1
|
Leuthardt AS, Boyle CN, Raun K, Lutz TA, John LM, Le Foll C. Body weight lowering effect of glucose-dependent insulinotropic polypeptide and glucagon-like peptide receptor agonists is more efficient in RAMP1/3 KO than in WT mice. Eur J Pharmacol 2023; 955:175912. [PMID: 37454968 DOI: 10.1016/j.ejphar.2023.175912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
The glucose-dependent insulinotropic polypeptide (GIPR) and glucagon-like peptide (GLP-1R) receptor agonists are insulin secretagogues that have long been shown to improve glycemic control and dual agonists have demonstrated successful weight loss in the clinic. GIPR and GLP-1R populations are located in the dorsal vagal complex where receptor activity-modifying proteins (RAMPs) are also present. According to recent literature, RAMPs not only regulate the signaling of the calcitonin receptor, but also that of other class B G-protein coupled receptors, including members of the glucagon receptor family such as GLP-1R and GIPR. The aim of this study was to investigate whether the absence of RAMP1 and RAMP3 interferes with the action of GIPR and GLP-1R agonists on body weight maintenance and glucose control. To this end, WT and RAMP 1/3 KO mice were fed a 45% high fat diet for 22 weeks and were injected daily with GLP-1R agonist (2 nmol/kg/d; NN0113-2220), GIPR agonist (30 nmol/kg/d; NN0441-0329) or both for 3 weeks. While the mono-agonists exerted little to no body weight lowering and anorectic effects in WT or RAMP1/3 KO mice, but at the given doses, when both compounds were administered together, they synergistically reduced body weight, with a greater effect observed in KO mice. Finally, GLP-1R and GIP/GLP-1R agonist treatment led to improved glucose tolerance, but the absence of RAMPs resulted in an improvement of the HOMA-IR score. These data suggest that RAMPs may play a crucial role in modulating the pharmacological actions of GLP-1 and GIP receptors.
Collapse
Affiliation(s)
- Andrea S Leuthardt
- Institute of Veterinary Physiology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Christina N Boyle
- Institute of Veterinary Physiology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Kirsten Raun
- Global Research, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, CH-8057, Zurich, Switzerland
| | - Linu M John
- Global Research, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, CH-8057, Zurich, Switzerland.
| |
Collapse
|
2
|
Babin KM, Karim JA, Gordon PH, Lennon J, Dickson A, Pioszak AA. Adrenomedullin 2/intermedin is a slow off-rate, long-acting endogenous agonist of the adrenomedullin 2 G protein-coupled receptor. J Biol Chem 2023:104785. [PMID: 37146967 DOI: 10.1016/j.jbc.2023.104785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/20/2023] [Accepted: 05/01/2023] [Indexed: 05/07/2023] Open
Abstract
Adrenomedullin 2/intermedin (AM2/IMD), adrenomedullin (AM), and calcitonin gene-related peptide (CGRP) have signaling functions in the cardiovascular, lymphatic, and nervous systems by activating three heterodimeric receptors comprised of the class B GPCR CLR and a RAMP1, -2, or -3 modulatory subunit. CGRP and AM prefer the RAMP1 and RAMP2/3 complexes, respectively, whereas AM2/IMD is thought to be relatively non-selective. Accordingly, AM2/IMD exhibits overlapping actions with CGRP and AM, so the rationale for this third agonist for the CLR-RAMP complexes is unclear. Here, we report that AM2/IMD is kinetically selective for CLR-RAMP3, known as the AM2R, and we define the structural basis for its distinct kinetics. In live cell biosensor assays, AM2/IMD-AM2R elicited substantially longer duration cAMP signaling than the eight other peptide-receptor combinations. AM2/IMD and AM bound the AM2R with similar equilibrium affinities, but AM2/IMD had a much slower off-rate and longer receptor residence time, thus explaining its prolonged signaling capacity. Peptide and receptor chimeras and mutagenesis were used to map the regions responsible for the distinct binding and signaling kinetics to the AM2/IMD mid-region and the RAMP3 extracellular domain (ECD). Molecular dynamics simulations revealed how the former forms stable interactions at the CLR ECD-transmembrane domain interface and how the latter augments the CLR ECD binding pocket to anchor the AM2/IMD C-terminus. These two strong binding components only combine in the AM2R. Our findings uncover AM2/IMD-AM2R as a cognate pair with unique temporal features, reveal how AM2/IMD and RAMP3 collaborate to shape CLR signaling, and have significant implications for AM2/IMD biology.
Collapse
Affiliation(s)
- Katie M Babin
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Jordan A Karim
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Peyton H Gordon
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - James Lennon
- Departments of Biochemistry and Molecular Biology and Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI 48824
| | - Alex Dickson
- Departments of Biochemistry and Molecular Biology and Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI 48824.
| | - Augen A Pioszak
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104.
| |
Collapse
|
3
|
Babin KM, Karim JA, Gordon PH, Lennon J, Dickson A, Pioszak AA. Adrenomedullin 2/intermedin is a slow off-rate, long-acting endogenous agonist of the adrenomedullin 2 G protein-coupled receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523955. [PMID: 36711519 PMCID: PMC9882245 DOI: 10.1101/2023.01.13.523955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The signaling peptides adrenomedullin 2/intermedin (AM2/IMD), adrenomedullin (AM), and CGRP have overlapping and distinct functions in the cardiovascular, lymphatic, and nervous systems by activating three shared receptors comprised of the class B GPCR CLR in complex with a RAMP1, -2, or -3 modulatory subunit. Here, we report that AM2/IMD, which is thought to be a non-selective agonist, is kinetically selective for CLR-RAMP3, known as the AM 2 R. AM2/IMD-AM 2 R elicited substantially longer duration cAMP signaling than the eight other peptide-receptor combinations due to AM2/IMD slow off-rate binding kinetics. The regions responsible for the slow off-rate were mapped to the AM2/IMD mid-region and the RAMP3 extracellular domain. MD simulations revealed how these bestow enhanced stability to the complex. Our results uncover AM2/IMD-AM 2 R as a cognate pair with unique temporal features, define the mechanism of kinetic selectivity, and explain how AM2/IMD and RAMP3 collaborate to shape the signaling output of a clinically important GPCR.
Collapse
Affiliation(s)
- Katie M. Babin
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Jordan A. Karim
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Peyton H. Gordon
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - James Lennon
- Departments of Biochemistry and Molecular Biology and Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI 48824
| | - Alex Dickson
- Departments of Biochemistry and Molecular Biology and Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI 48824
| | - Augen A. Pioszak
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Lead contact
| |
Collapse
|
4
|
Kotliar IB, Lorenzen E, Schwenk JM, Hay DL, Sakmar TP. Elucidating the Interactome of G Protein-Coupled Receptors and Receptor Activity-Modifying Proteins. Pharmacol Rev 2023; 75:1-34. [PMID: 36757898 PMCID: PMC9832379 DOI: 10.1124/pharmrev.120.000180] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 09/27/2022] [Indexed: 12/13/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are known to interact with several other classes of integral membrane proteins that modulate their biology and pharmacology. However, the extent of these interactions and the mechanisms of their effects are not well understood. For example, one class of GPCR-interacting proteins, receptor activity-modifying proteins (RAMPs), comprise three related and ubiquitously expressed single-transmembrane span proteins. The RAMP family was discovered more than two decades ago, and since then GPCR-RAMP interactions and their functional consequences on receptor trafficking and ligand selectivity have been documented for several secretin (class B) GPCRs, most notably the calcitonin receptor-like receptor. Recent bioinformatics and multiplexed experimental studies suggest that GPCR-RAMP interactions might be much more widespread than previously anticipated. Recently, cryo-electron microscopy has provided high-resolution structures of GPCR-RAMP-ligand complexes, and drugs have been developed that target GPCR-RAMP complexes. In this review, we provide a summary of recent advances in techniques that allow the discovery of GPCR-RAMP interactions and their functional consequences and highlight prospects for future advances. We also provide an up-to-date list of reported GPCR-RAMP interactions based on a review of the current literature. SIGNIFICANCE STATEMENT: Receptor activity-modifying proteins (RAMPs) have emerged as modulators of many aspects of G protein-coupled receptor (GPCR)biology and pharmacology. The application of new methodologies to study membrane protein-protein interactions suggests that RAMPs interact with many more GPCRs than had been previously known. These findings, especially when combined with structural studies of membrane protein complexes, have significant implications for advancing GPCR-targeted drug discovery and the understanding of GPCR pharmacology, biology, and regulation.
Collapse
Affiliation(s)
- Ilana B Kotliar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Emily Lorenzen
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Jochen M Schwenk
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Debbie L Hay
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| |
Collapse
|
5
|
Pearce A, Redfern-Nichols T, Harris M, Poyner DR, Wigglesworth M, Ladds G. Determining the Effects of Differential Expression of GRKs and β-arrestins on CLR-RAMP Agonist Bias. Front Physiol 2022; 13:840763. [PMID: 35422711 PMCID: PMC9001978 DOI: 10.3389/fphys.2022.840763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/08/2022] [Indexed: 11/17/2022] Open
Abstract
Signalling of the calcitonin-like receptor (CLR) is multifaceted, due to its interaction with receptor activity modifying proteins (RAMPs), and three endogenous peptide agonists. Previous studies have focused on the bias of G protein signalling mediated by the receptor and receptor internalisation of the CLR-RAMP complex has been assumed to follow the same pattern as other Class B1 G Protein-Coupled Receptors (GPCRs). Here we sought to measure desensitisation of the three CLR-RAMP complexes in response to the three peptide agonists, through the measurement of β-arrestin recruitment and internalisation. We then delved further into the mechanism of desensitisation through modulation of β-arrestin activity and the expression of GPCR kinases (GRKs), a key component of homologous GPCR desensitisation. First, we have shown that CLR-RAMP1 is capable of potently recruiting β-arrestin1 and 2, subsequently undergoing rapid endocytosis, and that CLR-RAMP2 and -RAMP3 also utilise these pathways, although to a lesser extent. Following this we have shown that agonist-dependent internalisation of CLR is β-arrestin dependent, but not required for full agonism. Overexpression of GRK2-6 was then found to decrease receptor signalling, due to an agonist-independent reduction in surface expression of the CLR-RAMP complex. These results represent the first systematic analysis of the importance of β-arrestins and GRKs in CLR-RAMP signal transduction and pave the way for further investigation regarding other Class B1 GPCRs.
Collapse
Affiliation(s)
- Abigail Pearce
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | | | - Matthew Harris
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - David R. Poyner
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Mark Wigglesworth
- Hit Discovery, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, London, United Kingdom
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Graham Ladds,
| |
Collapse
|
6
|
Shao L, Chen Y, Zhang S, Zhang Z, Cao Y, Yang D, Wang MW. Modulating effects of RAMPs on signaling profiles of the glucagon receptor family. Acta Pharm Sin B 2022; 12:637-650. [PMID: 35256936 PMCID: PMC8897147 DOI: 10.1016/j.apsb.2021.07.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/04/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
Receptor activity-modulating proteins (RAMPs) are accessory molecules that form complexes with specific G protein-coupled receptors (GPCRs) and modulate their functions. It is established that RAMP interacts with the glucagon receptor family of GPCRs but the underlying mechanism is poorly understood. In this study, we used a bioluminescence resonance energy transfer (BRET) approach to comprehensively investigate such interactions. In conjunction with cAMP accumulation, Gαq activation and β-arrestin1/2 recruitment assays, we not only verified the GPCR–RAMP pairs previously reported, but also identified new patterns of GPCR–RAMP interaction. While RAMP1 was able to modify the three signaling events elicited by both glucagon receptor (GCGR) and glucagon-like peptide-1 receptor (GLP-1R), and RAMP2 mainly affected β-arrestin1/2 recruitment by GCGR, GLP-1R and glucagon-like peptide-2 receptor, RAMP3 showed a widespread negative impact on all the family members except for growth hormone-releasing hormone receptor covering the three pathways. Our results suggest that RAMP modulates both G protein dependent and independent signal transduction among the glucagon receptor family members in a receptor-specific manner. Mapping such interactions provides new insights into the role of RAMP in ligand recognition and receptor activation.
Collapse
Key Words
- AMY, amylin
- Allosteric modulation
- BRET, bioluminescence resonance energy transfer
- Bmax, maximum measured BRET value
- CGRP, calcitonin gene-related peptide
- CLR, calcitonin-like receptor
- EC50, half maximal effective concentration
- ECD, extracellular domain
- Emax, maximal response
- G protein-coupled receptor
- GCGR, glucagon receptor
- GHRHR, hormone-releasing hormone receptor
- GIPR, gastric inhibitory polypeptide receptor or glucose-dependent insulinotropic polypeptide
- GLP-1R, glucagon-like peptide-1 receptor
- GLP-2R, glucagon-like peptide-2 receptor
- GPCRs, G protein-coupled receptors
- GPCR–RAMP interaction
- Glucagon receptor family
- Ligand selectivity
- RAMP, receptor activity-modulating protein
- Receptor activity-modulating protein
- Receptor pharmacology
- Rluc, Renilla luciferase
- SBA, suspension bead array
- SCTR, secretin receptor
- SV, splice variant
- Signaling
- TMD, transmembrane domain
- VPAC2R, vasoactive intestinal polypeptide 2 receptor
- cAMP, cyclic adenosine monophosphate
- pEC50, negative logarithm of EC50
- β2-AR, β2-adrenergic receptor
Collapse
Affiliation(s)
- Lijun Shao
- The National Center for Drug Screening and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Chen
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shikai Zhang
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Zhihui Zhang
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yongbing Cao
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Dehua Yang
- The National Center for Drug Screening and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Corresponding authors.
| | - Ming-Wei Wang
- The National Center for Drug Screening and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Pharmacy, Fudan University, Shanghai 201203, China
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Corresponding authors.
| |
Collapse
|
7
|
Vázquez R, Riveiro ME, Berenguer-Daizé C, O'Kane A, Gormley J, Touzelet O, Rezai K, Bekradda M, Ouafik L. Targeting Adrenomedullin in Oncology: A Feasible Strategy With Potential as Much More Than an Alternative Anti-Angiogenic Therapy. Front Oncol 2021; 10:589218. [PMID: 33489885 PMCID: PMC7815935 DOI: 10.3389/fonc.2020.589218] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/02/2020] [Indexed: 12/18/2022] Open
Abstract
The development, maintenance and metastasis of solid tumors are highly dependent on the formation of blood and lymphatic vessels from pre-existing ones through a series of processes that are respectively known as angiogenesis and lymphangiogenesis. Both are mediated by specific growth-stimulating molecules, such as the vascular endothelial growth factor (VEGF) and adrenomedullin (AM), secreted by diverse cell types which involve not only the cancerogenic ones, but also those constituting the tumor stroma (i.e., macrophages, pericytes, fibroblasts, and endothelial cells). In this sense, anti-angiogenic therapy represents a clinically-validated strategy in oncology. Current therapeutic approaches are mainly based on VEGF-targeting agents, which, unfortunately, are usually limited by toxicity and/or tumor-acquired resistance. AM is a ubiquitous peptide hormone mainly secreted in the endothelium with an important involvement in blood vessel development and cardiovascular homeostasis. In this review, we will introduce the state-of-the-art in terms of AM physiology, while putting a special focus on its pro-tumorigenic role, and discuss its potential as a therapeutic target in oncology. A large amount of research has evidenced AM overexpression in a vast majority of solid tumors and a correlation between AM levels and disease stage, progression and/or vascular density has been observed. The analysis presented here indicates that the involvement of AM in the pathogenesis of cancer arises from: 1) direct promotion of cell proliferation and survival; 2) increased vascularization and the subsequent supply of nutrients and oxygen to the tumor; 3) and/or alteration of the cell phenotype into a more aggressive one. Furthermore, we have performed a deep scrutiny of the pathophysiological prominence of each of the AM receptors (AM1 and AM2) in different cancers, highlighting their differential locations and functions, as well as regulatory mechanisms. From the therapeutic point of view, we summarize here an exhaustive series of preclinical studies showing a reduction of tumor angiogenesis, metastasis and growth following treatment with AM-neutralizing antibodies, AM receptor antagonists, or AM receptor interference. Anti-AM therapy is a promising strategy to be explored in oncology, not only as an anti-angiogenic alternative in the context of acquired resistance to VEGF treatment, but also as a potential anti-metastatic approach.
Collapse
Affiliation(s)
- Ramiro Vázquez
- Preclinical Department, Early Drug Development Group (E2DG), Boulogne-Billancourt, France.,Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Maria E Riveiro
- Preclinical Department, Early Drug Development Group (E2DG), Boulogne-Billancourt, France
| | | | - Anthony O'Kane
- Discovery and Scientific Affairs Department, Fusion Antibodies plc., Belfast, United Kingdom
| | - Julie Gormley
- Discovery and Scientific Affairs Department, Fusion Antibodies plc., Belfast, United Kingdom
| | - Olivier Touzelet
- Discovery and Scientific Affairs Department, Fusion Antibodies plc., Belfast, United Kingdom
| | - Keyvan Rezai
- Department of Radio-Pharmacology, Institute Curie-René Huguenin Hospital, Saint-Cloud, France
| | - Mohamed Bekradda
- Preclinical Department, Early Drug Development Group (E2DG), Boulogne-Billancourt, France
| | - L'Houcine Ouafik
- Aix Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France.,APHM, CHU Nord, Service de Transfert d'Oncologie Biologique, Marseille, France
| |
Collapse
|
8
|
Fischer JP, Els-Heindl S, Beck-Sickinger AG. Adrenomedullin - Current perspective on a peptide hormone with significant therapeutic potential. Peptides 2020; 131:170347. [PMID: 32569606 DOI: 10.1016/j.peptides.2020.170347] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 12/11/2022]
Abstract
The peptide hormone adrenomedullin (ADM) consists of 52 amino acids and plays a pivotal role in the regulation of many physiological processes, particularly those of the cardiovascular and lymphatic system. Like calcitonin (CT), calcitonin gene-related peptide (CGRP), intermedin (IMD) and amylin (AMY), it belongs to the CT/CGRP family of peptide hormones, which despite their low little sequence identity share certain characteristic structural features as well as a complex multicomponent receptor system. ADM, IMD and CGRP exert their biological effects by activation of the calcitonin receptor-like receptor (CLR) as a complex with one of three receptor activity-modifying proteins (RAMP), which alter the ligand affinity. Selectivity within the receptor system is largely mediated by the amidated C-terminus of the peptide hormones, which bind to the extracellular domains of the receptors. This enables their N-terminus consisting of a disulfide-bonded ring structure and a helical segment to bind within the transmembrane region and to induce an active receptor confirmation. ADM is expressed in a variety of tissues in the human body and is fundamentally involved in multitude biological processes. Thus, it is of interest as a diagnostic marker and a promising candidate for therapeutic interventions. In order to fully exploit the potential of ADM, it is necessary to improve its pharmacological profile by increasing the metabolic stability and, ideally, creating receptor subtype-selective analogs. While several successful attempts to prolong the half-life of ADM were recently reported, improving or even retaining receptor selectivity remains challenging.
Collapse
Affiliation(s)
- Jan-Patrick Fischer
- Institut für Biochemie, Universität Leipzig, Brüderstraße 34, 04103 Leipzig, Germany
| | - Sylvia Els-Heindl
- Institut für Biochemie, Universität Leipzig, Brüderstraße 34, 04103 Leipzig, Germany
| | | |
Collapse
|
9
|
Tao YX. Molecular chaperones and G protein-coupled receptor maturation and pharmacology. Mol Cell Endocrinol 2020; 511:110862. [PMID: 32389798 DOI: 10.1016/j.mce.2020.110862] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022]
Abstract
G protein-coupled receptors (GPCRs) are highly conserved versatile signaling molecules located at the plasma membrane that respond to diverse extracellular signals. They regulate almost all physiological processes in the vertebrates. About 35% of current drugs target these receptors. Mutations in these genes have been identified as causes of numerous diseases. The seven transmembrane domain structure of GPCRs implies that the folding of these transmembrane proteins is extremely complicated and difficult. Indeed, many wild type GPCRs are not folded optimally. The most common defect in genetic diseases caused by GPCR mutations is misfolding and failure to reach the plasma membrane where it functions. General molecular chaperones aid the folding of all proteins, including GPCRs, by preventing aggregation, promoting folding and disaggregating small aggregates. Some GPCRs need additional receptor-specific chaperones to assist their folding. Many of these receptor-specific chaperones interact with additional receptors and alter receptor pharmacology, expanding the understanding of these chaperone proteins.
Collapse
Affiliation(s)
- Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849-5519, USA.
| |
Collapse
|
10
|
Garelja M, Au M, Brimble MA, Gingell JJ, Hendrikse ER, Lovell A, Prodan N, Sexton PM, Siow A, Walker CS, Watkins HA, Williams GM, Wootten D, Yang SH, Harris PWR, Hay DL. Molecular Mechanisms of Class B GPCR Activation: Insights from Adrenomedullin Receptors. ACS Pharmacol Transl Sci 2020; 3:246-262. [PMID: 32296766 PMCID: PMC7155197 DOI: 10.1021/acsptsci.9b00083] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Indexed: 02/07/2023]
Abstract
Adrenomedullin (AM) is a 52 amino acid peptide that plays a regulatory role in the vasculature. Receptors for AM comprise the class B G protein-coupled receptor, the calcitonin-like receptor (CLR), in complex with one of three receptor activity-modifying proteins (RAMPs). The C-terminus of AM is involved in binding to the extracellular domain of the receptor, while the N-terminus is proposed to interact with the juxtamembranous portion of the receptor to activate signaling. There is currently limited information on the molecular determinants involved in AM signaling, thus we set out to define the importance of the AM N-terminus through five signaling pathways (cAMP production, ERK phosphorylation, CREB phosphorylation, Akt phosphorylation, and IP1 production). We characterized the three CLR:RAMP complexes through the five pathways, finding that each had a distinct repertoire of intracellular signaling pathways that it is able to regulate. We then performed an alanine scan of AM from residues 15-31 and found that most residues could be substituted with only small effects on signaling, and that most substitutions affected signaling through all receptors and pathways in a similar manner. We identify F18, T20, L26, and I30 as being critical for AM function, while also identifying an analogue (AM15-52 G19A) which has unique signaling properties relative to the unmodified AM. We interpret our findings in the context of new structural information, highlighting the complementary nature of structural biology and functional assays.
Collapse
Affiliation(s)
- Michael
L. Garelja
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Maggie Au
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
- Maurice
Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, 1010, New Zealand
| | - Margaret A. Brimble
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
- Maurice
Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, 1010, New Zealand
- School
of Chemical Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Joseph J. Gingell
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
- Maurice
Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, 1010, New Zealand
| | - Erica R. Hendrikse
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Annie Lovell
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Nicole Prodan
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Patrick M. Sexton
- Drug
Discovery Biology and Department of Pharmacology, Monash Institute
of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Andrew Siow
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
- School
of Chemical Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Christopher S. Walker
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
- Maurice
Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, 1010, New Zealand
| | - Harriet A. Watkins
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
- Maurice
Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, 1010, New Zealand
| | - Geoffrey M. Williams
- Maurice
Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, 1010, New Zealand
- School
of Chemical Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Denise Wootten
- Drug
Discovery Biology and Department of Pharmacology, Monash Institute
of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Sung H. Yang
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
- Maurice
Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, 1010, New Zealand
| | - Paul W. R. Harris
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
- Maurice
Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, 1010, New Zealand
- School
of Chemical Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Debbie L. Hay
- School
of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
- Maurice
Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, 1010, New Zealand
| |
Collapse
|
11
|
Serafin DS, Harris NR, Nielsen NR, Mackie DI, Caron KM. Dawn of a New RAMPage. Trends Pharmacol Sci 2020; 41:249-265. [PMID: 32115276 PMCID: PMC7236817 DOI: 10.1016/j.tips.2020.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 01/08/2023]
Abstract
Receptor activity-modifying proteins (RAMPs) interact with G-protein-coupled receptors (GPCRs) to modify their functions, imparting significant implications upon their physiological and therapeutic potentials. Resurging interest in identifying RAMP-GPCR interactions has recently been fueled by coevolution studies and orthogonal technological screening platforms. These new studies reveal previously unrecognized RAMP-interacting GPCRs, many of which expand beyond Class B GPCRs. The consequences of these interactions on GPCR function and physiology lays the foundation for new molecular therapeutic targets, as evidenced by the recent success of erenumab. Here, we highlight recent papers that uncovered novel RAMP-GPCR interactions, human RAMP-GPCR disease-causing mutations, and RAMP-related human pathologies, paving the way for a new era of RAMP-targeted drug development.
Collapse
Affiliation(s)
- D Stephen Serafin
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie R Harris
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Duncan I Mackie
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
12
|
Gingell JJ, Rees TA, Hendrikse ER, Siow A, Rennison D, Scotter J, Harris PWR, Brimble MA, Walker CS, Hay DL. Distinct Patterns of Internalization of Different Calcitonin Gene-Related Peptide Receptors. ACS Pharmacol Transl Sci 2020; 3:296-304. [PMID: 32296769 DOI: 10.1021/acsptsci.9b00089] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Indexed: 02/06/2023]
Abstract
Calcitonin gene-related peptide (CGRP) is a neuropeptide that is involved in the transmission of pain. Drugs targeting CGRP or a CGRP receptor are efficacious in the treatment of migraine. The canonical CGRP receptor is a complex of a G protein-coupled receptor, the calcitonin-like receptor (CLR), with an accessory protein, receptor activity-modifying protein 1 (RAMP1). A second receptor, the AMY1 receptor, a complex of the calcitonin receptor with RAMP1, is a dual high-affinity receptor for CGRP and amylin. Receptor regulatory processes, such as internalization, are crucial for controlling peptide and drug responsiveness. Given the importance of CGRP receptor activity in migraine we compared the internalization profiles of both receptors for CGRP using novel fluorescent probes and a combination of live cell imaging, fixed cell imaging, and ELISA. This revealed stark differences in the regulation of each receptor with the AMY1 receptor unexpectedly showing little internalization.
Collapse
Affiliation(s)
- Joseph J Gingell
- School of Biological Sciences, University of Auckland, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand
| | - Tayla A Rees
- School of Biological Sciences, University of Auckland, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand
| | - Erica R Hendrikse
- School of Biological Sciences, University of Auckland, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand
| | - Andrew Siow
- School of Biological Sciences, University of Auckland, Auckland 1142, New Zealand.,School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - David Rennison
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand.,School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - John Scotter
- Liggins Institute, University of Auckland, Auckland 1023, New Zealand
| | - Paul W R Harris
- School of Biological Sciences, University of Auckland, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand.,School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Margaret A Brimble
- School of Biological Sciences, University of Auckland, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand.,School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Christopher S Walker
- School of Biological Sciences, University of Auckland, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand
| | - Debbie L Hay
- School of Biological Sciences, University of Auckland, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand.,Centre for Brain Research, University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
13
|
Xie J, Guo J, Kanwal Z, Wu M, Lv X, Ibrahim NA, Li P, Buabeid MA, Arafa ESA, Sun Q. Calcitonin and Bone Physiology: In Vitro, In Vivo, and Clinical Investigations. Int J Endocrinol 2020; 2020:3236828. [PMID: 32963524 PMCID: PMC7501564 DOI: 10.1155/2020/3236828] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/18/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Calcitonin was discovered as a peptide hormone that was known to reduce the calcium levels in the systemic circulation. This hypocalcemic effect is produced due to multiple reasons such as inhibition of bone resorption or suppression of calcium release from the bone. Thus, calcitonin was said as a primary regulator of the bone resorption process. This is the reason why calcitonin has been used widely in clinics for the treatment of bone disorders such as osteoporosis, hypercalcemia, and Paget's disease. However, presently calcitonin usage is declined due to the development of efficacious formulations of new drugs. Calcitonin gene-related peptides and several other peptides such as intermedin, amylin, and adrenomedullin (ADM) are categorized in calcitonin family. These peptides are known for the structural similarity with calcitonin. Aside from having a similar structure, these peptides have few overlapping biological activities and signal transduction action through related receptors. However, several other activities are also present that are peptide specific. In vitro and in vivo studies documented the posttreatment effects of calcitonin peptides, i.e., positive effect on bone osteoblasts and their formation and negative effect on osteoclasts and their resorption. The recent research studies carried out on genetically modified mice showed the inhibition of osteoclast activity by amylin, while astonishingly calcitonin plays its role by suppressing osteoblast and bone turnover. This article describes the review of the bone, the activity of the calcitonin family of peptides, and the link between them.
Collapse
Affiliation(s)
- Jingbo Xie
- Department of Orthopedics, Fengcheng People's Hospital, Fengcheng, Jiangxi 331100, China
| | - Jian Guo
- Department of the Second Orthopedics, Hongdu Hospital of Traditional Chinese Medicine Affiliated to Jiangxi University of Traditional Chinese Medicine, Nanchang Hongdu Traditional Chinese Medicine Hospital, Nanchang, Jiangxi 330008, China
| | | | - Mingzheng Wu
- Department of Orthopaedics, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Xiangyang Lv
- Department of Orthopaedics, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, China
| | | | - Ping Li
- Department of Orthopaedics, Ya'an People's Hospital, Ya'an, Sichuan 625000, China
| | | | | | - Qingshan Sun
- Department of Orthopedics, The Third Hospital of Shandong Province, Jinan, Shandong 250031, China
| |
Collapse
|
14
|
Goel D, Un Nisa K, Reza MI, Rahman Z, Aamer S. Aberrant DNA Methylation Pattern may Enhance Susceptibility to Migraine: A Novel Perspective. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 18:504-515. [DOI: 10.2174/1871527318666190809162631] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 06/04/2019] [Accepted: 07/27/2019] [Indexed: 12/17/2022]
Abstract
In today’s world, migraine is one of the most frequent disorders with an estimated world prevalence of 14.7% characterized by attacks of a severe headache making people enfeebled and imposing a big socioeconomic burden. The pathophysiology of a migraine is not completely understood however there are pieces of evidence that epigenetics performs a primary role in the pathophysiology of migraine. Here, in this review, we highlight current evidence for an epigenetic link with migraine in particular DNA methylation of numerous genes involved in migraine pathogenesis. Outcomes of various studies have explained the function of DNA methylation of a several migraine related genes such as RAMP1, CALCA, NOS1, ESR1, MTHFR and NR4A3 in migraine pathogenesis. Mentioned data suggested there exist a strong association of DNA methylation of migraine-related genes in migraine. Although we now have a general understanding of the role of epigenetic modifications of a numerous migraine associated genes in migraine pathogenesis, there are many areas of active research are of key relevance to medicine. Future studies into the complexities of epigenetic modifications will bring a new understanding of the mechanisms of migraine processes and open novel approaches towards therapeutic intervention.
Collapse
Affiliation(s)
- Divya Goel
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education & Research, Guwahati, India
| | - Kaiser Un Nisa
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education & Research, SAS Nagar, India
| | - Mohammad Irshad Reza
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education & Research, SAS Nagar, India
| | - Ziaur Rahman
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education & Research, SAS Nagar, India
| | - Shaikh Aamer
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education & Research, SAS Nagar, India
| |
Collapse
|
15
|
RAMP3 determines rapid recycling of atypical chemokine receptor-3 for guided angiogenesis. Proc Natl Acad Sci U S A 2019; 116:24093-24099. [PMID: 31712427 DOI: 10.1073/pnas.1905561116] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Receptor-activity-modifying proteins (RAMPs) are single transmembrane-spanning proteins which serve as molecular chaperones and allosteric modulators of G-protein-coupled receptors (GPCRs) and their signaling pathways. Although RAMPs have been previously studied in the context of their effects on Family B GPCRs, the coevolution of RAMPs with many GPCR families suggests an expanded repertoire of potential interactions. Using bioluminescence resonance energy transfer-based and cell-surface expression approaches, we comprehensively screen for RAMP interactions within the chemokine receptor family and identify robust interactions between RAMPs and nearly all chemokine receptors. Most notably, we identify robust RAMP interaction with atypical chemokine receptors (ACKRs), which function to establish chemotactic gradients for directed cell migration. Specifically, RAMP3 association with atypical chemokine receptor 3 (ACKR3) diminishes adrenomedullin (AM) ligand availability without changing G-protein coupling. Instead, RAMP3 is required for the rapid recycling of ACKR3 to the plasma membrane through Rab4-positive vesicles following either AM or SDF-1/CXCL12 binding, thereby enabling formation of dynamic spatiotemporal chemotactic gradients. Consequently, genetic deletion of either ACKR3 or RAMP3 in mice abolishes directed cell migration of retinal angiogenesis. Thus, RAMP association with chemokine receptor family members represents a molecular interaction to control receptor signaling and trafficking properties.
Collapse
|
16
|
Naot D, Musson DS, Cornish J. The Activity of Peptides of the Calcitonin Family in Bone. Physiol Rev 2019; 99:781-805. [PMID: 30540227 DOI: 10.1152/physrev.00066.2017] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Calcitonin was discovered over 50 yr ago as a new hormone that rapidly lowers circulating calcium levels. This effect is caused by the inhibition of calcium efflux from bone, as calcitonin is a potent inhibitor of bone resorption. Calcitonin has been in clinical use for conditions of accelerated bone turnover, including Paget's disease and osteoporosis; although in recent years, with the development of drugs that are more potent inhibitors of bone resorption, its use has declined. A number of peptides that are structurally similar to calcitonin form the calcitonin family, which currently includes calcitonin gene-related peptides (αCGRP and βCGRP), amylin, adrenomedullin, and intermedin. Apart from being structurally similar, the peptides signal through related receptors and have some overlapping biological activities, although other activities are peptide specific. In bone, in vitro studies and administration of the peptides to animals generally found inhibitory effects on osteoclasts and bone resorption and positive effects on osteoblasts and bone formation. Surprisingly, studies in genetically modified mice have demonstrated that the physiological role of calcitonin appears to be the inhibition of osteoblast activity and bone turnover, whereas amylin inhibits osteoclast activity. The review article focuses on the activities of peptides of the calcitonin family in bone and the challenges in understanding the relationship between the pharmacological effects and the physiological roles of these peptides.
Collapse
Affiliation(s)
- Dorit Naot
- Department of Medicine, University of Auckland , Auckland , New Zealand
| | - David S Musson
- Department of Medicine, University of Auckland , Auckland , New Zealand
| | - Jillian Cornish
- Department of Medicine, University of Auckland , Auckland , New Zealand
| |
Collapse
|
17
|
Almeida LS, Castro‐Lopes JM, Neto FL, Potes CS. Amylin, a peptide expressed by nociceptors, modulates chronic neuropathic pain. Eur J Pain 2019; 23:784-799. [DOI: 10.1002/ejp.1347] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 11/05/2018] [Accepted: 11/27/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Lígia Sofia Almeida
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto Porto Portugal
- IBMC – Instituto de Biologia Molecular e Celular, Universidade do Porto Porto Portugal
- Departamento de Biomedicina – Unidade de Biologia Experimental, Faculdade de Medicina Universidade do Porto Porto Portugal
| | - José Manuel Castro‐Lopes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto Porto Portugal
- IBMC – Instituto de Biologia Molecular e Celular, Universidade do Porto Porto Portugal
- Departamento de Biomedicina – Unidade de Biologia Experimental, Faculdade de Medicina Universidade do Porto Porto Portugal
| | - Fani Lourença Neto
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto Porto Portugal
- IBMC – Instituto de Biologia Molecular e Celular, Universidade do Porto Porto Portugal
- Departamento de Biomedicina – Unidade de Biologia Experimental, Faculdade de Medicina Universidade do Porto Porto Portugal
| | - Catarina Soares Potes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto Porto Portugal
- IBMC – Instituto de Biologia Molecular e Celular, Universidade do Porto Porto Portugal
- Departamento de Biomedicina – Unidade de Biologia Experimental, Faculdade de Medicina Universidade do Porto Porto Portugal
| |
Collapse
|
18
|
Shindo T, Tanaka M, Kamiyoshi A, Ichikawa-Shindo Y, Kawate H, Yamauchi A, Sakurai T. Regulation of cardiovascular development and homeostasis by the adrenomedullin-RAMP system. Peptides 2019; 111:55-61. [PMID: 29689347 DOI: 10.1016/j.peptides.2018.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 11/18/2022]
Abstract
Adrenomedullin (AM), a member of the calcitonin peptide superfamily, is a peptide involved in both the pathogenesis of cardiovascular diseases and circulatory homeostasis. Its receptor, calcitonin receptor-like receptor (CLR), associates with an accessory protein, receptor activity-modifying protein (RAMP). Depending upon which the three RAMP isoforms (RAMP1-3) it interacts with, CLR functions as a receptor for AM or other calcitonin family peptides. AM knockout mice (-/-) died mid-gestation due to abnormalities in vascular development. We found that phenotypes similar to AM-/- were reproduced only in RAMP2-/- mice. We generated endothelial cell-specific RAMP2 knockout mice (E-RAMP2-/-) and found most E-RAMP2-/- mice died perinatally. In surviving adults, vasculitis and organ fibrosis occurred spontaneously. We next generated drug-inducible cardiac myocyte-specific RAMP2-/- (DI-C-RAMP2-/-) mice, which exhibited dilated cardiomyopathy-like heart failure with cardiac dilatation and myofibril disruption. DI-C-RAMP2-/- hearts also showed changes in mitochondrial structure and downregulation of mitochondria-related genes involved in oxidative phosphorylation and β-oxidation. In contrast to RAMP2-/- mice, RAMP3-/- mice were born with no major abnormalities. In adult RAMP3-/- mice, postnatal angiogenesis was normal, but drainage of subcutaneous lymphatic vessels was delayed. RAMP3-/- mice also showed more severe interstitial edema than in wild-type mice in a tail lymphedema model. These findings show that the AM-RAMP system is a key determinant of cardiovascular integrity and homeostasis from prenatal stages through adulthood. The AM-RAMP2 system mainly regulates vascular development and homeostasis, while the AM-RAMP3 system mainly regulates lymphatic function in adults. The AM-RAMP system may thus have therapeutic potential for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Takayuki Shindo
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan.
| | - Megumu Tanaka
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Akiko Kamiyoshi
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Yuka Ichikawa-Shindo
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Hisaka Kawate
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Akihiro Yamauchi
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan; Japan Bio Products Co., Ltd., Tokyo, Japan
| | - Takayuki Sakurai
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| |
Collapse
|
19
|
Gingell JJ, Hendrikse ER, Hay DL. New Insights into the Regulation of CGRP-Family Receptors. Trends Pharmacol Sci 2019; 40:71-83. [DOI: 10.1016/j.tips.2018.11.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 11/29/2022]
|
20
|
Hay DL, Garelja ML, Poyner DR, Walker CS. Update on the pharmacology of calcitonin/CGRP family of peptides: IUPHAR Review 25. Br J Pharmacol 2017; 175:3-17. [PMID: 29059473 DOI: 10.1111/bph.14075] [Citation(s) in RCA: 265] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 12/19/2022] Open
Abstract
The calcitonin/CGRP family of peptides includes calcitonin, α and β CGRP, amylin, adrenomedullin (AM) and adrenomedullin 2/intermedin (AM2/IMD). Their receptors consist of one of two GPCRs, the calcitonin receptor (CTR) or the calcitonin receptor-like receptor (CLR). Further diversity arises from heterodimerization of these GPCRs with one of three receptor activity-modifying proteins (RAMPs). This gives the CGRP receptor (CLR/RAMP1), the AM1 and AM2 receptors (CLR/RAMP2 or RAMP3) and the AMY1, AMY2 and AMY3 receptors (CTR/RAMPs1-3 complexes, respectively). Apart from the CGRP receptor, there are only peptide antagonists widely available for these receptors, and these have limited selectivity, thus defining the function of each receptor in vivo remains challenging. Further challenges arise from the probable co-expression of CTR with the CTR/RAMP complexes and species-dependent splice variants of the CTR (CT(a) and CT(b) ). Furthermore, the AMY1(a) receptor is activated equally well by both amylin and CGRP, and the preferred receptor for AM2/IMD has been unclear. However, there are clear therapeutic rationales for developing agents against the various receptors for these peptides. For example, many agents targeting the CGRP system are in clinical trials, and pramlintide, an amylin analogue, is an approved therapy for insulin-requiring diabetes. This review provides an update on the pharmacology of the calcitonin family of peptides by members of the corresponding subcommittee of the International Union of Basic and Clinical Pharmacology and colleagues.
Collapse
Affiliation(s)
- Debbie L Hay
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Michael L Garelja
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - David R Poyner
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | | |
Collapse
|
21
|
Roux BT, Bauer CC, McNeish AJ, Ward SG, Cottrell GS. The Role of Ubiquitination and Hepatocyte Growth Factor-Regulated Tyrosine Kinase Substrate in the Degradation of the Adrenomedullin Type I Receptor. Sci Rep 2017; 7:12389. [PMID: 28959041 PMCID: PMC5620052 DOI: 10.1038/s41598-017-12585-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 09/06/2017] [Indexed: 01/27/2023] Open
Abstract
Calcitonin receptor-like receptor (CLR) and the receptor activity-modifying protein 2 (RAMP2) comprise a receptor for adrenomedullin (AM). Although it is known that AM induces internalization of CLR•RAMP2, little is known about the molecular mechanisms that regulate the trafficking of CLR•RAMP2. Using HEK and HMEC-1 cells, we observed that AM-induced activation of CLR•RAMP2 promoted ubiquitination of CLR. A mutant (CLRΔ9KR), lacking all intracellular lysine residues was functional and trafficked similar to the wild-type receptor, but was not ubiquitinated. Degradation of CLR•RAMP2 and CLRΔ9KR•RAMP2 was not dependent on the duration of AM stimulation or ubiquitination and occurred via a mechanism that was partially prevented by peptidase inhibitors. Degradation of CLR•RAMP2 was sensitive to overexpression of hepatocyte growth factor-regulated tyrosine kinase substrate (HRS), but not to HRS knockdown, whereas CLRΔ9KR•RAMP2 degradation was unaffected. Overexpression, but not knockdown of HRS, promoted hyperubiquitination of CLR under basal conditions. Thus, we propose a role for ubiquitin and HRS in the regulation of AM-induced degradation of CLR•RAMP2.
Collapse
Affiliation(s)
- Benoît T Roux
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Claudia C Bauer
- Cellular and Molecular Neuroscience, Reading School of Pharmacy, University of Reading, Reading, RG6 6UB, UK
| | - Alister J McNeish
- Cellular and Molecular Neuroscience, Reading School of Pharmacy, University of Reading, Reading, RG6 6UB, UK
| | - Stephen G Ward
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Graeme S Cottrell
- Cellular and Molecular Neuroscience, Reading School of Pharmacy, University of Reading, Reading, RG6 6UB, UK.
| |
Collapse
|
22
|
Wesseling H, Elgersma Y, Bahn S. A brain proteomic investigation of rapamycin effects in the Tsc1+/- mouse model. Mol Autism 2017; 8:41. [PMID: 28775826 PMCID: PMC5540199 DOI: 10.1186/s13229-017-0151-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 06/14/2017] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) is a rare monogenic disorder characterized by benign tumors in multiple organs as well as a high prevalence of epilepsy, intellectual disability and autism. TSC is caused by inactivating mutations in the TSC1 or TSC2 genes. Heterozygocity induces hyperactivation of mTOR which can be inhibited by mTOR inhibitors, such as rapamycin, which have proven efficacy in the treatment of TSC-associated symptoms. The aim of the present study was (1) to identify molecular changes associated with social and cognitive deficits in the brain tissue of Tsc1+/- mice and (2) to investigate the molecular effects of rapamycin treatment, which has been shown to ameliorate genotype-related behavioural deficits. METHODS Molecular alterations in the frontal cortex and hippocampus of Tsc1+/- and control mice, with or without rapamycin treatment, were investigated. A quantitative mass spectrometry-based shotgun proteomic approach (LC-MSE) was employed as an unbiased method to detect changes in protein levels. Changes identified in the initial profiling stage were validated using selected reaction monitoring (SRM). Protein Set Enrichment Analysis was employed to identify dysregulated pathways. RESULTS LC-MSE analysis of Tsc1+/- mice and controls (n = 30) identified 51 proteins changed in frontal cortex and 108 in the hippocampus. Bioinformatic analysis combined with targeted proteomic validation revealed several dysregulated molecular pathways. Using targeted assays, proteomic alterations in the hippocampus validated the pathways "myelination", "dendrite," and "oxidative stress", an upregulation of ribosomal proteins and the mTOR kinase. LC-MSE analysis was also employed on Tsc1+/- and wildtype mice (n = 34) treated with rapamycin or vehicle. Rapamycin treatment exerted a stronger proteomic effect in Tsc1+/- mice with significant changes (mainly decreased expression) in 231 and 106 proteins, respectively. The cellular pathways "oxidative stress" and "apoptosis" were found to be affected in Tsc1+/- mice and the cellular compartments "myelin sheet" and "neurofilaments" were affected by rapamycin treatment. Thirty-three proteins which were altered in Tsc1+/- mice were normalized following rapamycin treatment, amongst them oxidative stress related proteins, myelin-specific and ribosomal proteins. CONCLUSIONS Molecular changes in the Tsc1+/- mouse brain were more prominent in the hippocampus compared to the frontal cortex. Pathways linked to myelination and oxidative stress response were prominently affected and, at least in part, normalized following rapamycin treatment. The results could aid in the identification of novel drug targets for the treatment of cognitive, social and psychiatric symptoms in autism spectrum disorders. Similar pathways have also been implicated in other psychiatric and neurodegenerative disorders and could imply similar disease processes. Thus, the potential efficacy of mTOR inhibitors warrants further investigation not only for autism spectrum disorders but also for other neuropsychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Hendrik Wesseling
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QT UK
| | - Ype Elgersma
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, 3000 CA The Netherlands
| | - Sabine Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QT UK
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, 3000 CA The Netherlands
| |
Collapse
|
23
|
Cegla J, Jones BJ, Gardiner JV, Hodson DJ, Marjot T, McGlone ER, Tan TM, Bloom SR. RAMP2 Influences Glucagon Receptor Pharmacology via Trafficking and Signaling. Endocrinology 2017; 158:2680-2693. [PMID: 28586439 PMCID: PMC5551549 DOI: 10.1210/en.2016-1755] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 05/08/2017] [Indexed: 01/20/2023]
Abstract
Endogenous satiety hormones provide an attractive target for obesity drugs. Glucagon causes weight loss by reducing food intake and increasing energy expenditure. To further understand the cellular mechanisms by which glucagon and related ligands activate the glucagon receptor (GCGR), we investigated the interaction of the GCGR with receptor activity modifying protein (RAMP)2, a member of the family of receptor activity modifying proteins. We used a combination of competition binding experiments, cell surface enzyme-linked immunosorbent assay, functional assays assessing the Gαs and Gαq pathways and β-arrestin recruitment, and small interfering RNA knockdown to examine the effect of RAMP2 on the GCGR. Ligands tested were glucagon; glucagonlike peptide-1 (GLP-1); oxyntomodulin; and analog G(X), a GLP-1/glucagon coagonist developed in-house. Confocal microscopy was used to assess whether RAMP2 affects the subcellular distribution of GCGR. Here we demonstrate that coexpression of RAMP2 and the GCGR results in reduced cell surface expression of the GCGR. This was confirmed by confocal microscopy, which demonstrated that RAMP2 colocalizes with the GCGR and causes significant GCGR cellular redistribution. Furthermore, the presence of RAMP2 influences signaling through the Gαs and Gαq pathways, as well as recruitment of β-arrestin. This work suggests that RAMP2 may modify the agonist activity and trafficking of the GCGR, with potential relevance to production of new peptide analogs with selective agonist activities.
Collapse
Affiliation(s)
- Jaimini Cegla
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London W12 0NN, United Kingdom
| | - Ben J. Jones
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London W12 0NN, United Kingdom
| | - James V. Gardiner
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London W12 0NN, United Kingdom
| | - David J. Hodson
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston B15 2TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, United Kingdom
- Centre of Membrane Proteins and Receptors, University of Birmingham, Birmingham B15 2TT, and University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Thomas Marjot
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London W12 0NN, United Kingdom
| | - Emma R. McGlone
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London W12 0NN, United Kingdom
| | - Tricia M. Tan
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London W12 0NN, United Kingdom
| | - Stephen R. Bloom
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London W12 0NN, United Kingdom
| |
Collapse
|
24
|
Schönauer R, Els-Heindl S, Beck-Sickinger AG. Adrenomedullin - new perspectives of a potent peptide hormone. J Pept Sci 2017; 23:472-485. [DOI: 10.1002/psc.2953] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 11/24/2016] [Accepted: 11/28/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Ria Schönauer
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry; Leipzig University; Brüderstraße 34 04103 Leipzig Germany
| | - Sylvia Els-Heindl
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry; Leipzig University; Brüderstraße 34 04103 Leipzig Germany
| | - Annette G. Beck-Sickinger
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry; Leipzig University; Brüderstraße 34 04103 Leipzig Germany
| |
Collapse
|
25
|
A Highly Polymorphic Copy Number Variant in the NSF Gene is Associated with Cocaine Dependence. Sci Rep 2016; 6:31033. [PMID: 27498889 PMCID: PMC4976312 DOI: 10.1038/srep31033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 07/12/2016] [Indexed: 12/25/2022] Open
Abstract
Cocaine dependence is a complex psychiatric disorder involving both genetic and environmental factors. Several neurotransmitter systems mediate cocaine’s effects, dependence and relapse, being the components of the neurotransmitter release machinery good candidates for the disorder. Previously, we identified a risk haplotype for cocaine dependence in the NSF gene, encoding the protein N-Ethylmaleimide-Sensitive Factor essential for synaptic vesicle turnover. Here we examined the possible contribution to cocaine dependence of a large copy number variant (CNV) that encompasses part of the NSF gene. We performed a case-control association study in a discovery sample (359 cases and 356 controls) and identified an association between cocaine dependence and the CNV (P = 0.013), that was confirmed in the replication sample (508 cases and 569 controls, P = 7.1e-03) and in a pooled analysis (P = 1.8e-04), with an over-representation of low number of copies in cases. Subsequently, we studied the functional impact of the CNV on gene expression and found that the levels of two NSF transcripts were significantly increased in peripheral blood mononuclear cells (PBMC) along with the number of copies of the CNV. These results, together with a previous study from our group, support the role of NSF in the susceptibility to cocaine dependence.
Collapse
|
26
|
Lindsley CW, Emmitte KA, Hopkins CR, Bridges TM, Gregory KJ, Niswender CM, Conn PJ. Practical Strategies and Concepts in GPCR Allosteric Modulator Discovery: Recent Advances with Metabotropic Glutamate Receptors. Chem Rev 2016; 116:6707-41. [PMID: 26882314 PMCID: PMC4988345 DOI: 10.1021/acs.chemrev.5b00656] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allosteric modulation of GPCRs has initiated a new era of basic and translational discovery, filled with therapeutic promise yet fraught with caveats. Allosteric ligands stabilize unique conformations of the GPCR that afford fundamentally new receptors, capable of novel pharmacology, unprecedented subtype selectivity, and unique signal bias. This review provides a comprehensive overview of the basics of GPCR allosteric pharmacology, medicinal chemistry, drug metabolism, and validated approaches to address each of the major challenges and caveats. Then, the review narrows focus to highlight recent advances in the discovery of allosteric ligands for metabotropic glutamate receptor subtypes 1-5 and 7 (mGlu1-5,7) highlighting key concepts ("molecular switches", signal bias, heterodimers) and practical solutions to enable the development of tool compounds and clinical candidates. The review closes with a section on late-breaking new advances with allosteric ligands for other GPCRs and emerging data for endogenous allosteric modulators.
Collapse
Affiliation(s)
- Craig W. Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kyle A. Emmitte
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, Texas 76107, United States
| | - Corey R. Hopkins
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Thomas M. Bridges
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Karen J. Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville VIC 3052, Australia
| | - Colleen M. Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - P. Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
27
|
Potes C, Pestana A, Pontes M, Caramelo A, Neto F. Amylin modulates the formalin-induced tonic pain behaviours in rats. Eur J Pain 2016; 20:1741-1752. [DOI: 10.1002/ejp.898] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2016] [Indexed: 12/12/2022]
Affiliation(s)
- C.S. Potes
- Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Porto Portugal
- IBMC-Instituto de Biologia Molecular e Celular; Universidade do Porto; Porto Portugal
- Departamento de Biologia Experimental; Faculdade de Medicina; Universidade do Porto; Porto Portugal
| | - A.C. Pestana
- Departamento de Biologia Experimental; Faculdade de Medicina; Universidade do Porto; Porto Portugal
| | - M. Pontes
- Departamento de Biologia Experimental; Faculdade de Medicina; Universidade do Porto; Porto Portugal
| | - A.S. Caramelo
- Departamento de Biologia Experimental; Faculdade de Medicina; Universidade do Porto; Porto Portugal
| | - F.L. Neto
- Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Porto Portugal
- IBMC-Instituto de Biologia Molecular e Celular; Universidade do Porto; Porto Portugal
- Departamento de Biologia Experimental; Faculdade de Medicina; Universidade do Porto; Porto Portugal
| |
Collapse
|
28
|
Klein KR, Matson BC, Caron KM. The expanding repertoire of receptor activity modifying protein (RAMP) function. Crit Rev Biochem Mol Biol 2016; 51:65-71. [PMID: 26740457 DOI: 10.3109/10409238.2015.1128875] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Receptor activity modifying proteins (RAMPs) associate with G-protein-coupled receptors (GPCRs) at the plasma membrane and together bind a variety of peptide ligands, serving as a communication interface between the extracellular and intracellular environments. The collection of RAMP-interacting GPCRs continues to expand and now consists of GPCRs from families A, B and C, suggesting that RAMP activity is extremely prevalent. RAMP association with GPCRs can regulate GPCR function by altering ligand binding, receptor trafficking and desensitization, and downstream signaling pathways. Here, we elaborate on these RAMP-dependent mechanisms of GPCR regulation, which provide opportunities for pharmacological intervention.
Collapse
Affiliation(s)
| | | | - Kathleen M Caron
- a Department of Cell Biology & Physiology and.,b Department of Genetics , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| |
Collapse
|
29
|
Abstract
It is now recognized that G protein-coupled receptors (GPCRs), once considered largely independent functional units, have a far more diverse molecular architecture. Receptor activity-modifying proteins (RAMPs) provide an important example of proteins that interact with GPCRs to modify their function. RAMPs are able to act as pharmacological switches and chaperones, and they can regulate signaling and/or trafficking in a receptor-dependent manner. This review covers recent discoveries in the RAMP field and summarizes the known GPCR partners and functions of RAMPs. We also discuss the first peptide-bound structures of RAMP-GPCR complexes, which give insight into the molecular mechanisms that enable RAMPs to alter the pharmacology and signaling of GPCRs.
Collapse
Affiliation(s)
- Debbie L Hay
- School of Biological Sciences and Maurice Wilkins Center, University of Auckland, Auckland 1142, New Zealand;
| | - Augen A Pioszak
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104;
| |
Collapse
|
30
|
Hay DL, Chen S, Lutz TA, Parkes DG, Roth JD. Amylin: Pharmacology, Physiology, and Clinical Potential. Pharmacol Rev 2015; 67:564-600. [PMID: 26071095 DOI: 10.1124/pr.115.010629] [Citation(s) in RCA: 266] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amylin is a pancreatic β-cell hormone that produces effects in several different organ systems. Here, we review the literature in rodents and in humans on amylin research since its discovery as a hormone about 25 years ago. Amylin is a 37-amino-acid peptide that activates its specific receptors, which are multisubunit G protein-coupled receptors resulting from the coexpression of a core receptor protein with receptor activity-modifying proteins, resulting in multiple receptor subtypes. Amylin's major role is as a glucoregulatory hormone, and it is an important regulator of energy metabolism in health and disease. Other amylin actions have also been reported, such as on the cardiovascular system or on bone. Amylin acts principally in the circumventricular organs of the central nervous system and functionally interacts with other metabolically active hormones such as cholecystokinin, leptin, and estradiol. The amylin-based peptide, pramlintide, is used clinically to treat type 1 and type 2 diabetes. Clinical studies in obesity have shown that amylin agonists could also be useful for weight loss, especially in combination with other agents.
Collapse
Affiliation(s)
- Debbie L Hay
- School of Biological Sciences, Maurice Wilkins Centre for Molecular Biodiscovery and Centre for Brain Research, University of Auckland, Auckland, New Zealand (D.L.H.); Amylin Pharmaceuticals LLC, San Diego, California (S.C., D.G.P.); Institute of Veterinary Physiology, Institute of Laboratory Animal Sciences and Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (T.A.L.); and Intercept Pharmaceuticals, Inc., San Diego, California (J.D.R.)
| | - Steve Chen
- School of Biological Sciences, Maurice Wilkins Centre for Molecular Biodiscovery and Centre for Brain Research, University of Auckland, Auckland, New Zealand (D.L.H.); Amylin Pharmaceuticals LLC, San Diego, California (S.C., D.G.P.); Institute of Veterinary Physiology, Institute of Laboratory Animal Sciences and Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (T.A.L.); and Intercept Pharmaceuticals, Inc., San Diego, California (J.D.R.)
| | - Thomas A Lutz
- School of Biological Sciences, Maurice Wilkins Centre for Molecular Biodiscovery and Centre for Brain Research, University of Auckland, Auckland, New Zealand (D.L.H.); Amylin Pharmaceuticals LLC, San Diego, California (S.C., D.G.P.); Institute of Veterinary Physiology, Institute of Laboratory Animal Sciences and Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (T.A.L.); and Intercept Pharmaceuticals, Inc., San Diego, California (J.D.R.)
| | - David G Parkes
- School of Biological Sciences, Maurice Wilkins Centre for Molecular Biodiscovery and Centre for Brain Research, University of Auckland, Auckland, New Zealand (D.L.H.); Amylin Pharmaceuticals LLC, San Diego, California (S.C., D.G.P.); Institute of Veterinary Physiology, Institute of Laboratory Animal Sciences and Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (T.A.L.); and Intercept Pharmaceuticals, Inc., San Diego, California (J.D.R.)
| | - Jonathan D Roth
- School of Biological Sciences, Maurice Wilkins Centre for Molecular Biodiscovery and Centre for Brain Research, University of Auckland, Auckland, New Zealand (D.L.H.); Amylin Pharmaceuticals LLC, San Diego, California (S.C., D.G.P.); Institute of Veterinary Physiology, Institute of Laboratory Animal Sciences and Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland (T.A.L.); and Intercept Pharmaceuticals, Inc., San Diego, California (J.D.R.)
| |
Collapse
|
31
|
Triplett JC, Zhang Z, Sultana R, Cai J, Klein JB, Büeler H, Butterfield DA. Quantitative expression proteomics and phosphoproteomics profile of brain from PINK1 knockout mice: insights into mechanisms of familial Parkinson's disease. J Neurochem 2015; 133:750-65. [PMID: 25626353 DOI: 10.1111/jnc.13039] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/08/2014] [Accepted: 01/12/2015] [Indexed: 12/27/2022]
Abstract
Parkinson's disease (PD) is an age-related, neurodegenerative motor disorder characterized by progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta and presence of α-synuclein-containing protein aggregates. Mutations in the mitochondrial Ser/Thr kinase PTEN-induced kinase 1 (PINK1) are associated with an autosomal recessive familial form of early-onset PD. Recent studies have suggested that PINK1 plays important neuroprotective roles against mitochondrial dysfunction by phosphorylating and recruiting Parkin, a cytosolic E3 ubiquitin ligase, to facilitate elimination of damaged mitochondria via autophagy-lysosomal pathways. Loss of PINK1 in cells and animals leads to various mitochondrial impairments and oxidative stress, culminating in dopaminergic neuronal death in humans. Using a 2-D polyacrylamide gel electrophoresis proteomics approach, the differences in expressed brain proteome and phosphoproteome between 6-month-old PINK1-deficient mice and wild-type mice were identified. The observed changes in the brain proteome and phosphoproteome of mice lacking PINK1 suggest that defects in signaling networks, energy metabolism, cellular proteostasis, and neuronal structure and plasticity are involved in the pathogenesis of familial PD. Mutations in PINK1 are associated with an early-onset form of Parkinson's disease (PD). This study examines changes in the proteome and phosphoproteome of the PINK1 knockout mouse brain. Alterations were noted in several key proteins associated with: increased oxidative stress, aberrant cellular signaling, altered neuronal structure, decreased synaptic plasticity, reduced neurotransmission, diminished proteostasis networks, and altered metabolism. 14-3-3ε, 14-3-3 protein epsilon; 3-PGDH, phosphoglycerate dehydrogenase; ALDOA, aldolase A; APT1, acyl-protein thioesterase 1; CaM, calmodulin; CBR3, carbonyl reductase [NADPH] 3; ENO2, gamma-enolase; HPRT, hypoxanthine-guanine phosphoribosyltransferase; HSP70, heat-shock-related 70 kDa protein 2; IDHc, cytoplasmic isocitrate dehydrogenase [NADP+]; MAPK1, mitogen-activated protein kinase 1; MEK1, MAP kinase kinase 1; MDHc, cytoplasmic malate dehydrogenase; NFM, neurofilament medium polypeptide; NSF, N-ethylmaleimide-sensitive fusion protein; PHB, prohibitin; PINK1, PTEN-induced putative kinase 1; PPIaseA, peptidyl-prolyl cis-trans isomerase A; PSA2, proteasome subunit alpha type-2; TK, transketolase; VDAC-2, voltage-dependent anion-selective channel protein 2.
Collapse
Affiliation(s)
- Judy C Triplett
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Nag K, Sultana N, Kato A, Dranik A, Nakamura N, Kutsuzawa K, Hirose S, Akaike T. Ligand-induced internalization, recycling, and resensitization of adrenomedullin receptors depend not on CLR or RAMP alone but on the receptor complex as a whole. Gen Comp Endocrinol 2015; 212:156-62. [PMID: 24815888 DOI: 10.1016/j.ygcen.2014.04.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 04/28/2014] [Indexed: 11/24/2022]
Abstract
Adrenomedullins (AM) is a multifaceted distinct subfamily of peptides that belongs to the calcitonin gene-related peptide (CGRP) superfamily. These peptides exert their functional activities via associations of calcitonin receptor-like receptors (CLRs) and receptor activity-modifying proteins (RAMPs) RAMP2 and RAMP3. Recent studies established that RAMPs and CLRs can modify biochemical properties such as trafficking and glycosylation of each other. However there is very little or no understanding regarding how RAMP or CLR influence ligand-induced events of AM-receptor complex. In this study, using pufferfish homologs of CLR (mfCLR1-3) and RAMP (mfRAMP2 and mfRAMP3), we revealed that all combinations of CLR and RAMP quickly underwent ligand-induced internalization; however, their recycling rates were different as follows: mfCLR1-mfRAMP3>mfCLR2-mfRAMP3>mfCLR3-mfRAMP3. Functional receptor assay confirmed that the recycled receptors were resensitized on the plasma membrane. In contrast, a negligible amount of mfCLR1-mfRAMP2 was recycled and reconstituted. Immunocytochemistry results indicated that the lower recovery rate of mfCLR3-mfRAMP3 and mfCLR1-mfRAMP2 was correlated with higher proportion of lysosomal localization of these receptor complexes compared to the other combinations. Collectively our results indicate, for the first time, that the ligand-induced internalization, recycling, and reconstitution properties of RAMP-CLR receptor complexes depend on the receptor-complex as a whole, and not on individual CLR or RAMP alone.
Collapse
Affiliation(s)
- Kakon Nag
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B19 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan; Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W. Hamilton, ON L8N 3Z5, Canada; Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259-B19 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan.
| | - Naznin Sultana
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B19 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Akira Kato
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B19 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Anna Dranik
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main St. W. Hamilton, ON L8N 3Z5, Canada
| | - Nobuhiro Nakamura
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B19 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Koichi Kutsuzawa
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259-B19 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Shigehisa Hirose
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B19 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Toshihiro Akaike
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259-B19 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| |
Collapse
|
33
|
Tao YX, Conn PM. Chaperoning G protein-coupled receptors: from cell biology to therapeutics. Endocr Rev 2014; 35:602-47. [PMID: 24661201 PMCID: PMC4105357 DOI: 10.1210/er.2013-1121] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 03/14/2014] [Indexed: 12/13/2022]
Abstract
G protein-coupled receptors (GPCRs) are membrane proteins that traverse the plasma membrane seven times (hence, are also called 7TM receptors). The polytopic structure of GPCRs makes the folding of GPCRs difficult and complex. Indeed, many wild-type GPCRs are not folded optimally, and defects in folding are the most common cause of genetic diseases due to GPCR mutations. Both general and receptor-specific molecular chaperones aid the folding of GPCRs. Chemical chaperones have been shown to be able to correct the misfolding in mutant GPCRs, proving to be important tools for studying the structure-function relationship of GPCRs. However, their potential therapeutic value is very limited. Pharmacological chaperones (pharmacoperones) are potentially important novel therapeutics for treating genetic diseases caused by mutations in GPCR genes that resulted in misfolded mutant proteins. Pharmacoperones also increase cell surface expression of wild-type GPCRs; therefore, they could be used to treat diseases that do not harbor mutations in GPCRs. Recent studies have shown that indeed pharmacoperones work in both experimental animals and patients. High-throughput assays have been developed to identify new pharmacoperones that could be used as therapeutics for a number of endocrine and other genetic diseases.
Collapse
Affiliation(s)
- Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology (Y.-X.T.), College of Veterinary Medicine, Auburn University, Auburn, Alabama 36849-5519; and Departments of Internal Medicine and Cell Biology (P.M.C.), Texas Tech University Health Science Center, Lubbock, Texas 79430-6252
| | | |
Collapse
|
34
|
G protein-coupled receptors: what a difference a 'partner' makes. Int J Mol Sci 2014; 15:1112-42. [PMID: 24441568 PMCID: PMC3907859 DOI: 10.3390/ijms15011112] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 12/20/2013] [Accepted: 01/08/2014] [Indexed: 01/16/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are important cell signaling mediators, involved in essential physiological processes. GPCRs respond to a wide variety of ligands from light to large macromolecules, including hormones and small peptides. Unfortunately, mutations and dysregulation of GPCRs that induce a loss of function or alter expression can lead to disorders that are sometimes lethal. Therefore, the expression, trafficking, signaling and desensitization of GPCRs must be tightly regulated by different cellular systems to prevent disease. Although there is substantial knowledge regarding the mechanisms that regulate the desensitization and down-regulation of GPCRs, less is known about the mechanisms that regulate the trafficking and cell-surface expression of newly synthesized GPCRs. More recently, there is accumulating evidence that suggests certain GPCRs are able to interact with specific proteins that can completely change their fate and function. These interactions add on another level of regulation and flexibility between different tissue/cell-types. Here, we review some of the main interacting proteins of GPCRs. A greater understanding of the mechanisms regulating their interactions may lead to the discovery of new drug targets for therapy.
Collapse
|
35
|
Desai AJ, Roberts DJ, Richards GO, Skerry TM. Role of receptor activity modifying protein 1 in function of the calcium sensing receptor in the human TT thyroid carcinoma cell line. PLoS One 2014; 9:e85237. [PMID: 24454825 PMCID: PMC3890319 DOI: 10.1371/journal.pone.0085237] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 12/02/2013] [Indexed: 12/15/2022] Open
Abstract
The Calcium Sensing Receptor (CaSR) plays a role in calcium homeostasis by sensing minute changes in serum Ca(2+) and modulating secretion of calciotropic hormones. It has been shown in transfected cells that accessory proteins known as Receptor Activity Modifying Proteins (RAMPs), specifically RAMPs 1 and 3, are required for cell-surface trafficking of the CaSR. These effects have only been demonstrated in transfected cells, so their physiological relevance is unclear. Here we explored CaSR/RAMP interactions in detail, and showed that in thyroid human carcinoma cells, RAMP1 is required for trafficking of the CaSR. Furthermore, we show that normal RAMP1 function is required for intracellular responses to ligands. Specifically, to confirm earlier studies with tagged constructs, and to provide the additional benefit of quantitative stoichiometric analysis, we used fluorescence resonance energy transfer to show equal abilities of RAMP1 and 3 to chaperone CaSR to the cell surface, though RAMP3 interacted more efficiently with the receptor. Furthermore, a higher fraction of RAMP3 than RAMP1 was observed in CaSR-complexes on the cell-surface, suggesting different ratios of RAMPs to CaSR. In order to determine relevance of these findings in an endogenous expression system we assessed the effect of RAMP1 siRNA knock-down in medullary thyroid carcinoma TT cells, (which express RAMP1, but not RAMP3 constitutively) and measured a significant 50% attenuation of signalling in response to CaSR ligands Cinacalcet and neomycin. Blockade of RAMP1 using specific antibodies induced a concentration-dependent reduction in CaSR-mediated signalling in response to Cinacalcet in TT cells, suggesting a novel functional role for RAMP1 in regulation of CaSR signalling in addition to its known role in receptor trafficking. These data provide evidence that RAMPs traffic the CaSR as higher-level oligomers and play a role in CaSR signalling even after cell surface localisation has occurred.
Collapse
Affiliation(s)
- Aditya J. Desai
- The Mellanby Centre for Bone Research, Department of Human Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - David J. Roberts
- The Mellanby Centre for Bone Research, Department of Human Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Gareth O. Richards
- The Mellanby Centre for Bone Research, Department of Human Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Timothy M. Skerry
- The Mellanby Centre for Bone Research, Department of Human Metabolism, University of Sheffield, Sheffield, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Localization of receptors for calcitonin-gene-related peptide to intraganglionic laminar endings of the mouse esophagus: peripheral interaction between vagal and spinal afferents? Histochem Cell Biol 2013; 141:321-35. [PMID: 24203088 DOI: 10.1007/s00418-013-1162-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2013] [Indexed: 12/11/2022]
Abstract
The calcitonin-gene-related peptide (CGRP) receptor is a heterodimer of calcitonin-receptor-like receptor (CLR) and receptor-activity-modifying protein 1 (RAMP1). Despite the importance of CGRP in regulating gastrointestinal functions, nothing is known about the distribution and function of CLR/RAMP1 in the esophagus, where up to 90 % of spinal afferent neurons contain CGRP. We detected CLR/RAMP1 in the mouse esophagus using immunofluorescence and confocal laser scanning microscopy and examined their relationship with neuronal elements of the myenteric plexus. Immunoreactivity for CLR and RAMP1 colocalized with VGLUT2-positive intraganglionic laminar endings (IGLEs), which were contacted by CGRP-positive varicose axons presumably of spinal afferent origin, typically at sites of CRL/RAMP1 immunoreactivity. This provides an anatomical basis for interaction between spinal afferent fibers and IGLEs. Immunoreactive CLR and RAMP1 also colocalized in myenteric neurons. Thus, CGRP-containing spinal afferents may interact with both vagal IGLEs and myenteric neurons in the mouse esophagus, possibly modulating motility reflexes and inflammatory hypersensitivity.
Collapse
|
37
|
Moore EL, Salvatore CA. Targeting a family B GPCR/RAMP receptor complex: CGRP receptor antagonists and migraine. Br J Pharmacol 2012; 166:66-78. [PMID: 21871019 DOI: 10.1111/j.1476-5381.2011.01633.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The clinical effectiveness of antagonizing the calcitonin gene-related peptide (CGRP) receptor for relief of migraine pain has been clearly demonstrated, but the road to the development of these small molecule antagonists has been daunting. The key hurdle that needed to be overcome was the CGRP receptor itself. The vast majority of the current antagonists recognize similar epitopes on the calcitonin receptor-like receptor (CLR) and receptor activity-modifying protein 1 (RAMP1). RAMP1 is a relatively small, single, transmembrane-spanning protein and along with the G-protein-coupled receptor CLR comprise a functional CGRP receptor. The tri-helical extracellular domain of RAMP1 plays a key role in the high affinity binding of CGRP receptor antagonists and drives their species-selective pharmacology. Over the years, a significant amount of mutagenesis data has been generated to identify specific amino acids or regions within CLR and RAMP1 that are critical to antagonist binding and has directed attention to the CLR/RAMP1 extracellular domain (ECD) complex. Recently, the crystal structure of the CGRP receptor ECD has been elucidated and not only reinforces the early mutagenesis data, but provides critical insight into the molecular mechanism of CGRP receptor antagonism. This review will highlight the drug design hurdles that must be overcome to meet the desired potency, selectivity and pharmacokinetic profile while retaining drug-like properties. Although the development of these antagonists has proved challenging, blocking the CGRP receptor may one day represent a new way to manage migraine and offer hope to migraine sufferers.
Collapse
Affiliation(s)
- Eric L Moore
- Department of Pain & Migraine Research, Merck Research Laboratories, West Point, PA, USA.
| | | |
Collapse
|
38
|
Magalhaes AC, Dunn H, Ferguson SS. Regulation of GPCR activity, trafficking and localization by GPCR-interacting proteins. Br J Pharmacol 2012; 165:1717-1736. [PMID: 21699508 DOI: 10.1111/j.1476-5381.2011.01552.x] [Citation(s) in RCA: 254] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
GPCRs represent the largest family of integral membrane proteins and were first identified as receptor proteins that couple via heterotrimeric G-proteins to regulate a vast variety of effector proteins to modulate cellular function. It is now recognized that GPCRs interact with a myriad of proteins that not only function to attenuate their signalling but also function to couple these receptors to heterotrimeric G-protein-independent signalling pathways. In addition, intracellular and transmembrane proteins associate with GPCRs and regulate their processing in the endoplasmic reticulum, trafficking to the cell surface, compartmentalization to plasma membrane microdomains, endocytosis and trafficking between intracellular membrane compartments. The present review will overview the functional consequence of β-arrestin, receptor activity-modifying proteins (RAMPS), regulators of G-protein signalling (RGS), GPCR-associated sorting proteins (GASPs), Homer, small GTPases, PSD95/Disc Large/Zona Occludens (PDZ), spinophilin, protein phosphatases, calmodulin, optineurin and Src homology 3 (SH3) containing protein interactions with GPCRs.
Collapse
Affiliation(s)
- Ana C Magalhaes
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, London, ON, CanadaThe Department of Physiology & Pharmacology, the University of Western Ontario, London, ON, Canada
| | - Henry Dunn
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, London, ON, CanadaThe Department of Physiology & Pharmacology, the University of Western Ontario, London, ON, Canada
| | - Stephen Sg Ferguson
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, London, ON, CanadaThe Department of Physiology & Pharmacology, the University of Western Ontario, London, ON, Canada
| |
Collapse
|
39
|
Kuwasako K, Kitamura K, Nagata S, Nozaki N, Kato J. Characterization of the single transmembrane domain of human receptor activity-modifying protein 3 in adrenomedullin receptor internalization. Biochem Biophys Res Commun 2012; 420:582-7. [PMID: 22445753 DOI: 10.1016/j.bbrc.2012.03.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 03/08/2012] [Indexed: 01/27/2023]
Abstract
Two receptor activity-modifying proteins (RAMP2 and RAMP3) enable calcitonin receptor-like receptor (CLR) to function as two heterodimeric receptors (CLR/RAMP2 and CLR/RAMP3) for adrenomedullin (AM), a potent cardiovascular protective peptide. Following AM stimulation, both receptors undergo rapid internalization through a clathrin-dependent pathway, after which CLR/RAMP3, but not CLR/RAMP2, can be recycled to the cell surface for resensitization. However, human (h)RAMP3 mediates CLR internalization much less efficiently than does hRAMP2. Therefore, the molecular basis of the single transmembrane domain (TMD) and the intracellular domain of hRAMP3 during AM receptor internalization was investigated by transiently transfecting various RAMP chimeras and mutants into HEK-293 cells stably expressing hCLR. Flow cytometric analysis revealed that substituting the RAMP3 TMD with that of RAMP2 markedly enhanced AM-induced internalization of CLR. However, this replacement did not enhance the cell surface expression of CLR, [(125)I]AM binding affinity or AM-induced cAMP response. More detailed analyses showed that substituting the Thr(130)-Val(131) sequence in the RAMP3 TMD with the corresponding sequence (Ile(157)-Pro(158)) from RAMP2 significantly enhanced AM-mediated CLR internalization. In contrast, substituting the RAMP3 target sequence with Ala(130)-Ala(131) did not significantly affect CLR internalization. Thus, the RAMP3 TMD participates in the negative regulation of CLR/RAMP3 internalization, and the aforementioned introduction of the Ile-Pro sequence into the RAMP3 TMD may be a strategy for promoting receptor internalization/resensitization.
Collapse
Affiliation(s)
- Kenji Kuwasako
- Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, Miyazaki 889-1692, Japan.
| | | | | | | | | |
Collapse
|
40
|
Fernàndez-Castillo N, Cormand B, Roncero C, Sánchez-Mora C, Grau-Lopez L, Gonzalvo B, Miquel L, Corominas R, Ramos-Quiroga JA, Casas M, Ribasés M. Candidate pathway association study in cocaine dependence: the control of neurotransmitter release. World J Biol Psychiatry 2012; 13:126-34. [PMID: 21426264 DOI: 10.3109/15622975.2010.551406] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVES Cocaine is the second most used illegal drug in Europe. The transition from use to dependence involves both genetic and environmental factors. Genetic variation in neurotransmitter systems is involved in the susceptibility to cocaine dependence. We examined the possible contribution to cocaine dependence of 16 genes involved in the cellular machinery that controls neurotransmitter release: genes encoding proteins of the SNARE complex (STX1A, SNAP25, VAMP1 and VAMP2), fusion control elements (SYT1, SYT2, CPLX1, CPLX2, CPLX3 and CPLX4) and regulatory elements (STXBP1, SYP, SNPH, NSF, NAPA and RAB3A). METHODS We genotyped 121 SNPs, selected according to genetic coverage criteria, in 360 cocaine-dependent patients and 360 controls from Spain. RESULTS Single and multiple-marker analyses revealed a strong association between cocaine dependence and the NSF gene, encoding the N-ethylmaleimide-sensitive factor (P = 5.1e-04, OR = 2.44 (1.45-4.00) and P = 0.001, OR = 1.82 (1.28-2.59), respectively). The presence and absence of psychotic symptoms were also studied. Interestingly, when we considered the time between initial consumption and the onset of cocaine dependence, we observed that the association was mainly restricted to the group of patients that rapidly developed drug dependence (≤ 2 years; P = 2.98e-06, OR = 1.33 (1.20-1.47)). CONCLUSIONS Our data show preliminary evidence that NSF may predispose not only to cocaine dependence, but also to an early onset of the dependence.
Collapse
|
41
|
Nag K, Sultana N, Hirose S. Calcitonin receptor-like receptor (CLR) influences posttranslational events of receptor activity-modifying proteins (RAMPs). Biochem Biophys Res Commun 2012; 418:824-9. [DOI: 10.1016/j.bbrc.2012.01.116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Accepted: 01/24/2012] [Indexed: 11/29/2022]
|
42
|
|
43
|
Parameswaran N, Spielman WS. Introduction to RAMPs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 744:1-11. [PMID: 22434103 DOI: 10.1007/978-1-4614-2364-5_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Receptor activity modifying proteins (RAMPs) are single transmembrane proteins discovered for their role in the regulation of translocation of certain G-protein coupled receptors (GPCRs) to the plasma membrane. Since its discovery in 1998, several pivotal advances have been made in understanding the function of this family of proteins. This chapter provides a basic introduction to RAMPs as well as details on the various chapters in this book.
Collapse
|
44
|
Sexton PM, Poyner DR, Simms J, Christopoulos A, Hay DL. RAMPs as drug targets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 744:61-74. [PMID: 22434108 DOI: 10.1007/978-1-4614-2364-5_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The receptor activity-modifying protein (RAMP) family of membrane proteins regulates G protein-coupled receptor (GPCR) function in several ways. RAMPs can alter their pharmacology and signalling as well as the trafficking of these receptors to and from the cell surface. Accordingly, RAMPs may be exploited as drug targets, offering new opportunities for regulating the function of therapeutically relevant RAMP-interacting GPCRs. For example, several small molecule antagonists of RAMP1/ calcitonin receptor-like receptor complexes, which block the actions of the neuropeptide calcitonin gene-related peptide are in development for the treatment of migraine headache.
Collapse
Affiliation(s)
- Patrick M Sexton
- Department of Pharmacology, Monash University, Victoria, Australia
| | | | | | | | | |
Collapse
|
45
|
Bomberger JM, Parameswaran N, Spielman WS. Regulation of GPCR trafficking by RAMPs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 744:25-37. [PMID: 22434105 DOI: 10.1007/978-1-4614-2364-5_3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AM and CGRP receptors undergo differential intracellular receptor trafficking upon ligand stimulation. Intracellular trafficking of CLR/RAMP receptor complexes is regulated by posttranslational modifications and protein-protein interactions that differ for each cell type. Recent evidence is accumulating to suggest that the RAMP isoform in complex with CLR may play a role in determining the intracellular trafficking and fate of ligand-stimulated receptor complexes. In this chapter, we will review the current literature on mechanisms of regulating receptor trafficking and roles that have been demonstrated for RAMPs in this regulation.
Collapse
|
46
|
Karpinich NO, Hoopes SL, Kechele DO, Lenhart PM, Caron KM. Adrenomedullin Function in Vascular Endothelial Cells: Insights from Genetic Mouse Models. Curr Hypertens Rev 2011; 7:228-239. [PMID: 22582036 PMCID: PMC3349984 DOI: 10.2174/157340211799304761] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 08/18/2011] [Accepted: 08/21/2011] [Indexed: 01/29/2023]
Abstract
Adrenomedullin is a highly conserved peptide implicated in a variety of physiological processes ranging from pregnancy and embryonic development to tumor progression. This review highlights past and present studies that have contributed to our current appreciation of the important roles adrenomedullin plays in both normal and disease conditions. We provide a particular emphasis on the functions of adrenomedullin in vascular endothelial cells and how experimental approaches in genetic mouse models have helped to drive the field forward.
Collapse
Affiliation(s)
- Natalie O Karpinich
- Department of Cell and Molecular Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | | | | |
Collapse
|
47
|
Ardura JA, Friedman PA. Regulation of G protein-coupled receptor function by Na+/H+ exchange regulatory factors. Pharmacol Rev 2011; 63:882-900. [PMID: 21873413 PMCID: PMC3186079 DOI: 10.1124/pr.110.004176] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Many G protein-coupled receptors (GPCR) exert patterns of cell-specific signaling and function. Mounting evidence now supports the view that cytoplasmic adapter proteins contribute critically to this behavior. Adapter proteins recognize highly conserved motifs such as those for Src homology 3 (SH3), phosphotyrosine-binding (PTB), and postsynaptic density 95/discs-large/zona occludens (PDZ) docking sequences in candidate GPCRs. Here we review the behavior of the Na+/H+ exchange regulatory factor (NHERF) family of PDZ adapter proteins on GPCR signalling, trafficking, and function. Structural determinants of NHERF proteins that allow them to recognize targeted GPCRs are considered. NHERF1 and NHERF2 are capable also of modifying the assembled complex of accessory proteins such as β-arrestins, which have been implicated in regulating GPCR signaling. In addition, NHERF1 and NHERF2 modulate GPCR signaling by altering the G protein to which the receptor binds or affect other regulatory proteins that affect GTPase activity, protein kinase A, phospholipase C, or modify downstream signaling events. Small molecules targeting the site of NHERF1-GPCR interaction are being developed and may become important and selective drug candidates.
Collapse
Affiliation(s)
- Juan A Ardura
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
48
|
Holzmann B. Modulation of immune responses by the neuropeptide CGRP. Amino Acids 2011; 45:1-7. [PMID: 22113645 DOI: 10.1007/s00726-011-1161-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 11/14/2011] [Indexed: 01/15/2023]
Abstract
The peripheral nervous system is connected with lymphoid organs through sensory nerves that mediate pain reflexes and may influence immune responses through the release of neuropeptides such as calcitonin gene-related peptide (CGRP). Local and systemic levels of CGRP increase rapidly during inflammatory responses. CGRP inhibits effector functions of various immune cells and dampens inflammation by distinct pathways involving the amplification of IL-10 production and/or the induction of the transcriptional repressor inducible cAMP early repressor (ICER). Thus, available evidence suggests that, in neuro-immunological interactions, CGRP mediates a potent peptidergic anti-inflammatory pathway.
Collapse
Affiliation(s)
- Bernhard Holzmann
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München, Germany.
| |
Collapse
|
49
|
Barrick CJ, Lenhart PM, Dackor RT, Nagle E, Caron KM. Loss of receptor activity-modifying protein 3 exacerbates cardiac hypertrophy and transition to heart failure in a sex-dependent manner. J Mol Cell Cardiol 2011; 52:165-74. [PMID: 22100352 DOI: 10.1016/j.yjmcc.2011.10.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 10/18/2011] [Accepted: 10/24/2011] [Indexed: 10/15/2022]
Abstract
Sex differences exist in the hypertrophic response, cardiac remodeling, and transition to heart failure of hypertensive patients, and while some of these differences are likely influenced by estrogen, the genetic pathways downstream of estrogen that impact on cardioprotection have yet to be fully elucidated. We have previously shown that the cardioprotective effects of adrenomedullin (AM), an emerging clinical biomarker for cardiovascular disease severity, vary with sex in mouse models. AM signaling during cardiovascular stress is strongly modulated by receptor activity-modifying protein 3 (RAMP3) via its interaction with the G protein-coupled receptor calcitonin receptor-like receptor (CLR). Like AM, RAMP3 expression is potently regulated by estrogen, and so we sought to determine the consequences of genetic Ramp3 loss on cardiac adaptation to chronic hypertension, with a particular focus on characterizing potential sex differences. We generated and bred RAMP3(-/-) mice to RenTgMK mice that consistently display severe angiotensin II-mediated CV disease and compared CV disease progression in RenTgMK to that of RenTgMK:RAMP3(-/-) offspring. As expected, RAMP3 gene expression was higher in cardiovascular tissues of RenTgMK mice and more strongly up-regulated in female RenTgMK mice relative to wildtype controls. RAMP3 loss did not affect the development of hypertension or the presence and severity of perivascular and interstitial fibrosis in the left ventricle (LV). However, echocardiography revealed that while RenTgMK mice developed concentric cardiac hypertrophy with sustained systolic function, male RenTgMK:RAMP3(-/-) mice showed evidence of LV chamber dilatation and depressed systolic function, suggestive of cardiac decompensation. Consistent with these measures of heart failure, male RenTgMK:RAMP3(-/-) mice had increased cardiac apoptosis and elevated activation of Akt. These phenotypes were not present in female RenTgMK:RAMP3(-/-) mice. Collectively, these data demonstrate a sex-dependant, cardioprotective role of RAMP3 in the setting of chronic hypertension.
Collapse
Affiliation(s)
- Cordelia J Barrick
- Department of Cell & Molecular Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
50
|
Kuwasako K, Kitamura K, Nagata S, Hikosaka T, Takei Y, Kato J. Shared and separate functions of the RAMP-based adrenomedullin receptors. Peptides 2011; 32:1540-50. [PMID: 21645567 DOI: 10.1016/j.peptides.2011.05.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 05/20/2011] [Accepted: 05/20/2011] [Indexed: 11/25/2022]
Abstract
Adrenomedullin (AM) is a novel hypotensive peptide that exerts a variety of strongly protective effects against multiorgan damage. AM-specific receptors were first identified as heterodimers composed of calcitonin-receptor-like receptor (CLR), a G protein coupled receptor, and one of two receptor activity-modifying proteins (RAMP2 or RAMP3), which are accessory proteins containing a single transmembrane domain. RAMPs are required for the surface delivery of CLR and the determination of its phenotype. CLR/RAMP2 (AM₁ receptor) is more highly AM-specific than CLR/RAMP3 (AM₂ receptor). Although there have been no reports showing differences in intracellular signaling via the two AM receptors, in vitro studies have shed light on their distinct trafficking and functionality. In addition, the tissue distributions of RAMP2 and RAMP3 differ, and their gene expression is differentially altered under pathophysiological conditions, which is suggestive of the separate roles played by AM₁ and AM₂ receptors in vivo. Both AM and the AM₁ receptor, but not the AM₂ receptor, are crucial for the development of the fetal cardiovascular system and are able to effectively protect against various vascular diseases. However, AM₂ receptors reportedly play an important role in maintaining a normal body weight in old age and may be involved in immune function. In this review article, we focus on the shared and separate functions of the AM receptor subtypes and also discuss the potential for related drug discovery. In addition, we mention their possible function as receptors for AM2 (or intermedin), an AM-related peptide whose biological functions are similar to those of AM.
Collapse
Affiliation(s)
- Kenji Kuwasako
- Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, Miyazaki 889-1692, Japan.
| | | | | | | | | | | |
Collapse
|