectious Diseases, Department of Internal Medicine, University of Iowa Carver College of Medicine and Veterans Affairs Medical Center, Iowa City, Iowa 52245, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Suga H, Haga T. Ligand screening system using fusion proteins of G protein-coupled receptors with G protein alpha subunits. Neurochem Int 2007; 51:140-64. [PMID: 17659814 DOI: 10.1016/j.neuint.2007.06.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2007] [Revised: 06/07/2007] [Accepted: 06/08/2007] [Indexed: 01/04/2023]
Abstract
G protein-coupled receptors (GPCRs) constitute one of the largest families of genes in the human genome, and are the largest targets for drug development. Although a large number of GPCR genes have recently been identified, ligands have not yet been identified for many of them. Various assay systems have been employed to identify ligands for orphan GPCRs, but there is still no simple and general method to screen for ligands of such GPCRs, particularly of G(i)-coupled receptors. We have examined whether fusion proteins of GPCRs with G protein alpha subunit (Galpha) could be utilized for ligand screening and showed that the fusion proteins provide an effective method for the purpose. This article focuses on the followings: (1) characterization of GPCR genes and GPCRs, (2) identification of ligands for orphan GPCRs, (3) characterization of GPCR-Galpha fusion proteins, and (4) identification of ligands for orphan GPCRs using GPCR-Galpha fusion proteins.
Collapse
Affiliation(s)
- Hinako Suga
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | | |
Collapse
|
46
|
Tanaka S, Ishii K, Kasai K, Yoon SO, Saeki Y. Neural expression of G protein-coupled receptors GPR3, GPR6, and GPR12 up-regulates cyclic AMP levels and promotes neurite outgrowth. J Biol Chem 2007; 282:10506-15. [PMID: 17284443 DOI: 10.1074/jbc.m700911200] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cyclic AMP regulates multiple neuronal functions, including neurite outgrowth and axonal regeneration. GPR3, GPR6, and GPR12 make up a family of constitutively active G protein-coupled receptors (GPCRs) that share greater than 50% identity and 65% similarity at the amino acid level. They are highly expressed in the central nervous system, and their expression in various cell lines results in constitutive stimulation of cAMP production. When the constitutively active GPCRs were overexpressed in rat cerebellar granule neurons in culture, the transfected neurons exhibited significantly enhanced neurite outgrowth and overcame growth inhibition caused by myelin-associated glycoprotein. GPR12-mediated neurite outgrowth was the most prominent and was shown to depend on G(s) and cAMP-dependent protein kinase. Moreover, the GPR12-mediated rescue from myelin-associated glycoprotein inhibition was attributable to cAMP-dependent protein kinase-mediated inhibition of the small GTPase, RhoA. Among the three receptors, GPR3 was revealed to be enriched in the developing rat cerebellar granule neurons. When the endogenous GPR3 was knocked down, significant reduction of neurite growth was observed, which was reversed by expression of either GPR3 or GPR12. Taken together, our results indicate that expression of the constitutively active GPCRs up-regulates cAMP production in neurons, stimulates neurite outgrowth, and counteracts myelin inhibition. Further characterization of the GPCRs in developing and injured mammalian neurons should provide insights into how basal cAMP levels are regulated in neurons and could establish a firm scientific foundation for applying receptor biology to treatment of various neurological disorders.
Collapse
Affiliation(s)
- Shigeru Tanaka
- Dardinger Laboratory for Neuro-oncology and Neurosciences, Department of Neurological Surgery, the Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | |
Collapse
|
47
|
Meyer zu Heringdorf D, Jakobs KH. Lysophospholipid receptors: signalling, pharmacology and regulation by lysophospholipid metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2006; 1768:923-40. [PMID: 17078925 DOI: 10.1016/j.bbamem.2006.09.026] [Citation(s) in RCA: 282] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2006] [Accepted: 09/28/2006] [Indexed: 12/17/2022]
Abstract
The lysophospholipids, sphingosine-1-phosphate (S1P), lysophosphatidic acid (LPA), sphingosylphosphorylcholine (SPC) and lysophosphatidylcholine (LPC), activate diverse groups of G-protein-coupled receptors that are widely expressed and regulate decisive cellular functions. Receptors of the endothelial differentiation gene family are activated by S1P (S1P(1-5)) or LPA (LPA(1-3)); two more distantly related receptors are activated by LPA (LPA(4/5)); the GPR(3/6/12) receptors have a high constitutive activity but are further activated by S1P and/or SPC; and receptors of the OGR1 cluster (OGR1, GPR4, G2A, TDAG8) appear to be activated by SPC, LPC, psychosine and/or protons. G-protein-coupled lysophospholipid receptors regulate cellular Ca(2+) homoeostasis and the cytoskeleton, proliferation and survival, migration and adhesion. They have been implicated in development, regulation of the cardiovascular, immune and nervous systems, inflammation, arteriosclerosis and cancer. The availability of S1P and LPA at their G-protein-coupled receptors is regulated by enzymes that generate or metabolize these lysophospholipids, and localization plays an important role in this process. Besides FTY720, which is phosphorylated by sphingosine kinase-2 and then acts on four of the five S1P receptors of the endothelial differentiation gene family, other compounds have been identified that interact with more ore less selectivity with lysophospholipid receptors.
Collapse
|
48
|
Süsens U, Hermans-Borgmeyer I, Urny J, Schaller HC. Characterisation and differential expression of two very closely related G-protein-coupled receptors, GPR139 and GPR142, in mouse tissue and during mouse development. Neuropharmacology 2006; 50:512-20. [PMID: 16378626 DOI: 10.1016/j.neuropharm.2005.11.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2005] [Revised: 10/31/2005] [Accepted: 11/02/2005] [Indexed: 11/17/2022]
Abstract
By searching the human and mouse genomic databases we found two G-protein-coupled receptors, GPR139 and GPR142, with characteristic motifs of the rhodopsin family of receptors. The gene for GPR139 maps to chromosome 7F1 of mouse and 16p12.3 of human and that for GPR142 to 11E2 of mouse and 17q25.1 of human. We isolated GPR139 from a cDNA library of adult mouse brain and GPR142 from a cDNA library of brains from 15-day-old mouse embryos. GPR139 mRNA was predominantly expressed in specific areas of human and mouse brains, whereas GPR142 mRNA showed a more ubiquitous expression both in the brain and in various peripheral glands and organs. A 50% identity and a 67% homology at the amino-acid level between the two receptors and only 20-25% identity with other G-protein-coupled receptors established them as a new subbranch within the phylogenetic tree and hints at a common or similar ligand(s). Preliminary results suggest that the cognate ligand is present in brain extracts and is, most likely, a small peptide. GPR139 signal transduction in Chinese hamster ovary cells requires coupling to an inhibitory G-protein and is mediated by phospholipase C. Dimer formation may be necessary for proper function.
Collapse
Affiliation(s)
- Ute Süsens
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Germany
| | | | | | | |
Collapse
|
49
|
Rezgaoui M, Süsens U, Ignatov A, Gelderblom M, Glassmeier G, Franke I, Urny J, Imai Y, Takahashi R, Schaller HC. The neuropeptide head activator is a high-affinity ligand for the orphan G-protein-coupled receptor GPR37. J Cell Sci 2006; 119:542-9. [PMID: 16443751 DOI: 10.1242/jcs.02766] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The neuropeptide head activator (HA) is a mitogen for mammalian cell lines of neuronal or neuroendocrine origin. HA signalling is mediated by a G-protein-coupled receptor (GPCR). Orphan GPCRs with homology to peptide receptors were screened for HA interaction. Electrophysiological recordings in frog oocytes and in mammalian cell lines as well as Ca2+ mobilisation assays revealed nanomolar affinities of HA to GPR37. HA signal transduction through GPR37 was mediated by an inhibitory G protein and required Ca2+ influx through a channel of the transient receptor potential (TRP) family. It also required activation of Ca2+-dependent calmodulin kinase and phosphoinositide 3-kinase. Respective inhibitors blocked HA signalling and HA-induced mitosis in GPR37-expressing cells. HA treatment resulted in internalisation of GPR37. Overexpression of GPR37 led to aggregate formation, retention of the receptor in the cytoplasm and low survival rates of transfected cells, confirming the notion that misfolded GPR37 contributes to cell death, as observed in Parkinson's disease.
Collapse
Affiliation(s)
- Meriem Rezgaoui
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Inagaki Y, Pham T, Fujiwara Y, Kohno T, Osborne D, Igarashi Y, Tigyi G, Parrill A. Sphingosine 1-phosphate analogue recognition and selectivity at S1P4 within the endothelial differentiation gene family of receptors. Biochem J 2005; 389:187-95. [PMID: 15733055 PMCID: PMC1184551 DOI: 10.1042/bj20050046] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Synergistic computational and experimental studies provided previously unforeseen details concerning the structural basis of S1P (sphingosine 1-phosphate) recognition by the S1P4 G-protein-coupled receptor. Similarly to reports on the S1P1 receptor, cationic and anionic residues in the third transmembrane domain (R3.28 and E3.29 at positions 124 and 125) form ion pairs with the phosphate and ammonium of S1P, and alanine mutations at these positions abolished specific S1P binding, S1P-induced receptor activation and cell migration. Unlike findings on the S1P1 receptor, no cationic residue in the seventh transmembrane domain interacts with the phosphate. Additionally, two previously undiscovered interactions with the S1P polar headgroup have been identified. Trp186 at position 4.64 in the fourth transmembrane domain interacts by a cation-pi interaction with the ammonium group of S1P. Lys204 at position 5.38 forms an ion pair with the S1P. The S1P4 and S1P1 receptors show differences in binding-pocket shape and electrostatic distributions that correlate with the published structure-activity relationships. In particular, the binding pocket of mS1P4 (mouse S1P4) has recognition sites for the anionic phosphate and cationic ammonium groups that are equidistant from the end of the non-polar tail. In contrast, the binding pocket of hS1P1 (human S1P4) places the ammonium recognition site 2 A (1 A=0.1 nm) closer to the end of the non-polar tail than the phosphate recognition site.
Collapse
Affiliation(s)
- Yuichi Inagaki
- *Department of Biomembrane and Biofunctional Chemistry, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12-jo, Nishi 6-choume, Kita-ku, Sapporo 060-0812, Japan
| | - TrucChi T. Pham
- †Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152, U.S.A
| | - Yuko Fujiwara
- ‡Department of Physiology, University of Tennessee Health Sciences Center, Memphis, TN 38163, U.S.A
| | - Takayuki Kohno
- *Department of Biomembrane and Biofunctional Chemistry, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12-jo, Nishi 6-choume, Kita-ku, Sapporo 060-0812, Japan
| | - Daniel A. Osborne
- †Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152, U.S.A
| | - Yasuyuki Igarashi
- *Department of Biomembrane and Biofunctional Chemistry, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12-jo, Nishi 6-choume, Kita-ku, Sapporo 060-0812, Japan
| | - Gabor Tigyi
- †Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152, U.S.A
- ‡Department of Physiology, University of Tennessee Health Sciences Center, Memphis, TN 38163, U.S.A
| | - Abby L. Parrill
- †Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152, U.S.A
- ‡Department of Physiology, University of Tennessee Health Sciences Center, Memphis, TN 38163, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|