51
|
Anti-atherosclerotic action of GW9508 - Free fatty acid receptors activator - In apoE-knockout mice. Pharmacol Rep 2019; 71:551-555. [PMID: 31129318 DOI: 10.1016/j.pharep.2019.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND In the past two decades, enhanced understanding of the biology of G-protein-coupled receptors (GPRs) has led to the identification of several such receptors as novel targets for free fatty acids (FFAs). Two GPRs, FFAR1 and FFAR4, have received special attention in the context of chronic inflammatory diseases, thanks to their anti-inflammatory activities. METHODS The present study investigates the influence of prolonged treatment with GW9508 - agonist of FFAR1 and FFAR4 - on the development of atherosclerosis plaque in apoE-knockout mice, using morphometric and molecular methods. RESULTS GW9508 administration has led to the reduction of atheroscletoric plaque size in an apoE-knockout mice model. Moreover, a FFAR1/FFAR4 agonist reduced the content of macrophages by almost 20%, attributed by immunohistochemical phenotyping to the pro-inflammatory M1-like activation state macrophages. CONCLUSIONS Prolonged administration of GW9508 resulted in significant amelioration of atherogenesis, providing evidence that the strategy based on macrophage phenotype switching toward an M2-like activation state via stimulation of FFAR1/FFAR4 receptors holds promise for a new approach to the prevention or treatment of atherosclerosis.
Collapse
|
52
|
Erdei AI, Borbély A, Magyar A, Szűcs E, Ötvös F, Gombos D, Al-Khrasani M, Stefanucci A, Dimmito MP, Luisi G, Mollica A, Benyhe S. Biochemical and pharmacological investigation of novel nociceptin/OFQ analogues and N/OFQ-RYYRIK hybrid peptides. Peptides 2019; 112:106-113. [PMID: 30513351 DOI: 10.1016/j.peptides.2018.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 10/27/2022]
Abstract
The endogenous ligand nociceptin (N/OFQ) and a positively charged synthetic peptide RYYRIK are both selective for the nociceptin opioid receptor (NOPr). Despite their structural dissimilarity, N/OFQ and RYYRIK compete for the same binding site of NOP receptor possessing full and partial agonistic character, respectively. In the view of the message-address concept, hybrid peptide constructs were probed for the NOP receptor combining different regions of N/OFQ and RYYRIK related peptide sequences. Nine novel nociceptin- or Ac-RYYRIK-NH2 peptide variants or hybrid peptides were synthesized and characterized. Peptides P2 and P8 contain fragments of native N/OFQ. The other seven analogues (P1, P3-7, P9) are composed of Ac-RYYRIK-NH2 fragments and parts of the original nociceptin sequence. The analogues were characterized in receptor binding assays and G-protein activation experiments on rat brain membranes, as well as by electrically stimulated mouse vas deferens bioassay. In receptor binding assays ligands P2, P4, P6 (Ki 0.37 nM) and P7 showed higher affinity (Ki 0.65 nM, 0.6 nM, 0.37 nM and 0.44 nM, respectively) for NOP receptor than their parent compounds N/OFQ (Ki 2.8 nM) or Ac-RYYRIK-NH2 (Ki 4.2 nM). In [35S]GTPγS binding experiments P2 and P3 behaved as full agonists. The other variants exhibited partial agonist properties characterized by submaximal stimulatory effects. In mouse vas deferens bioassay only P2 showed agonist activity. P4, P5, P6 inhibited the biological activity of N/OFQ more effectively than the NOP receptor selective antagonist JTC-801. In summary, hybrid peptides P4, P5 and P6 proved to be NOP receptor partial agonists even antagonists, while P2 peptide retained the full agonist property.
Collapse
Affiliation(s)
- Anna I Erdei
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62., Hungary; Doctoral School of Theoretical Medicine, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Adina Borbély
- MTA-ELTE Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös Loránd University, H-1117, Budapest, Pázmány Péter sétány 1/A, Hungary
| | - Anna Magyar
- MTA-ELTE Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös Loránd University, H-1117, Budapest, Pázmány Péter sétány 1/A, Hungary
| | - Edina Szűcs
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62., Hungary; Doctoral School of Theoretical Medicine, University of Szeged, Faculty of Medicine, Szeged, Hungary
| | - Ferenc Ötvös
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62., Hungary
| | - Dávid Gombos
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62., Hungary
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1445 Budapest, Nagyvárad tér 4., Hungary
| | - Azzurra Stefanucci
- Dipartimento di Farmacia, Università̀ degli Studi "G. d'Annunzio" di Chieti-Pescara, Via dei Vestini 31, Chieti, 66100, Italy
| | - Marilisa Pia Dimmito
- Dipartimento di Farmacia, Università̀ degli Studi "G. d'Annunzio" di Chieti-Pescara, Via dei Vestini 31, Chieti, 66100, Italy
| | - Grazia Luisi
- Dipartimento di Farmacia, Università̀ degli Studi "G. d'Annunzio" di Chieti-Pescara, Via dei Vestini 31, Chieti, 66100, Italy
| | - Adriano Mollica
- Dipartimento di Farmacia, Università̀ degli Studi "G. d'Annunzio" di Chieti-Pescara, Via dei Vestini 31, Chieti, 66100, Italy
| | - Sándor Benyhe
- Institute of Biochemistry, Biological Research Center, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62., Hungary.
| |
Collapse
|
53
|
Ye N, Li B, Mao Q, Wold EA, Tian S, Allen JA, Zhou J. Orphan Receptor GPR88 as an Emerging Neurotherapeutic Target. ACS Chem Neurosci 2019; 10:190-200. [PMID: 30540906 DOI: 10.1021/acschemneuro.8b00572] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although G protein-coupled receptors (GPCRs) are recognized as pivotal drug targets involved in multiple physiological and pathological processes, the majority of GPCRs including orphan GPCRs (oGPCRs) are unexploited. GPR88, a brain-specific oGPCR with particularly robust expression in the striatum, regulates diverse brain and behavioral functions, including cognition, mood, movement control, and reward-based learning, and is thus emerging as a novel drug target for central nervous system disorders including schizophrenia, Parkinson's disease, anxiety, and addiction. Nevertheless, no effective GPR88 synthetic ligands have yet entered into clinical trials, and GPR88 endogenous ligands remain unknown. Despite the recent discovery and early stage study of several GPR88 agonists, such as 2-PCCA, RTI-13951-33, and phenylglycinol derivatives, further research into GPR88 pharmacology, medicinal chemistry, and chemical biology is urgently needed to yield structurally diversified GPR88-specific ligands. Drug-like pharmacological tool function and relevant signaling elucidation will also accelerate the evaluation of this receptor as a viable neurotherapeutic target.
Collapse
Affiliation(s)
- Na Ye
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Bang Li
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Qi Mao
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Eric A. Wold
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Sheng Tian
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - John A. Allen
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
54
|
Abstract
The nociceptin/orphanin FQ (N/OFQ) peptide receptor (NOP) is a G protein-coupled receptor involved in the regulation of several physiological functions and pathological conditions. Thus, researchers from academia and industry are pursuing NOP to discover and study novel pharmacological entities. In a multidisciplinary effort of pharmacologists, medicinal chemists, and molecular and structural biologists the mechanisms of NOP activation and inhibition have been, at least partially, disentangled. Here, we review the in vitro methodologies employed, which have contributed to our understanding of this target. We hope this chapter guides the reader through the mostly established assay platforms to investigate NOP pharmacology, and gives some hints taking advantage from what has already illuminated the function of other GPCRs. We analyzed the pharmacological results obtained with a large panel of NOP ligands investigated in several assays including receptor binding, stimulation of GTPγS binding, decrease of cAMP levels, calcium flux stimulation via chimeric G proteins, NOP/G protein and NOP/β-arrestin interaction, label-free assays such as dynamic mass redistribution, and bioassays such as the electrically stimulated mouse vas deferens.
Collapse
Affiliation(s)
- Davide Malfacini
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Girolamo Caló
- Section of Pharmacology, Department of Medical Sciences, National Institute of Neurosciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
55
|
Ren Z, Chen L, Wang Y, Wei X, Zeng S, Zheng Y, Gao C, Liu H. Activation of the Omega-3 Fatty Acid Receptor GPR120 Protects against Focal Cerebral Ischemic Injury by Preventing Inflammation and Apoptosis in Mice. THE JOURNAL OF IMMUNOLOGY 2018; 202:747-759. [PMID: 30598514 DOI: 10.4049/jimmunol.1800637] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 11/22/2018] [Indexed: 12/28/2022]
Abstract
G protein-coupled receptor 120 (GPR120) has been shown to negatively regulate inflammation and apoptosis, but its role in cerebral ischemic injury remains unclear. Using an in vivo model of middle cerebral artery occlusion (MCAO) and an in vitro model of oxygen-glucose deprivation (OGD), we investigated the potential role and molecular mechanisms of GPR120 in focal cerebral ischemic injury. Increased GPR120 expression was observed in microglia and neurons following MCAO-induced ischemia in wild type C57BL/6 mice. Treatment with docosahexaenoic acid (DHA) inhibited OGD-induced inflammatory response in primary microglia and murine microglial BV2 cells, whereas silencing of GPR120 strongly exacerbated the inflammation induced by OGD and abolished the anti-inflammatory effects of DHA. Mechanistically, DHA inhibited OGD-induced inflammation through GPR120 interacting with β-arrestin2. In addition to its anti-inflammatory function, GPR120 also played a role in apoptosis as its knockdown impaired the antiapoptotic effect of DHA in OGD-induced rat pheochromocytoma (PC12) cells. Finally, using MCAO mouse model, we demonstrated that GPR120 activation protected against focal cerebral ischemic injury by preventing inflammation and apoptosis. Our study indicated that pharmacological targeting of GPR120 may provide a novel approach for the treatment of patients with ischemic stroke.
Collapse
Affiliation(s)
- Zhiping Ren
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Lin Chen
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yimeng Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xinbing Wei
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Shenglan Zeng
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yi Zheng
- State Key Laboratory of Microbial Technology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; and.,Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Chengjiang Gao
- State Key Laboratory of Microbial Technology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; .,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; and.,Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Huiqing Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China;
| |
Collapse
|
56
|
Bao C, Yang Y, Zeng C, Huang H, Ye H. Identifying neuropeptide GPCRs in the mud crab, Scylla paramamosain, by combinatorial bioinformatics analysis. Gen Comp Endocrinol 2018; 269:122-130. [PMID: 30189191 DOI: 10.1016/j.ygcen.2018.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 08/23/2018] [Accepted: 09/02/2018] [Indexed: 11/24/2022]
Abstract
Neuropeptides, ubiquitous signaling molecules, commonly achieve their signaling function via interaction with cell membrane-spanning G-protein coupled receptors (GPCRs). In recent years, in the midst of the rapid development of next-generation sequencing technology, the amount of available information on encoded neuropeptides and their GPCRs sequences have increased dramatically. The repertoire of neuropeptides has been determined in many crustaceans, including the commercially important mud crab, Scylla paramamosain; however, determination of GPCRs is known to be more difficult and usually requires in vitro binding tests. In this study, we adopted a combinatorial bioinformatics analysis to identify S. paramamosain neuropeptide GPCRs. A total of 65 assembled GPCR sequences were collected from the transcriptome database. Subsequently these GPCRs were identified by comparison to known neuropeptide GPCRs based on the sequence-similarity-based clustering and phylogenetic analysis, which showed that many of them are closely related to insect GPCR families. Of these GPCRs, most of them were detected in various tissues of the mud crab and some of them showed differential expression by gender, suggesting they are involved in different physiological processes, such as sex differentiation. By employing ligand-receptor binding tests, we demonstrated that the predicted crustacean cardioactive peptide (CCAP) receptor was activated by CCAP peptide in a dose-dependent manner. This is the first CCAP receptor that has been functionally defined in crustaceans. In summary, the present study shortlists candidate neuropeptide GPCRs for ligand-receptor binding tests, and provides information for subsequent future research on the neuropeptide/GPCR signaling pathway in S. paramamosain.
Collapse
Affiliation(s)
- Chenchang Bao
- College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Yanan Yang
- College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Chaoshu Zeng
- College of Science & Engineering, James Cook University, Townsville, Queensland 4811, Australia
| | - Huiyang Huang
- College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Haihui Ye
- College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen 361102, Fujian Province, China.
| |
Collapse
|
57
|
da Fonseca NJ, Afonso MQL, de Oliveira LC, Bleicher L. A new method bridging graph theory and residue co-evolutionary networks for specificity determinant positions detection. Bioinformatics 2018; 35:1478-1485. [DOI: 10.1093/bioinformatics/bty846] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 09/11/2018] [Accepted: 10/04/2018] [Indexed: 12/22/2022] Open
Affiliation(s)
- Néli José da Fonseca
- Departmento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Pampulha, Belo Horizonte – MG, Brazil
| | - Marcelo Querino Lima Afonso
- Departmento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Pampulha, Belo Horizonte – MG, Brazil
| | - Lucas Carrijo de Oliveira
- Departmento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Pampulha, Belo Horizonte – MG, Brazil
| | - Lucas Bleicher
- Departmento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Pampulha, Belo Horizonte – MG, Brazil
| |
Collapse
|
58
|
Hoque M, Ali S, Hoda M. Current status of G-protein coupled receptors as potential targets against type 2 diabetes mellitus. Int J Biol Macromol 2018; 118:2237-2244. [DOI: 10.1016/j.ijbiomac.2018.07.091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 06/09/2018] [Accepted: 07/14/2018] [Indexed: 12/15/2022]
|
59
|
Kubota R, Nomura W, Iwasaka T, Ojima K, Kiyonaka S, Hamachi I. Chemogenetic Approach Using Ni(II) Complex-Agonist Conjugates Allows Selective Activation of Class A G-Protein-Coupled Receptors. ACS CENTRAL SCIENCE 2018; 4:1211-1221. [PMID: 30276255 PMCID: PMC6161059 DOI: 10.1021/acscentsci.8b00390] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Indexed: 05/04/2023]
Abstract
Investigating individual G-protein-coupled receptors (GPCRs) involved in various signaling cascades can unlock a myriad of invaluable physiological findings. One of the promising strategies for addressing the activity of each subtype of receptor is to design chemical turn-on switches on the target receptors. However, valid methods to selectively control class A GPCRs, the largest receptor family encoded in the human genome, remain limited. Here, we describe a novel approach to chemogenetically manipulate activity of engineered class A GPCRs carrying a His4 tag, using metal complex-agonist conjugates (MACs). This manipulation is termed coordination tethering. With the assistance of coordination bonds, MACs showed 10-100-fold lower EC50 values in the engineered receptors, compared with wild-type receptors. Such coordination tethering enabled selective activation of β2-adrenoceptors and muscarinic acetylcholine receptors, without loss of natural receptor responses, in living mammalian cells, including primary cultured astrocytes. Our generalized, modular chemogenetic approach should facilitate more precise control and deeper understanding of individual GPCR signaling pathways in living systems.
Collapse
Affiliation(s)
- Ryou Kubota
- Department
of Synthetic Chemistry and Biological Chemistry, Graduate School of
Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Wataru Nomura
- Department
of Synthetic Chemistry and Biological Chemistry, Graduate School of
Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Takuma Iwasaka
- Department
of Synthetic Chemistry and Biological Chemistry, Graduate School of
Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Kento Ojima
- Department
of Synthetic Chemistry and Biological Chemistry, Graduate School of
Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Shigeki Kiyonaka
- Department
of Synthetic Chemistry and Biological Chemistry, Graduate School of
Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Itaru Hamachi
- Department
of Synthetic Chemistry and Biological Chemistry, Graduate School of
Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- Core
Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 5 Sanbancho, Chiyoda-ku, Tokyo 102-0075, Japan
- E-mail:
| |
Collapse
|
60
|
Venthur H, Zhou JJ. Odorant Receptors and Odorant-Binding Proteins as Insect Pest Control Targets: A Comparative Analysis. Front Physiol 2018; 9:1163. [PMID: 30197600 PMCID: PMC6117247 DOI: 10.3389/fphys.2018.01163] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 08/03/2018] [Indexed: 01/09/2023] Open
Abstract
Recently, two alternative targets in insect periphery nerve system have been explored for environmentally-friendly approaches in insect pest management, namely odorant-binding proteins (OBPs) and odorant receptors (ORs). Located in insect antennae, OBPs are thought to be involved in the transport of odorants to ORs for the specific signal transduction of behaviorally active odorants. There is rich information on OBP binding affinity and molecular docking to bioactive compounds as well as ample 3D crystal structures due to feasible production of recombinant proteins. Although these provide excellent opportunities for them to be considered as pest control targets and a tool to design pest control agents, the debates on their binding specificity represent an obstacle. On the other hand, ORs have recently been functionally characterized with increasing evidence for their specificity, sensitivity and functional roles in pest behaviors. However, a major barrier to use ORs for semiochemical discovery is the lack of 3D crystal structures. Thus, OBPs and ORs have not been analyzed comparatively together so far for their feasibility as pest control targets. Here, we summarize the state of OBPs and ORs research in terms of its application in insect pest management. We discuss the suitability of both proteins as pest control targets and their selection toward the discovery of new potent semiochemicals. We argue that both proteins represent promising targets for pest control and can be used to identify new super-ligands likely present in nature and with reduced risk of resistance development than insect pesticides currently used in agriculture. We discuss that with the massive identification of OBPs through RNA-seq and improved binding affinity measurements, these proteins could be reconsidered as suitable targets for semiochemical discovery.
Collapse
Affiliation(s)
- Herbert Venthur
- Laboratorio de Química Ecológica, Departamento de Ciencias Químicas y Recursos Naturales, Universidad de La Frontera, Temuco, Chile.,Center of Excellence in Biotechnology Research Applied to the Environment (CIBAMA), Universidad de La Frontera, Temuco, Chile
| | - Jing-Jiang Zhou
- Department of Biological Chemistry and Crop Protection, Rothamsted Research, Harpenden, United Kingdom.,Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
| |
Collapse
|
61
|
Optical functionalization of human Class A orphan G-protein-coupled receptors. Nat Commun 2018; 9:1950. [PMID: 29769519 PMCID: PMC5956105 DOI: 10.1038/s41467-018-04342-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/20/2018] [Indexed: 12/21/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) form the largest receptor family, relay environmental stimuli to changes in cell behavior and represent prime drug targets. Many GPCRs are classified as orphan receptors because of the limited knowledge on their ligands and coupling to cellular signaling machineries. Here, we engineer a library of 63 chimeric receptors that contain the signaling domains of human orphan and understudied GPCRs functionally linked to the light-sensing domain of rhodopsin. Upon stimulation with visible light, we identify activation of canonical cell signaling pathways, including cAMP-, Ca2+-, MAPK/ERK-, and Rho-dependent pathways, downstream of the engineered receptors. For the human pseudogene GPR33, we resurrect a signaling function that supports its hypothesized role as a pathogen entry site. These results demonstrate that substituting unknown chemical activators with a light switch can reveal information about protein function and provide an optically controlled protein library for exploring the physiology and therapeutic potential of understudied GPCRs. G-protein coupled receptors (GPCRs) represent the largest receptor family and are prime drug targets, but many orphan GPCRs are poorly characterized. Here authors engineer human orphan GPCRs to be activated by light which allows studying the receptors ligand identity and downstream signaling.
Collapse
|
62
|
Abstract
Neuropeptides are the largest class of intercellular signaling molecules, contributing to a wide variety of physiological processes. Neuropeptide receptors are therapeutic targets for a broad range of drugs, including medications to treat pain, addiction, sleep disorders, and nausea. In addition to >100 peptides with known functions, many peptides have been identified in mammalian brain for which the cognate receptors have not been identified. Similarly, dozens of "orphan" G protein-coupled receptors have been identified in the mammalian genome. While it would seem straightforward to match the orphan peptides and receptors, this is not always easily accomplished. In this review we focus on peptides named PEN and big LEN, which are among the most abundant neuropeptides in mouse brain, and their recently identified receptors: GPR83 and GPR171. These receptors are co-expressed in some brain regions and are able to interact. Because PEN and big LEN are produced from the same precursor protein and co-secreted, the interaction of GPR83 and GPR171 is physiologically relevant. In addition to interactions of these two peptides/receptors, PEN and LEN are co-localized with neuropeptide Y and Agouti-related peptide in neurons that regulate feeding. In this review, using these peptide receptors as an example, we highlight the multiple modes of regulation of receptors and present the emerging view that neuropeptides function combinatorially to generate a network of signaling messages. The complexity of neuropeptides, receptors, and their signaling pathways is important to consider both in the initial deorphanization of peptides and receptors, and in the subsequent development of therapeutic applications.
Collapse
Affiliation(s)
- Lloyd D Fricker
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, United States
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
63
|
Schöneberg T, Meister J, Knierim AB, Schulz A. The G protein-coupled receptor GPR34 - The past 20 years of a grownup. Pharmacol Ther 2018; 189:71-88. [PMID: 29684466 DOI: 10.1016/j.pharmthera.2018.04.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Research on GPR34, which was discovered in 1999 as an orphan G protein-coupled receptor of the rhodopsin-like class, disclosed its physiologic relevance only piece by piece. Being present in all recent vertebrate genomes analyzed so far it seems to improve the fitness of species although it is not essential for life and reproduction as GPR34-deficient mice demonstrate. However, closer inspection of macrophages and microglia, where it is mainly expressed, revealed its relevance in immune cell function. Recent data clearly demonstrate that GPR34 function is required to arrest microglia in the M0 homeostatic non-phagocytic phenotype. Herein, we summarize the current knowledge on its evolution, genomic and structural organization, physiology, pharmacology and relevance in human diseases including neurodegenerative diseases and cancer, which accumulated over the last 20 years.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany.
| | - Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Alexander Bernd Knierim
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; Leipzig University Medical Center, IFB AdiposityDiseases, 04103 Leipzig, Germany
| | - Angela Schulz
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
64
|
The G protein-coupled receptors deorphanization landscape. Biochem Pharmacol 2018; 153:62-74. [PMID: 29454621 DOI: 10.1016/j.bcp.2018.02.016] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) are usually highlighted as being both the largest family of membrane proteins and the most productive source of drug targets. However, most of the GPCRs are understudied and hence cannot be used immediately for innovative therapeutic strategies. Besides, there are still around 100 orphan receptors, with no described endogenous ligand and no clearly defined function. The race to discover new ligands for these elusive receptors seems to be less intense than before. Here, we present an update of the various strategies employed to assign a function to these receptors and to discover new ligands. We focus on the recent advances in the identification of endogenous ligands with a detailed description of newly deorphanized receptors. Replication being a key parameter in these endeavors, we also discuss the latest controversies about problematic ligand-receptor pairings. In this context, we propose several recommendations in order to strengthen the reporting of new ligand-receptor pairs.
Collapse
|
65
|
Sutton LP, Orlandi C, Song C, Oh WC, Muntean BS, Xie K, Filippini A, Xie X, Satterfield R, Yaeger JDW, Renner KJ, Young SM, Xu B, Kwon H, Martemyanov KA. Orphan receptor GPR158 controls stress-induced depression. eLife 2018; 7:33273. [PMID: 29419376 PMCID: PMC5823542 DOI: 10.7554/elife.33273] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/06/2018] [Indexed: 01/23/2023] Open
Abstract
Stress can be a motivational force for decisive action and adapting to novel environment; whereas, exposure to chronic stress contributes to the development of depression and anxiety. However, the molecular mechanisms underlying stress-responsive behaviors are not fully understood. Here, we identified the orphan receptor GPR158 as a novel regulator operating in the prefrontal cortex (PFC) that links chronic stress to depression. GPR158 is highly upregulated in the PFC of human subjects with major depressive disorder. Exposure of mice to chronic stress also increased GPR158 protein levels in the PFC in a glucocorticoid-dependent manner. Viral overexpression of GPR158 in the PFC induced depressive-like behaviors. In contrast GPR158 ablation, led to a prominent antidepressant-like phenotype and stress resiliency. We found that GPR158 exerts its effects via modulating synaptic strength altering AMPA receptor activity. Taken together, our findings identify a new player in mood regulation and introduce a pharmacological target for managing depression.
Collapse
Affiliation(s)
- Laurie P Sutton
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Chenghui Song
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Won Chan Oh
- Max Planck Florida Institute for Neuroscience, Jupiter, United States
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Keqiang Xie
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Alice Filippini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Xiangyang Xie
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | | | - Jazmine D W Yaeger
- Center for Brain and Behavior Research, University of South Dakota, Vermillion, United States.,Department of Biology, University of South Dakota, Vermillion, United States
| | - Kenneth J Renner
- Center for Brain and Behavior Research, University of South Dakota, Vermillion, United States.,Department of Biology, University of South Dakota, Vermillion, United States
| | - Samuel M Young
- Max Planck Florida Institute for Neuroscience, Jupiter, United States.,Department of Anatomy and Cell Biology, University of Iowa, Iowa, United States.,Aging Mind and Brain Initiative, University of Iowa, Iowa, United States.,Department of Otolaryngology, Carver College of Medicine, University of Iowa, Iowa, United States
| | - Baoji Xu
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Hyungbae Kwon
- Max Planck Florida Institute for Neuroscience, Jupiter, United States.,Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| |
Collapse
|
66
|
Jékely G, Melzer S, Beets I, Kadow ICG, Koene J, Haddad S, Holden-Dye L. The long and the short of it - a perspective on peptidergic regulation of circuits and behaviour. J Exp Biol 2018; 221:jeb166710. [PMID: 29439060 DOI: 10.1242/jeb.166710] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Neuropeptides are the most diverse class of chemical modulators in nervous systems. They contribute to extensive modulation of circuit activity and have profound influences on animal physiology. Studies on invertebrate model organisms, including the fruit fly Drosophila melanogaster and the nematode Caenorhabditis elegans, have enabled the genetic manipulation of peptidergic signalling, contributing to an understanding of how neuropeptides pattern the output of neural circuits to underpin behavioural adaptation. Electrophysiological and pharmacological analyses of well-defined microcircuits, such as the crustacean stomatogastric ganglion, have provided detailed insights into neuropeptide functions at a cellular and circuit level. These approaches can be increasingly applied in the mammalian brain by focusing on circuits with a defined and identifiable sub-population of neurons. Functional analyses of neuropeptide systems have been underpinned by systematic studies to map peptidergic networks. Here, we review the general principles and mechanistic insights that have emerged from these studies. We also highlight some of the challenges that remain for furthering our understanding of the functional relevance of peptidergic modulation.
Collapse
Affiliation(s)
- Gáspár Jékely
- Living Systems Institute, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - Sarah Melzer
- Howard Hughes Medical Institute, Department of Neurobiology, 200 Longwood Avenue, Boston, MA 02115, USA
| | - Isabel Beets
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Ilona C Grunwald Kadow
- Technical University of Munich, TUM School of Life Sciences, ZIEL - Institute for Food and Health, 85354 Freising, Germany
| | - Joris Koene
- Vrije Universiteit - Ecological Science, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Sara Haddad
- Volen Center for Complex Systems, Brandeis University, Mailstop 013, 415 South Street, Waltham, MA 02454, USA
| | - Lindy Holden-Dye
- Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, UK
| |
Collapse
|
67
|
Oishi A, Cecon E, Jockers R. Melatonin Receptor Signaling: Impact of Receptor Oligomerization on Receptor Function. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:59-77. [DOI: 10.1016/bs.ircmb.2018.02.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
68
|
De Francesco EM, Sotgia F, Clarke RB, Lisanti MP, Maggiolini M. G Protein-Coupled Receptors at the Crossroad between Physiologic and Pathologic Angiogenesis: Old Paradigms and Emerging Concepts. Int J Mol Sci 2017; 18:ijms18122713. [PMID: 29240722 PMCID: PMC5751314 DOI: 10.3390/ijms18122713] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/11/2017] [Accepted: 12/11/2017] [Indexed: 12/14/2022] Open
Abstract
G protein-coupled receptors (GPCRs) have been implicated in transmitting signals across the extra- and intra-cellular compartments, thus allowing environmental stimuli to elicit critical biological responses. As GPCRs can be activated by an extensive range of factors including hormones, neurotransmitters, phospholipids and other stimuli, their involvement in a plethora of physiological functions is not surprising. Aberrant GPCR signaling has been regarded as a major contributor to diverse pathologic conditions, such as inflammatory, cardiovascular and neoplastic diseases. In this regard, solid tumors have been demonstrated to activate an angiogenic program that relies on GPCR action to support cancer growth and metastatic dissemination. Therefore, the manipulation of aberrant GPCR signaling could represent a promising target in anticancer therapy. Here, we highlight the GPCR-mediated angiogenic function focusing on the molecular mechanisms and transduction effectors driving the patho-physiological vasculogenesis. Specifically, we describe evidence for the role of heptahelic receptors and associated G proteins in promoting angiogenic responses in pathologic conditions, especially tumor angiogenesis and progression. Likewise, we discuss opportunities to manipulate aberrant GPCR-mediated angiogenic signaling for therapeutic benefit using innovative GPCR-targeted and patient-tailored pharmacological strategies.
Collapse
Affiliation(s)
- Ernestina M De Francesco
- Department of Pharmacy, Health and Nutrition Sciences, University of Calabria via Savinio, 87036 Rende, Italy.
- Breast Cancer Now Research Unit, Division of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester M20 4GJ, UK.
| | - Federica Sotgia
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre, University of Salford, Greater Manchester M5 4WT, UK.
| | - Robert B Clarke
- Breast Cancer Now Research Unit, Division of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester M20 4GJ, UK.
| | - Michael P Lisanti
- Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre, University of Salford, Greater Manchester M5 4WT, UK.
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutrition Sciences, University of Calabria via Savinio, 87036 Rende, Italy.
| |
Collapse
|
69
|
Kozielewicz P, Alomar H, Yusof S, Grafton G, Cooper AJ, Curnow SJ, Ironside JW, Pall H, Barnes NM. N-glycosylation and expression in human tissues of the orphan GPR61 receptor. FEBS Open Bio 2017; 7:1982-1993. [PMID: 29226084 PMCID: PMC5715243 DOI: 10.1002/2211-5463.12339] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/19/2017] [Accepted: 10/23/2017] [Indexed: 12/21/2022] Open
Abstract
A number of members of the G protein-coupled receptor class of cell surface receptors are 'orphans' with no known endogenous ligand. One of these orphan receptors is GPR61; there are little data about its expression in human cells and tissues. In this study, we investigated the post-translational modification of GPR61 by N-glycosylation at an identified consensus N-glycosylation site (N12) and the impact of this modification upon the subcellular expression of the protein. The N-glycosylation inhibitor tunicamycin reduced the apparent molecular weight of immunoreactivity associated with myc-tagged GPR61 by 1-2 kDa, which was comparable to the evident molecular weight of the myc-tagged N12S GPR61 mutant with disrupted consensus N-glycosylation site. Analysis of GPR61 expression demonstrated that tunicamycin treatment reduced considerably heterologous expression of GPR61 in the cell membrane despite the N12S GPR61 mutant being readily expressed at the cell surface. These results demonstrate that GPR61 is subject to N-glycosylation but suggest this is not a prerequisite for cell surface expression, although N-glycosylation of other proteins may be important for cell membrane expression of GPR61. Expression of GPR61 protein was demonstrated at the cellular level in human hippocampus and human peripheral blood mononuclear cells. In the latter, there was a significantly higher expression of GPR61 in the Th17 cell subset in comparison with resting CD4+ cells, which may point toward a potential role for the GPR61 receptor in autoimmune diseases. This is the first report that GPR61 protein is subject to post-translational modification and is expressed in immune cell subsets and the hippocampus. These findings will help guide studies to investigate the function of GPR61.
Collapse
Affiliation(s)
- Paweł Kozielewicz
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK.,Present address: Department of Physiology and Pharmacology Karolinska Institutet Nanna Svartz väg 217 177 Stockholm Sweden
| | - Hatun Alomar
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK.,Present address: Pharmacology and Toxicology Department College of Pharmacy King Saud University Riyadh 12372 Saudi Arabia
| | - Syaratul Yusof
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK.,Present address: Faculty of Pharmacy Universiti Kebangsaan Malaysia 50300 Kuala Lumpur Malaysia
| | - Gillian Grafton
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK
| | - Alison J Cooper
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK
| | - S John Curnow
- Institute of Inflammation and Ageing College of Medical and Dental Sciences University of Birmingham UK
| | - James W Ironside
- National CJD Research and Surveillance Unit Centre for Clinical Brain Sciences University of Edinburgh UK
| | - Hardev Pall
- Neurology Department Queen Elizabeth Hospital Birmingham UK
| | - Nicholas M Barnes
- Institute of Clinical Sciences College of Medical and Dental Sciences University of Birmingham UK
| |
Collapse
|
70
|
Fricker LD. Carboxypeptidase E and the Identification of Novel Neuropeptides as Potential Therapeutic Targets. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:85-102. [PMID: 29413529 DOI: 10.1016/bs.apha.2017.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Peptides and small molecules that bind to peptide receptors are important classes of drugs that are used for a wide variety of different applications. The search for novel neuropeptides traditionally involved a time-consuming approach to purify each peptide to homogeneity and determine its amino acid sequence. The discovery in the 1980s of enkephalin convertase/carboxypeptidase E (CPE), and the observation that this enzyme was involved in the production of nearly every known neuropeptide led to the idea for a one-step affinity purification of CPE substrates. This approach was successfully used to isolate hundreds of known neuropeptides in mouse brain, as well as over a dozen novel peptides. Some of the novel peptides found using this approach are among the most abundant peptides present in brain, but had not been previously identified by traditional approaches. Recently, receptors for two of the novel peptides have been identified, confirming their role as neuropeptides that function in cell-cell signaling. Small molecules that bind to one of these receptors have been developed and found to significantly reduce food intake and anxiety-like behavior in an animal model. This review describes the entire project, from discovery of CPE to the novel peptides and their receptors.
Collapse
Affiliation(s)
- Lloyd D Fricker
- Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
71
|
Milligan G. G protein-coupled receptors not currently in the spotlight: free fatty acid receptor 2 and GPR35. Br J Pharmacol 2017; 175:2543-2553. [PMID: 28940377 PMCID: PMC6003633 DOI: 10.1111/bph.14042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/17/2017] [Accepted: 08/30/2017] [Indexed: 01/05/2023] Open
Abstract
It is widely appreciated that G protein‐coupled receptors have been the most successfully exploited class of targets for the development of small molecule medicines. Despite this, to date, less than 15% of the non‐olfactory G protein‐coupled receptors in the human genome are the targets of a clinically used medicine. In many cases, this is likely to reflect a lack of understanding of the basic underpinning biology of many G protein‐coupled receptors that are not currently in the spotlight, as well as a paucity of pharmacological tool compounds and appropriate animal models to test in vivo function of such G protein‐coupled receptors in both normal physiology and in the context of disease. ‘Open Innovation’ arrangements, in which pharmaceutical companies and public–private partnerships provide wider access to tool compounds identified from ligand screening programmes, alongside enhanced medicinal chemistry support to convert such screening ‘hits’ into useful ‘tool’ compounds will provide important routes to improved understanding. However, in parallel, novel approaches to define and fully appreciate the selectivity and mode of action of such tool compounds, as well as better understanding of potential species orthologue variability in the pharmacology and/or signalling profile of a wide range of currently poorly understood and understudied G protein‐coupled receptors, will be vital to fully exploit the therapeutic potential of this large target class. I consider these themes using as exemplars two G protein‐coupled receptors, free fatty acid receptor 2 and GPR35.
Collapse
Affiliation(s)
- Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
72
|
Bombesin-like receptor 3 (Brs3) expression in glutamatergic, but not GABAergic, neurons is required for regulation of energy metabolism. Mol Metab 2017; 6:1540-1550. [PMID: 29107299 PMCID: PMC5681273 DOI: 10.1016/j.molmet.2017.08.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 02/03/2023] Open
Abstract
Objective Bombesin-like receptor 3 (BRS-3) is an orphan G protein-coupled receptor. Brs3 null mice have reduced resting metabolic rate and body temperature, increased food intake, and obesity. Here we study the role of Brs3 in different neuron types. Methods Mice able to undergo Cre recombinase-dependent inactivation or re-expression of Brs3 were generated, respectively Brs3fl/y and Brs3loxTB/y. We then studied four groups of mice with Brs3 selectively inactivated or re-expressed in cells expressing Vglut2-Cre or Vgat-Cre. Results Deletion of Brs3 in glutamatergic neurons expressing Vglut2 reproduced the global null phenotype for regulation of food intake, metabolic rate, body temperature, adiposity, and insulin resistance. These mice also no longer responded to a BRS-3 agonist, MK-5046. In contrast, deletion of Brs3 in GABAergic neurons produced no detectable phenotype. Conversely, the wild type phenotype was restored by selective re-expression of Brs3 in glutamatergic neurons, with no normalization achieved by re-expressing Brs3 in GABAergic neurons. Conclusions Brs3 expression in glutamatergic neurons is both necessary and sufficient for full Brs3 function in energy metabolism. In these experiments, no function was identified for Brs3 in GABAergic neurons. The data suggest that the anti-obesity pharmacologic actions of BRS-3 agonists occur via agonism of receptors on glutamatergic neurons. Brs3 in glutamatergic neurons regulates food intake, metabolic rate, and body weight. Brs3 in glutamatergic neurons is both necessary and sufficient for these functions. No phenotypes were identified by Brs3 loss or re-expression in GABAergic neurons. BRS-3 agonists likely act on glutamatergic neurons for their anti-obesity effects.
Collapse
|
73
|
Oishi A, Karamitri A, Gerbier R, Lahuna O, Ahmad R, Jockers R. Orphan GPR61, GPR62 and GPR135 receptors and the melatonin MT 2 receptor reciprocally modulate their signaling functions. Sci Rep 2017; 7:8990. [PMID: 28827538 PMCID: PMC5566548 DOI: 10.1038/s41598-017-08996-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 07/14/2017] [Indexed: 01/14/2023] Open
Abstract
Understanding the function of orphan G protein-coupled receptors (GPCRs), whose cognate ligand is unknown, is of major importance as GPCRs are privileged drug targets for many diseases. Recent phylogenetic studies classified three orphan receptors, GPR61, GPR62 and GPR135 among the melatonin receptor subfamily, but their capacity to bind melatonin and their biochemical functions are not well characterized yet. We show here that GPR61, GPR62 and GPR135 do not bind [3H]-melatonin nor 2-[125I]iodomelatonin and do not respond to melatonin in several signaling assays. In contrast, the three receptors show extensive spontaneous ligand-independent activities on the cAMP, inositol phosphate and ß-arrestin pathways with distinct pathway-specific profiles. Spontaneous ß-arrestin recruitment internalizes all three GPRs in the endosomal compartment. Co-expression of the melatonin binding MT2 receptor with GPR61, GPR62 or GPR135 has several consequences such as (i) the formation of receptor heteromers, (ii) the inhibition of melatonin-induced ß-arrestin2 recruitment to MT2 and (iii) the decrease of elevated cAMP levels upon melatonin stimulation in cells expressing spontaneously active GPR61 and GPR62. Collectively, these data show that GPR61, GPR62 and GPR135 are unable to bind melatonin, but show a reciprocal regulatory interaction with MT2 receptors.
Collapse
Affiliation(s)
- Atsuro Oishi
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR, 8104, Paris, France.,University Paris Descartes, Paris, France
| | - Angeliki Karamitri
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR, 8104, Paris, France.,University Paris Descartes, Paris, France
| | - Romain Gerbier
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR, 8104, Paris, France.,University Paris Descartes, Paris, France
| | - Olivier Lahuna
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR, 8104, Paris, France.,University Paris Descartes, Paris, France
| | - Raise Ahmad
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR, 8104, Paris, France.,University Paris Descartes, Paris, France
| | - Ralf Jockers
- Inserm, U1016, Institut Cochin, Paris, France. .,CNRS UMR, 8104, Paris, France. .,University Paris Descartes, Paris, France.
| |
Collapse
|
74
|
Gacasan SB, Baker DL, Parrill AL. G protein-coupled receptors: the evolution of structural insight. AIMS BIOPHYSICS 2017; 4:491-527. [PMID: 29951585 DOI: 10.3934/biophy.2017.3.491] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
G protein-coupled receptors (GPCR) comprise a diverse superfamily of over 800 proteins that have gained relevance as biological targets for pharmaceutical drug design. Although these receptors have been investigated for decades, three-dimensional structures of GPCR have only recently become available. In this review, we focus on the technological advancements that have facilitated efforts to gain insights into GPCR structure. Progress in these efforts began with the initial crystal structure determination of rhodopsin (PDB: 1F88) in 2000 and has continued to the most recently published structure of the A1AR (PDB: 5UEN) in 2017. Numerous experimental developments over the past two decades have opened the door for widespread GPCR structural characterization. These efforts have resulted in the determination of three-dimensional structures for over 40 individual GPCR family members. Herein we present a comprehensive list and comparative analysis of over 180 individual GPCR structures. This includes a summary of different GPCR functional states crystallized with agonists, dual agonists, partial agonists, inverse agonists, antagonists, and allosteric modulators.
Collapse
Affiliation(s)
- Samantha B Gacasan
- Department of Chemistry, University of Memphis, 3744 Walker Ave, Memphis, TN 38152, USA
| | - Daniel L Baker
- Department of Chemistry, University of Memphis, 3744 Walker Ave, Memphis, TN 38152, USA
| | - Abby L Parrill
- Department of Chemistry, University of Memphis, 3744 Walker Ave, Memphis, TN 38152, USA
| |
Collapse
|
75
|
Identification of two novel chicken GPR133 variants and their expression in different tissues. Funct Integr Genomics 2017; 17:687-696. [DOI: 10.1007/s10142-017-0564-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 05/08/2017] [Accepted: 05/18/2017] [Indexed: 12/13/2022]
|
76
|
Irving A, Abdulrazzaq G, Chan SLF, Penman J, Harvey J, Alexander SPH. Cannabinoid Receptor-Related Orphan G Protein-Coupled Receptors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 80:223-247. [PMID: 28826536 DOI: 10.1016/bs.apha.2017.04.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Of the druggable group of G protein-coupled receptors in the human genome, a number remain which have yet to be paired with an endogenous ligand-orphan GPCRs. Among these 100 or so entities, 3 have been linked to the cannabinoid system. GPR18, GPR55, and GPR119 exhibit limited sequence homology with the established CB1 and CB2 cannabinoid receptors. However, the pharmacology of these orphan receptors displays overlap with CB1 and CB2 receptors, particularly for GPR18 and GPR55. The linking of GPR119 to the cannabinoid receptors is less convincing and emanates from structural similarities of endogenous ligands active at these GPCRs, but which do not cross-react. This review describes the evidence for describing these orphan GPCRs as cannabinoid receptor-like receptors.
Collapse
Affiliation(s)
- Andrew Irving
- The Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.
| | - Ghayth Abdulrazzaq
- Life Sciences, University of Nottingham Medical School, Nottingham, United Kingdom
| | - Sue L F Chan
- Life Sciences, University of Nottingham Medical School, Nottingham, United Kingdom
| | - June Penman
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | - Jenni Harvey
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | | |
Collapse
|
77
|
Dupuis N, Laschet C, Franssen D, Szpakowska M, Gilissen J, Geubelle P, Soni A, Parent AS, Pirotte B, Chevigné A, Twizere JC, Hanson J. Activation of the Orphan G Protein-Coupled Receptor GPR27 by Surrogate Ligands Promotes β-Arrestin 2 Recruitment. Mol Pharmacol 2017; 91:595-608. [PMID: 28314853 DOI: 10.1124/mol.116.107714] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/16/2017] [Indexed: 01/14/2023] Open
Abstract
G protein-coupled receptors are the most important drug targets for human diseases. An important number of them remain devoid of confirmed ligands. GPR27 is one of these orphan receptors, characterized by a high level of conservation among vertebrates and a predominant expression in the central nervous system. In addition, it has recently been linked to insulin secretion. However, the absence of endogenous or surrogate ligands for GPR27 complicates the examination of its biologic function. Our aim was to validate GPR27 signaling pathways, and therefore we sought to screen a diversity-oriented synthesis library to identify GPR27-specific surrogate agonists. To select an optimal screening assay, we investigated GPR27 ligand-independent activity. Both in G protein-mediated pathways and in β-arrestin 2 recruitment, no ligand-independent activity could be measured. However, we observed a recruitment of β-arrestin 2 to a GPR27V2 chimera in the presence of membrane-anchored G protein-coupled receptor kinase-2. Therefore, we optimized a firefly luciferase complementation assay to screen against this chimeric receptor. We identified two compounds [N-[4-(anilinocarbonyl)phenyl]-2,4-dichlorobenzamide (ChemBridge, San Diego, CA; ID5128535) and 2,4-dichloro-N-{4-[(1,3-thiazol-2-ylamino)sulfonyl]phenyl}benzamide (ChemBridge ID5217941)] sharing a N-phenyl-2,4-dichlorobenzamide scaffold, which were selective for GPR27 over its closely related family members GPR85 and GPR173. The specificity of the activity was confirmed with a NanoLuc Binary Technology β-arrestin 2 assay, imaging of green fluorescent protein-tagged β-arrestin 2, and PathHunter β-arrestin 2 assay. Interestingly, no G protein activation was detected upon activation of GPR27 by these compounds. Our study provides the first selective surrogate agonists for the orphan GPR27.
Collapse
Affiliation(s)
- Nadine Dupuis
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| | - Céline Laschet
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| | - Delphine Franssen
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| | - Martyna Szpakowska
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| | - Julie Gilissen
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| | - Pierre Geubelle
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| | - Arvind Soni
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| | - Anne-Simone Parent
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| | - Bernard Pirotte
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| | - Andy Chevigné
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| | - Jean-Claude Twizere
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| | - Julien Hanson
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases (N.D., C.L., J.G., P.G., A.S., J.H.), Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (N.D., B.P., J.H.), Neuroendocrinology Unit, GIGA-Neurosciences (D.F., A.-S.P.), Laboratory of Protein Signaling and Interactions, GIGA-Molecular Biology of Diseases (J.-C.T.), University of Liège, Liège, Belgium; and Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg (M.S., A.C.)
| |
Collapse
|
78
|
Brüser A, Zimmermann A, Crews BC, Sliwoski G, Meiler J, König GM, Kostenis E, Lede V, Marnett LJ, Schöneberg T. Prostaglandin E 2 glyceryl ester is an endogenous agonist of the nucleotide receptor P2Y 6. Sci Rep 2017; 7:2380. [PMID: 28539604 PMCID: PMC5443783 DOI: 10.1038/s41598-017-02414-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 04/10/2017] [Indexed: 11/10/2022] Open
Abstract
Cyclooxygenase-2 catalyses the biosynthesis of prostaglandins from arachidonic acid but also the biosynthesis of prostaglandin glycerol esters (PG-Gs) from 2-arachidonoylglycerol. Previous studies identified PG-Gs as signalling molecules involved in inflammation. Thus, the glyceryl ester of prostaglandin E2, PGE2-G, mobilizes Ca2+ and activates protein kinase C and ERK, suggesting the involvement of a G protein-coupled receptor (GPCR). To identify the endogenous receptor for PGE2-G, we performed a subtractive screening approach where mRNA from PGE2-G response-positive and -negative cell lines was subjected to transcriptome-wide RNA sequencing analysis. We found several GPCRs that are only expressed in the PGE2-G responder cell lines. Using a set of functional readouts in heterologous and endogenous expression systems, we identified the UDP receptor P2Y6 as the specific target of PGE2-G. We show that PGE2-G and UDP are both agonists at P2Y6, but they activate the receptor with extremely different EC50 values of ~1 pM and ~50 nM, respectively. The identification of the PGE2-G/P2Y6 pair uncovers the signalling mode of PG-Gs as previously under-appreciated products of cyclooxygenase-2.
Collapse
Affiliation(s)
- Antje Brüser
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103, Leipzig, Germany.
| | - Anne Zimmermann
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103, Leipzig, Germany
| | - Brenda C Crews
- Department of Biochemistry, Chemistry and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, USA
| | - Gregory Sliwoski
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, 37232-8725, USA
| | - Jens Meiler
- Department of Chemistry, Center for Structural Biology, Vanderbilt University, Nashville, TN, 37232-8725, USA
| | - Gabriele M König
- Institute of Pharmaceutical Biology, University of Bonn, 53115, Bonn, Germany
| | - Evi Kostenis
- Institute of Pharmaceutical Biology, University of Bonn, 53115, Bonn, Germany
| | - Vera Lede
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103, Leipzig, Germany
| | - Lawrence J Marnett
- Department of Biochemistry, Chemistry and Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, USA
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103, Leipzig, Germany.
| |
Collapse
|
79
|
Simon K, Merten N, Schröder R, Hennen S, Preis P, Schmitt NK, Peters L, Schrage R, Vermeiren C, Gillard M, Mohr K, Gomeza J, Kostenis E. The Orphan Receptor GPR17 Is Unresponsive to Uracil Nucleotides and Cysteinyl Leukotrienes. Mol Pharmacol 2017; 91:518-532. [PMID: 28254957 DOI: 10.1124/mol.116.107904] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/01/2017] [Indexed: 12/27/2022] Open
Abstract
Pairing orphan G protein–coupled receptors (GPCRs) with their cognate endogenous ligands is expected to have a major impact on our understanding of GPCR biology. It follows that the reproducibility of orphan receptor ligand pairs should be of fundamental importance to guide meaningful investigations into the pharmacology and function of individual receptors. GPR17 is an orphan receptor characterized by some as a dualistic uracil nucleotide/cysteinyl leukotriene receptor and by others as inactive toward these stimuli altogether. Whereas regulation of central nervous system myelination by GPR17 is well established, verification of activity of its putative endogenous ligands has proven elusive so far. Herein we report that uracil nucleotides and cysteinyl leukotrienes do not activate human, mouse, or rat GPR17 in various cellular backgrounds, including primary cells, using eight distinct functional assay platforms based on labelfree pathway-unbiased biosensor technologies, as well as canonical second-messenger or biochemical assays. Appraisal of GPR17 activity can neither be accomplished with co-application of both ligand classes, nor with exogenous transfection of partner receptors (nucleotide P2Y12, cysteinyl-leukotriene CysLT1) to reconstitute the elusive pharmacology. Moreover, our study does not support the inhibition of GPR17 by the marketed antiplatelet drugs cangrelor and ticagrelor, previously suggested to antagonize GPR17. Whereas our data do not disagree with a role of GPR17 per se as an orchestrator of central nervous system functions, they challenge the utility of the proposed (ant)agonists as tools to imply direct contribution of GPR17 in complex biologic settings.
Collapse
Affiliation(s)
- Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Nicole Merten
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Ralf Schröder
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Stephanie Hennen
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Philip Preis
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Nina-Katharina Schmitt
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Lucas Peters
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Ramona Schrage
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Celine Vermeiren
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Michel Gillard
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Klaus Mohr
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Jesus Gomeza
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.)
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology (K.S., N.M., Ral.S., S.H., P.P., N.-K.S, L.P., J.G., E.K.), Research Training Group 1873 (K.S., E.K.), Pharmacology and Toxicology Section, Institute of Pharmacy (Ram.S., K.M.), University of Bonn, Bonn, Germany; UCB Pharma, CNS Research, Braine l'Alleud, Belgium (C.V., M.G.).
| |
Collapse
|
80
|
Development and validation of a high-throughput calcium mobilization assay for the orphan receptor GPR88. J Biomed Sci 2017; 24:23. [PMID: 28347302 PMCID: PMC5369193 DOI: 10.1186/s12929-017-0330-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/20/2017] [Indexed: 12/02/2022] Open
Abstract
Background GPR88 is an orphan G protein-coupled receptor highly expressed in the striatum and is implicated in basal ganglia-associated disorders. However, the receptor functions of GPR88 are still largely unknown due to the lack of potent and selective ligands appropriate for central nervous system investigation. Development of a high-throughput screening assay for GPR88 should facilitate the discovery of novel ligands to probe GPR88 functions. Methods In this paper, we describe the development of a CHO-Gαqi5-GPR88 cell-based calcium mobilization assay. The assay takes advantage of functional coupling of GPR88 with the promiscuous Gαqi5 protein and consequent mobilization of intracellular calcium, which can be measured in a 384-well format with a Fluorescent Imaging Plate Reader. Results The CHO-Gαqi5-GPR88 cell-based calcium mobilization assay was validated by the structure-activity relationship study of known GPR88 agonist (1R,2R)-2-PCCA analogues. The assay was automated and miniaturized to a 384-well format, and was deemed robust and reproducible with a Z’-factor of 0.72 and tolerated dimethyl sulfoxide to a final concentration of 2%. Screening a pilot neurotransmitter library consisting of 228 compounds yielded 10 hits, but none of the hits were confirmed as GPR88 agonists in follow-up assays. Conclusions We have developed a high-throughput calcium mobilization assay for the orphan receptor GPR88. This calcium mobilization assay can be used to identify several different types of GPR88 ligands including agonists, competitive and noncompetitive antagonists, inverse agonists, and allosteric modulators. These ligands will serve as valuable tools to probe signaling mechanisms and in vivo functions of GPR88, and could expedite development of novel therapies for diseases potentially mediated by GPR88. Electronic supplementary material The online version of this article (doi:10.1186/s12929-017-0330-3) contains supplementary material, which is available to authorized users.
Collapse
|
81
|
Freyd T, Warszycki D, Mordalski S, Bojarski AJ, Sylte I, Gabrielsen M. Ligand-guided homology modelling of the GABAB2 subunit of the GABAB receptor. PLoS One 2017; 12:e0173889. [PMID: 28323850 PMCID: PMC5360267 DOI: 10.1371/journal.pone.0173889] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/28/2017] [Indexed: 11/18/2022] Open
Abstract
γ-aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the central nervous system, and disturbances in the GABAergic system have been implicated in numerous neurological and neuropsychiatric diseases. The GABAB receptor is a heterodimeric class C G protein-coupled receptor (GPCR) consisting of GABAB1a/b and GABAB2 subunits. Two GABAB receptor ligand binding sites have been described, namely the orthosteric GABA binding site located in the extracellular GABAB1 Venus fly trap domain and the allosteric binding site found in the GABAB2 transmembrane domain. To date, the only experimentally solved three-dimensional structures of the GABAB receptor are of the Venus fly trap domain. GABAB receptor allosteric modulators, however, show great therapeutic potential, and elucidating the structure of the GABAB2 transmembrane domain may lead to development of novel drugs and increased understanding of the allosteric mechanism of action. Despite the lack of x-ray crystal structures of the GABAB2 transmembrane domain, multiple crystal structures belonging to other classes of GPCRs than class A have been released within the last years. More closely related template structures are now available for homology modelling of the GABAB receptor. Here, multiple homology models of the GABAB2 subunit of the GABAB receptor have been constructed using templates from class A, B and C GPCRs, and docking of five clusters of positive allosteric modulators and decoys has been undertaken to select models that enrich the active compounds. Using this ligand-guided approach, eight GABAB2 homology models have been chosen as possible structural representatives of the transmembrane domain of the GABAB2 subunit. To the best of our knowledge, the present study is the first to describe homology modelling of the transmembrane domain of the GABAB2 subunit and the docking of positive allosteric modulators in the receptor.
Collapse
Affiliation(s)
- Thibaud Freyd
- Department of Medical Biology, Faculty of Health Sciences, UiT - the Arctic University of Norway, Tromsø, Norway
| | - Dawid Warszycki
- Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Stefan Mordalski
- Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Andrzej J. Bojarski
- Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Ingebrigt Sylte
- Department of Medical Biology, Faculty of Health Sciences, UiT - the Arctic University of Norway, Tromsø, Norway
- * E-mail:
| | - Mari Gabrielsen
- Department of Medical Biology, Faculty of Health Sciences, UiT - the Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
82
|
Kechele DO, Blue RE, Zwarycz B, Espenschied ST, Mah AT, Siegel MB, Perou CM, Ding S, Magness ST, Lund PK, Caron KM. Orphan Gpr182 suppresses ERK-mediated intestinal proliferation during regeneration and adenoma formation. J Clin Invest 2017; 127:593-607. [PMID: 28094771 DOI: 10.1172/jci87588] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 11/22/2016] [Indexed: 12/16/2022] Open
Abstract
Orphan GPCRs provide an opportunity to identify potential pharmacological targets, yet their expression patterns and physiological functions remain challenging to elucidate. Here, we have used a genetically engineered knockin reporter mouse to map the expression pattern of the Gpr182 during development and adulthood. We observed that Gpr182 is expressed at the crypt base throughout the small intestine, where it is enriched in crypt base columnar stem cells, one of the most active stem cell populations in the body. Gpr182 knockdown had no effect on homeostatic intestinal proliferation in vivo, but led to marked increases in proliferation during intestinal regeneration following irradiation-induced injury. In the ApcMin mouse model, which forms spontaneous intestinal adenomas, reductions in Gpr182 led to more adenomas and decreased survival. Loss of Gpr182 enhanced organoid growth efficiency ex vivo in an EGF-dependent manner. Gpr182 reduction led to increased activation of ERK1/2 in basal and challenge models, demonstrating a potential role for this orphan GPCR in regulating the proliferative capacity of the intestine. Importantly, GPR182 expression was profoundly reduced in numerous human carcinomas, including colon adenocarcinoma. Together, these results implicate Gpr182 as a negative regulator of intestinal MAPK signaling-induced proliferation, particularly during regeneration and adenoma formation.
Collapse
|
83
|
Kolar GR, Grote SM, Yosten GLC. Targeting orphan G protein-coupled receptors for the treatment of diabetes and its complications: C-peptide and GPR146. J Intern Med 2017; 281:25-40. [PMID: 27306986 PMCID: PMC6092955 DOI: 10.1111/joim.12528] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
G protein-coupled receptors (GPCRs) are the most abundant receptor family encoded by the human genome and are the targets of a high percentage of drugs currently in use or in clinical trials for the treatment of diseases such as diabetes and its associated complications. Thus, orphan GPCRs, for which the ligand is unknown, represent an important untapped source of therapeutic potential for the treatment of many diseases. We have identified the previously orphan GPCR, GPR146, as the putative receptor of proinsulin C-peptide, which may prove to be an effective treatment for diabetes-associated complications. For example, we have found a potential role of C-peptide and GPR146 in regulating the function of the retinal pigment epithelium, a monolayer of cells in the retina that serves as part of the blood-retinal barrier and is disrupted in diabetic macular oedema. However, C-peptide signalling in this cell type appears to depend at least in part on extracellular glucose concentration and its interaction with insulin. In this review, we discuss the therapeutic potential of orphan GPCRs with a special focus on C-peptide and GPR146, including past and current strategies used to 'deorphanize' this diverse family of receptors, past successes and the inherent difficulties of this process.
Collapse
Affiliation(s)
- G R Kolar
- Department of Pathology, St Louis University School of Medicine, St Louis, MO, USA
| | - S M Grote
- Department of Pharmacology and Physiology, St Louis University School of Medicine, St Louis, MO, USA
| | - G L C Yosten
- Department of Pharmacology and Physiology, St Louis University School of Medicine, St Louis, MO, USA
| |
Collapse
|
84
|
Zhang Q, Zhang D, Li N, Lu Y, Yao Y, Li S, Liu Q. Zinc Nanoparticles-equipped Bioelectronic Nose Using a Microelectrode Array for Odorant Detection. ANAL SCI 2016; 32:387-93. [PMID: 27063709 DOI: 10.2116/analsci.32.387] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Bioelectronic noses, such as olfactory cell- and receptor-based biosensors, have important applications for biomimetic odorant detection in various fields. Here, a nanoparticle-equipped biosensor was designed to record extracellular potentials from olfactory receptor cells effectively. In this research, a microelectrode array (MEA) was combined with olfactory epitheliums as the olfactory biosensor to record electrophysiological signals of receptor cells in the epitheliums. Zinc nanoparticles (NanoZn) were employed along with the biosensor for different kinds of odorant measurements, which improved the electrophysiological responses to odor molecules. The NanoZn-equipped biosensor showed greater performance, such as a higher sensitivity and a larger signal-to-noise ratio, than that without the nanoparticles. Thus, this approach provided a promising method to improve the detecting performance of biosensors based on olfactory cells and receptors, which would bring broad application prospects for bioelectronic noses in environmental monitoring, food analysis, and healthcare diagnosis.
Collapse
Affiliation(s)
- Qian Zhang
- Biosensor National Special Laboratory, Department of Biomedical Engineering, Zhejiang University
| | | | | | | | | | | | | |
Collapse
|
85
|
Orphan receptor ligand discovery by pickpocketing pharmacological neighbors. Nat Chem Biol 2016; 13:235-242. [PMID: 27992882 DOI: 10.1038/nchembio.2266] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 10/11/2016] [Indexed: 12/20/2022]
Abstract
Understanding the pharmacological similarity of G protein-coupled receptors (GPCRs) is paramount for predicting ligand off-target effects, drug repurposing, and ligand discovery for orphan receptors. Phylogenetic relationships do not always correctly capture pharmacological similarity. Previous family-wide attempts to define pharmacological relationships were based on three-dimensional structures and/or known receptor-ligand pairings, both unavailable for orphan GPCRs. Here, we present GPCR-CoINPocket, a novel contact-informed neighboring pocket metric of GPCR binding-site similarity that is informed by patterns of ligand-residue interactions observed in crystallographically characterized GPCRs. GPCR-CoINPocket is applicable to receptors with unknown structure or ligands and accurately captures known pharmacological relationships between GPCRs, even those undetected by phylogeny. When applied to orphan receptor GPR37L1, GPCR-CoINPocket identified its pharmacological neighbors, and transfer of their pharmacology aided in discovery of the first surrogate ligands for this orphan with a 30% success rate. Although primarily designed for GPCRs, the method is easily transferable to other protein families.
Collapse
|
86
|
Jin C, Decker AM, Langston TL. Design, synthesis and pharmacological evaluation of 4-hydroxyphenylglycine and 4-hydroxyphenylglycinol derivatives as GPR88 agonists. Bioorg Med Chem 2016; 25:805-812. [PMID: 27956039 DOI: 10.1016/j.bmc.2016.11.058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/29/2016] [Accepted: 11/30/2016] [Indexed: 12/29/2022]
Abstract
The orphan receptor GPR88 is an attractive therapeutic target because of its implications in a number of basal ganglia-associated disorders. To date, pharmacological characterization of GPR88 has been limited due to the lack of potent and selective agonists and antagonists appropriate for CNS investigations. We have previously reported that GPR88 couples to Gαi proteins and modulates cAMP levels upon treatment with a small molecule agonist 2-PCCA. Recently, another chemotype of GPR88 agonist, represented by 2-AMPP [(2S)-N-((1R)-2-amino-1-(4-(2-methylpentyloxy)-phenyl)ethyl)-2-phenylpropanamide], has also been discovered. In this report, a new series of 2-AMPP structurally related 4-hydroxyphenylglycine and 4-hydroxyphenylglycinol derivatives have been designed and evaluated for agonist activity at GPR88. The structure-activity relationship (SAR) studies suggest that the amine group in 2-AMPP can be replaced by hydroxyl, ester and amide groups, resulting in analogues with good to moderate potency, whereas the phenyl group on the amide cap is essential for activity and has limited size, shape and electronic tolerance.
Collapse
Affiliation(s)
- Chunyang Jin
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, United States.
| | - Ann M Decker
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, United States
| | - Tiffany L Langston
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, United States
| |
Collapse
|
87
|
Holmes FE, Kerr N, Chen YJ, Vanderplank P, McArdle CA, Wynick D. Targeted disruption of the orphan receptor Gpr151 does not alter pain-related behaviour despite a strong induction in dorsal root ganglion expression in a model of neuropathic pain. Mol Cell Neurosci 2016; 78:35-40. [PMID: 27913310 PMCID: PMC5235321 DOI: 10.1016/j.mcn.2016.11.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/03/2016] [Accepted: 11/28/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Gpr151 is an orphan GPCR whose function is unknown. The restricted pattern of neuronal expression in the habenula, dorsal horn of the spinal cord and dorsal root ganglion plus homology with the galanin family of receptors imply a role in nociception. RESULTS Real-time quantitative RT-PCR demonstrated a 49.9±2.9 fold highly significant (P<0.001) increase in Gpr151 mRNA expression in the dorsal root ganglion 7days after the spared nerve injury model of neuropathic pain. Measures of acute, inflammatory and neuropathic pain behaviours were not significantly different using separate groups of Gpr151 loss-of-function mutant mice and wild-type controls. Galanin at concentrations between 100nM and 10μM did not induce calcium signalling responses in ND7/23 cells transfected with Gpr151. CONCLUSIONS Our results indicate that despite the very large upregulation in the DRG after a nerve injury model of neuropathic pain, the Gpr151 orphan receptor does not appear to be involved in the modulation of pain-related behaviours. Further, galanin is unlikely to be an endogenous ligand for Gpr151.
Collapse
Affiliation(s)
- Fiona E Holmes
- School of Physiology, Pharmacology & Neuroscience, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK; School of Clinical Sciences, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Niall Kerr
- School of Physiology, Pharmacology & Neuroscience, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK; School of Clinical Sciences, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Ying-Ju Chen
- School of Physiology, Pharmacology & Neuroscience, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK; School of Clinical Sciences, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Penny Vanderplank
- School of Physiology, Pharmacology & Neuroscience, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK; School of Clinical Sciences, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Craig A McArdle
- School of Clinical Sciences, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - David Wynick
- School of Physiology, Pharmacology & Neuroscience, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK; School of Clinical Sciences, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK.
| |
Collapse
|
88
|
Orphan GPR110 (ADGRF1) targeted by N-docosahexaenoylethanolamine in development of neurons and cognitive function. Nat Commun 2016; 7:13123. [PMID: 27759003 PMCID: PMC5075789 DOI: 10.1038/ncomms13123] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/05/2016] [Indexed: 12/27/2022] Open
Abstract
Docosahexaenoic acid (DHA, 22:6n-3) is an omega-3 fatty acid essential for proper brain development. N-docosahexaenoylethanolamine (synaptamide), an endogenous metabolite of DHA, potently promotes neurogenesis, neuritogenesis and synaptogenesis; however, the underlying molecular mechanism is not known. Here, we demonstrate orphan G-protein coupled receptor 110 (GPR110, ADGRF1) as the synaptamide receptor, mediating synaptamide-induced bioactivity in a cAMP-dependent manner. Mass spectrometry-based proteomic characterization and cellular fluorescence tracing with chemical analogues of synaptamide reveal specific binding of GPR110 to synaptamide, which triggers cAMP production with low nM potency. Disruption of this binding or GPR110 gene knockout abolishes while GPR110 overexpression enhances synaptamide-induced bioactivity. GPR110 is highly expressed in fetal brains but rapidly decreases after birth. GPR110 knockout mice show significant deficits in object recognition and spatial memory. GPR110 deorphanized as a functional synaptamide receptor provides a novel target for neurodevelopmental control and new insight into mechanisms by which DHA promotes brain development and function.
Collapse
|
89
|
Jin C, Decker AM, Harris DL, Blough BE. Effect of Substitution on the Aniline Moiety of the GPR88 Agonist 2-PCCA: Synthesis, Structure-Activity Relationships, and Molecular Modeling Studies. ACS Chem Neurosci 2016; 7:1418-1432. [PMID: 27499251 DOI: 10.1021/acschemneuro.6b00182] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
GPR88, an orphan receptor richly expressed in the striatum, is implicated in a number of basal ganglia-associated disorders. In order to elucidate the functions of GPR88, an in vivo probe appropriate for CNS investigation is required. We previously reported that 2-PCCA was able to modulate GPR88-mediated cAMP production through a Gαi-coupled pathway. Early structure-activity relationship (SAR) studies suggested that the aniline moiety of 2-PCCA is a suitable site for diverse modifications. Aimed at elucidating structural requirements in this region, we have designed and synthesized a series of analogues bearing a variety of substituents at the phenyl ring of the aniline moiety. Several compounds (e.g., 5j, 5o) showed improved or comparable potency, but have lower lipophilicity than 2-PCCA (clogP 6.19). These compounds provide the basis for further optimization to probe GPR88 in vivo functions. Computational studies confirmed the SAR trends and supported the notion that 4'-substituents on the biphenyl ring exit through a largely hydrophobic binding site to the extracellular loop.
Collapse
Affiliation(s)
- Chunyang Jin
- Center for Drug Discovery, Research Triangle Institute, Research
Triangle Park, North Carolina 27709, United States
| | - Ann M. Decker
- Center for Drug Discovery, Research Triangle Institute, Research
Triangle Park, North Carolina 27709, United States
| | - Danni L. Harris
- Center for Drug Discovery, Research Triangle Institute, Research
Triangle Park, North Carolina 27709, United States
| | - Bruce E. Blough
- Center for Drug Discovery, Research Triangle Institute, Research
Triangle Park, North Carolina 27709, United States
| |
Collapse
|
90
|
Ngo T, Kufareva I, Coleman JL, Graham RM, Abagyan R, Smith NJ. Identifying ligands at orphan GPCRs: current status using structure-based approaches. Br J Pharmacol 2016; 173:2934-51. [PMID: 26837045 PMCID: PMC5341249 DOI: 10.1111/bph.13452] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 11/18/2015] [Accepted: 01/29/2016] [Indexed: 12/26/2022] Open
Abstract
GPCRs are the most successful pharmaceutical targets in history. Nevertheless, the pharmacology of many GPCRs remains inaccessible as their endogenous or exogenous modulators have not been discovered. Tools that explore the physiological functions and pharmacological potential of these 'orphan' GPCRs, whether they are endogenous and/or surrogate ligands, are therefore of paramount importance. Rates of receptor deorphanization determined by traditional reverse pharmacology methods have slowed, indicating a need for the development of more sophisticated and efficient ligand screening approaches. Here, we discuss the use of structure-based ligand discovery approaches to identify small molecule modulators for exploring the function of orphan GPCRs. These studies have been buoyed by the growing number of GPCR crystal structures solved in the past decade, providing a broad range of template structures for homology modelling of orphans. This review discusses the methods used to establish the appropriate signalling assays to test orphan receptor activity and provides current examples of structure-based methods used to identify ligands of orphan GPCRs. Linked Articles This article is part of a themed section on Molecular Pharmacology of G Protein-Coupled Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.20/issuetoc.
Collapse
Affiliation(s)
- Tony Ngo
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, USA
| | - James Lj Coleman
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Robert M Graham
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St. Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, USA
| | - Nicola J Smith
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.
- St. Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia.
| |
Collapse
|
91
|
Mogha A, D'Rozario M, Monk KR. G Protein-Coupled Receptors in Myelinating Glia. Trends Pharmacol Sci 2016; 37:977-987. [PMID: 27670389 DOI: 10.1016/j.tips.2016.09.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 12/21/2022]
Abstract
The G protein-coupled receptor (GPCR) superfamily represents the largest class of functionally selective drug targets for disease modulation and therapy. GPCRs have been studied in great detail in central nervous system (CNS) neurons, but these important molecules have been relatively understudied in glia. In recent years, however, exciting new roles for GPCRs in glial cell biology have emerged. We focus here on the key roles of GPCRs in a specialized subset of glia, myelinating glia. We highlight recent work firmly establishing GPCRs as regulators of myelinating glial cell development and myelin repair. These advances expand our understanding of myelinating glial cell biology and underscore the utility of targeting GPCRs to promote myelin repair in human disease.
Collapse
Affiliation(s)
- Amit Mogha
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mitchell D'Rozario
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
92
|
Xiao C, Reitman ML. Bombesin-Like Receptor 3: Physiology of a Functional Orphan. Trends Endocrinol Metab 2016; 27:603-605. [PMID: 27055378 PMCID: PMC4992652 DOI: 10.1016/j.tem.2016.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/01/2016] [Accepted: 03/04/2016] [Indexed: 12/19/2022]
Abstract
Bombesin-like receptor 3 (BRS-3) is an X-linked orphan Gq-coupled receptor that regulates food intake, metabolic rate, body temperature, heart rate, blood pressure, and insulin secretion. Most BRS-3 actions occur via the brain, through mechanisms including regulating sympathetic outflow. Ablation of Brs3 causes obesity, while synthetic agonists produce weight loss.
Collapse
Affiliation(s)
- Cuiying Xiao
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Marc L Reitman
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
93
|
Koziol U, Koziol M, Preza M, Costábile A, Brehm K, Castillo E. De novo discovery of neuropeptides in the genomes of parasitic flatworms using a novel comparative approach. Int J Parasitol 2016; 46:709-21. [PMID: 27388856 DOI: 10.1016/j.ijpara.2016.05.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/18/2016] [Accepted: 05/20/2016] [Indexed: 12/11/2022]
Abstract
Neuropeptide mediated signalling is an ancient mechanism found in almost all animals and has been proposed as a promising target for the development of novel drugs against helminths. However, identification of neuropeptides from genomic data is challenging, and knowledge of the neuropeptide complement of parasitic flatworms is still fragmentary. In this work, we have developed an evolution-based strategy for the de novo discovery of neuropeptide precursors, based on the detection of localised sequence conservation between possible prohormone convertase cleavage sites. The method detected known neuropeptide precursors with good precision and specificity in the models Drosophila melanogaster and Caenorhabditis elegans. Furthermore, it identified novel putative neuropeptide precursors in nematodes, including the first description of allatotropin homologues in this phylum. Our search for neuropeptide precursors in the genomes of parasitic flatworms resulted in the description of 34 conserved neuropeptide precursor families, including 13 new ones, and of hundreds of new homologues of known neuropeptide precursor families. Most neuropeptide precursor families show a wide phylogenetic distribution among parasitic flatworms and show little similarity to neuropeptide precursors of other bilaterian animals. However, we could also find orthologs of some conserved bilaterian neuropeptides including pyrokinin, crustacean cardioactive peptide, myomodulin, neuropeptide-Y, neuropeptide KY and SIF-amide. Finally, we determined the expression patterns of seven putative neuropeptide precursor genes in the protoscolex of Echinococcus multilocularis. All genes were expressed in the nervous system with different patterns, indicating a hidden complexity of peptidergic signalling in cestodes.
Collapse
Affiliation(s)
- Uriel Koziol
- Sección Bioquímica, Facultad de Ciencias, Universidad de la República, Iguá 4225, CP11400 Montevideo, Uruguay.
| | - Miguel Koziol
- Sección Bioquímica, Facultad de Ciencias, Universidad de la República, Iguá 4225, CP11400 Montevideo, Uruguay
| | - Matías Preza
- Sección Bioquímica, Facultad de Ciencias, Universidad de la República, Iguá 4225, CP11400 Montevideo, Uruguay
| | - Alicia Costábile
- Sección Bioquímica, Facultad de Ciencias, Universidad de la República, Iguá 4225, CP11400 Montevideo, Uruguay
| | - Klaus Brehm
- University of Würzburg, Institute for Hygiene and Microbiology, Josef-Schneider-Straße 2 / Bau E1, 97080 Würzburg, Germany
| | - Estela Castillo
- Sección Bioquímica, Facultad de Ciencias, Universidad de la República, Iguá 4225, CP11400 Montevideo, Uruguay
| |
Collapse
|
94
|
Di Liberto V, Mudò G, Garozzo R, Frinchi M, Fernandez-Dueñas V, Di Iorio P, Ciccarelli R, Caciagli F, Condorelli DF, Ciruela F, Belluardo N. The Guanine-Based Purinergic System: The Tale of An Orphan Neuromodulation. Front Pharmacol 2016; 7:158. [PMID: 27378923 PMCID: PMC4911385 DOI: 10.3389/fphar.2016.00158] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/30/2016] [Indexed: 11/17/2022] Open
Abstract
Guanine-based purines (GBPs) have been recently proposed to be not only metabolic agents but also extracellular signaling molecules that regulate important functions in the central nervous system. In such way, GBPs-mediated neuroprotection, behavioral responses and neuronal plasticity have been broadly described in the literature. However, while a number of these functions (i.e., GBPs neurothophic effects) have been well-established, the molecular mechanisms behind these GBPs-dependent effects are still unknown. Furthermore, no plasma membrane receptors for GBPs have been described so far, thus GBPs are still considered orphan neuromodulators. Interestingly, an intricate and controversial functional interplay between GBPs effects and adenosine receptors activity has been recently described, thus triggering the hypothesis that GBPs mechanism of action might somehow involve adenosine receptors. Here, we review recent data describing the GBPs role in the brain. We focus on the involvement of GBPs regulating neuronal plasticity, and on the new hypothesis based on putative GBPs receptors. Overall, we expect to shed some light on the GBPs world since although these molecules might represent excellent candidates for certain neurological diseases management, the lack of putative GBPs receptors precludes any high throughput screening intent for the search of effective GBPs-based drugs.
Collapse
Affiliation(s)
- Valentina Di Liberto
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy
| | - Giuseppa Mudò
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy
| | - Roberta Garozzo
- Department of Biomedical and Biotechnological Sciences, Unit of Medical Biochemistry, University of Catania Catania, Italy
| | - Monica Frinchi
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy
| | - Víctor Fernandez-Dueñas
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, Bellvitge Biomedical Research Institute, Institute of Neurosciences, University of Barcelona Barcelona, Spain
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotecnological Sciences, University of Chieti-Pescara Chieti, Italy
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotecnological Sciences, University of Chieti-Pescara Chieti, Italy
| | - Francesco Caciagli
- Department of Medical, Oral and Biotecnological Sciences, University of Chieti-Pescara Chieti, Italy
| | - Daniele F Condorelli
- Department of Biomedical and Biotechnological Sciences, Unit of Medical Biochemistry, University of Catania Catania, Italy
| | - Francisco Ciruela
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, Bellvitge Biomedical Research Institute, Institute of Neurosciences, University of Barcelona Barcelona, Spain
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy
| |
Collapse
|
95
|
Milligan G, Shimpukade B, Ulven T, Hudson BD. Complex Pharmacology of Free Fatty Acid Receptors. Chem Rev 2016; 117:67-110. [PMID: 27299848 DOI: 10.1021/acs.chemrev.6b00056] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are historically the most successful family of drug targets. In recent times it has become clear that the pharmacology of these receptors is far more complex than previously imagined. Understanding of the pharmacological regulation of GPCRs now extends beyond simple competitive agonism or antagonism by ligands interacting with the orthosteric binding site of the receptor to incorporate concepts of allosteric agonism, allosteric modulation, signaling bias, constitutive activity, and inverse agonism. Herein, we consider how evolving concepts of GPCR pharmacology have shaped understanding of the complex pharmacology of receptors that recognize and are activated by nonesterified or "free" fatty acids (FFAs). The FFA family of receptors is a recently deorphanized set of GPCRs, the members of which are now receiving substantial interest as novel targets for the treatment of metabolic and inflammatory diseases. Further understanding of the complex pharmacology of these receptors will be critical to unlocking their ultimate therapeutic potential.
Collapse
Affiliation(s)
- Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow G12 8QQ, Scotland, United Kingdom
| | - Bharat Shimpukade
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark , Campusvej 55, DK-5230 Odense M, Denmark
| | - Trond Ulven
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark , Campusvej 55, DK-5230 Odense M, Denmark
| | - Brian D Hudson
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow G12 8QQ, Scotland, United Kingdom
| |
Collapse
|
96
|
McCallum JE, Mackenzie AE, Divorty N, Clarke C, Delles C, Milligan G, Nicklin SA. G-Protein-Coupled Receptor 35 Mediates Human Saphenous Vein Vascular Smooth Muscle Cell Migration and Endothelial Cell Proliferation. J Vasc Res 2016; 52:383-95. [PMID: 27064272 PMCID: PMC4959467 DOI: 10.1159/000444754] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/14/2016] [Indexed: 12/14/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) migration and proliferation is central to neointima formation in vein graft failure following coronary artery bypass. However, there are currently no pharmacological interventions that prevent vein graft failure through intimal occlusion. It is hence a therapeutic target. Here, we investigated the contribution of GPR35 to human VSMC and endothelial cell (EC) migration, using a scratch-wound assay, and also the contribution to proliferation, using MTS and BrdU assays, in in vitro models using recently characterized human GPR35 ortholog-selective small-molecule agonists and antagonists. Real-time PCR studies showed GPR35 to be robustly expressed in human VSMCs and ECs. Stimulation of GPR35, with either the human-selective agonist pamoic acid or the reference agonist zaprinast, promoted VSMC migration in the scratch-wound assay. These effects were blocked by coincubation with either of the human GPR35-specific antagonists, CID-2745687 or ML-145. These GPR35-mediated effects were produced by inducing alterations in the actin cytoskeleton via the Rho A/Rho kinase signaling axis. Additionally, the agonist ligands stimulated a proliferative response in ECs. These studies highlight the potential that small molecules that stimulate or block GPR35 activity can modulate vascular proliferation and migration. These data propose GPR35 as a translational therapeutic target in vascular remodeling.
Collapse
Affiliation(s)
- Jennifer E McCallum
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | | | | | | | | | | |
Collapse
|
97
|
Toll L, Bruchas MR, Calo' G, Cox BM, Zaveri NT. Nociceptin/Orphanin FQ Receptor Structure, Signaling, Ligands, Functions, and Interactions with Opioid Systems. Pharmacol Rev 2016; 68:419-57. [PMID: 26956246 PMCID: PMC4813427 DOI: 10.1124/pr.114.009209] [Citation(s) in RCA: 226] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The NOP receptor (nociceptin/orphanin FQ opioid peptide receptor) is the most recently discovered member of the opioid receptor family and, together with its endogenous ligand, N/OFQ, make up the fourth members of the opioid receptor and opioid peptide family. Because of its more recent discovery, an understanding of the cellular and behavioral actions induced by NOP receptor activation are less well developed than for the other members of the opioid receptor family. All of these factors are important because NOP receptor activation has a clear modulatory role on mu opioid receptor-mediated actions and thereby affects opioid analgesia, tolerance development, and reward. In addition to opioid modulatory actions, NOP receptor activation has important effects on motor function and other physiologic processes. This review discusses how NOP pharmacology intersects, contrasts, and interacts with the mu opioid receptor in terms of tertiary structure and mechanism of receptor activation; location of receptors in the central nervous system; mechanisms of desensitization and downregulation; cellular actions; intracellular signal transduction pathways; and behavioral actions with respect to analgesia, tolerance, dependence, and reward. This is followed by a discussion of the agonists and antagonists that have most contributed to our current knowledge. Because NOP receptors are highly expressed in brain and spinal cord and NOP receptor activation sometimes synergizes with mu receptor-mediated actions and sometimes opposes them, an understanding of NOP receptor pharmacology in the context of these interactions with the opioid receptors will be crucial to the development of novel therapeutics that engage the NOP receptor.
Collapse
Affiliation(s)
- Lawrence Toll
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Michael R Bruchas
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Girolamo Calo'
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Brian M Cox
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Nurulain T Zaveri
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| |
Collapse
|
98
|
Gpr176 is a Gz-linked orphan G-protein-coupled receptor that sets the pace of circadian behaviour. Nat Commun 2016; 7:10583. [PMID: 26882873 PMCID: PMC4757782 DOI: 10.1038/ncomms10583] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/30/2015] [Indexed: 01/26/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) participate in a broad range of physiological functions. A priority for fundamental and clinical research, therefore, is to decipher the function of over 140 remaining orphan GPCRs. The suprachiasmatic nucleus (SCN), the brain's circadian pacemaker, governs daily rhythms in behaviour and physiology. Here we launch the SCN orphan GPCR project to (i) search for murine orphan GPCRs with enriched expression in the SCN, (ii) generate mutant animals deficient in candidate GPCRs, and (iii) analyse the impact on circadian rhythms. We thereby identify Gpr176 as an SCN-enriched orphan GPCR that sets the pace of circadian behaviour. Gpr176 is expressed in a circadian manner by SCN neurons, and molecular characterization reveals that it represses cAMP signalling in an agonist-independent manner. Gpr176 acts independently of, and in parallel to, the Vipr2 GPCR, not through the canonical Gi, but via the unique G-protein subclass Gz. The suprachiasmatic nucleus (SCN) is the central regulator of circadian rhythms. Here the authors identify mouse Gpr176 as a pace modulator of this circadian clock and characterize its mode of action as coupling to Gz rather than Gi subunits.
Collapse
|
99
|
Black JB, Premont RT, Daaka Y. Feedback regulation of G protein-coupled receptor signaling by GRKs and arrestins. Semin Cell Dev Biol 2016; 50:95-104. [PMID: 26773211 PMCID: PMC4779377 DOI: 10.1016/j.semcdb.2015.12.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 12/19/2015] [Indexed: 12/16/2022]
Abstract
GPCRs are ubiquitous in mammalian cells and present intricate mechanisms for cellular signaling and communication. Mechanistically, GPCR signaling was identified to occur vectorially through heterotrimeric G proteins that are negatively regulated by GRK and arrestin effectors. Emerging evidence highlights additional roles for GRK and Arrestin partners, and establishes the existence of interconnected feedback pathways that collectively define GPCR signaling. GPCRs influence cellular dynamics and can mediate pathologic development, such as cancer and cardiovascular remolding. Hence, a better understanding of their overall signal regulation is of great translational interest and research continues to exploit the pharmacologic potential for modulating their activity.
Collapse
Affiliation(s)
- Joseph B Black
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Richard T Premont
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Yehia Daaka
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, United States.
| |
Collapse
|
100
|
Moniri NH. Free-fatty acid receptor-4 (GPR120): Cellular and molecular function and its role in metabolic disorders. Biochem Pharmacol 2016; 110-111:1-15. [PMID: 26827942 DOI: 10.1016/j.bcp.2016.01.021] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/26/2016] [Indexed: 12/12/2022]
Abstract
Over the last decade, a subfamily of G protein-coupled receptors that are agonized by endogenous and dietary free-fatty acids (FFA) has been discovered. These free-fatty acid receptors include FFA2 and FFA3, which are agonized by short-chained FFA, as well as FFA1 and FFA4, which are agonized by medium-to-long chained FFA. Ligands for FFA1 and FFA4 comprise the family of long chain polyunsaturated omega-3 fatty acids including α-linolenic acid (ALA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), suggesting that many of the long-known beneficial effects of these fats may be receptor mediated. In this regard, FFA4 has gathered considerable interest due to its role in ameliorating inflammation, promoting insulin sensitization, and regulating energy metabolism in response to FFA ligands. The goal of this review is to summarize the body of evidence in regard to FFA4 signal transduction, its mechanisms of regulation, and its functional role in a variety of tissues. In addition, recent endeavors toward discovery of small molecules that modulate FFA4 activity are also presented.
Collapse
Affiliation(s)
- Nader H Moniri
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, GA 30341, United States.
| |
Collapse
|