1
|
Costanzi S, Stahr LG, Trivellin G, Stratakis CA. GPR101: Modeling a constitutively active receptor linked to X-linked acrogigantism. J Mol Graph Model 2024; 127:108676. [PMID: 38006624 PMCID: PMC10843723 DOI: 10.1016/j.jmgm.2023.108676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 11/27/2023]
Abstract
GPR101 is a G protein-coupled receptor (GPCR) implicated in a rare form of genetic gigantism known as X-linked acrogigantism, or X-LAG. In particular, X-LAG patients harbor microduplications in the long arm of the X-chromosome that invariably include the GPR101 gene. Duplications of the GPR101 gene lead to the formation of a new chromatin domain that causes over-expression of the receptor in the pituitary tumors of the patients. Notably, GPR101 is a constitutively active receptor, which stimulates cells to produce the second messenger cyclic AMP (cAMP) in the absence of ligands. Moreover, GPR101 was recently reported to constitutively activate not only the cAMP pathway via Gs, but also other G protein subunits (Gq/11 and G12/13). Hence, chemicals that block the constitutive activity of GPR101, known as inverse agonists, have the potential to be useful for the development of pharmacological tools for the treatment of X-LAG. In this study, we provide structural insights into the putative structure of GPR101 based on in-house built homology models, as well as third party models based on the machine learning methods AlphaFold and AlphaFold-Multistate. Moreover, we report a molecular dynamics study, meant to further probe the constitutive activity of GPR101. Finally, we provide a structural comparison with the closest GPCRs, which suggests that GPR101 does not share their natural ligands. While this manuscript was under review, cryo-electron microscopy structures of GPR101 were reported. These structures are expected to enable computer-aided ligand discovery efforts targeting GPR101.
Collapse
Affiliation(s)
- Stefano Costanzi
- American University, Department of Chemistry, Washington, DC, USA.
| | - Lea G Stahr
- American University, Department of Chemistry, Washington, DC, USA
| | - Giampaolo Trivellin
- Department of Biomedical Sciences, Humanitas University, Milan, Italy; IRCCS Humanitas Research Hospital, Milan, Italy
| | | |
Collapse
|
2
|
Liessmann F, Künze G, Meiler J. Improving the Modeling of Extracellular Ligand Binding Pockets in RosettaGPCR for Conformational Selection. Int J Mol Sci 2023; 24:7788. [PMID: 37175495 PMCID: PMC10178219 DOI: 10.3390/ijms24097788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest class of drug targets and undergo substantial conformational changes in response to ligand binding. Despite recent progress in GPCR structure determination, static snapshots fail to reflect the conformational space of putative binding pocket geometries to which small molecule ligands can bind. In comparative modeling of GPCRs in the absence of a ligand, often a shrinking of the orthosteric binding pocket is observed. However, the exact prediction of the flexible orthosteric binding site is crucial for adequate structure-based drug discovery. In order to improve ligand docking and guide virtual screening experiments in computer-aided drug discovery, we developed RosettaGPCRPocketSize. The algorithm creates a conformational ensemble of biophysically realistic conformations of the GPCR binding pocket between the TM bundle, which is consistent with a knowledge base of expected pocket geometries. Specifically, tetrahedral volume restraints are defined based on information about critical residues in the orthosteric binding site and their experimentally observed range of Cα-Cα-distances. The output of RosettaGPCRPocketSize is an ensemble of binding pocket geometries that are filtered by energy to ensure biophysically probable arrangements, which can be used for docking simulations. In a benchmark set, pocket shrinkage observed in the default RosettaGPCR was reduced by up to 80% and the binding pocket volume range and geometric diversity were increased. Compared to models from four different GPCR homology model databases (RosettaGPCR, GPCR-Tasser, GPCR-SSFE, and GPCRdb), the here-created models showed more accurate volumes of the orthosteric pocket when evaluated with respect to the crystallographic reference structure. Furthermore, RosettaGPCRPocketSize was able to generate an improved realistic pocket distribution. However, while being superior to other homology models, the accuracy of generated model pockets was comparable to AlphaFold2 models. Furthermore, in a docking benchmark using small-molecule ligands with a higher molecular weight between 400 and 700 Da, a higher success rate in creating native-like binding poses was observed. In summary, RosettaGPCRPocketSize can generate GPCR models with realistic orthosteric pocket volumes, which are useful for structure-based drug discovery applications.
Collapse
Affiliation(s)
- Fabian Liessmann
- Institute for Drug Discovery, Medical Faculty, Leipzig University, 04103 Leipzig, Germany; (F.L.)
| | - Georg Künze
- Institute for Drug Discovery, Medical Faculty, Leipzig University, 04103 Leipzig, Germany; (F.L.)
| | - Jens Meiler
- Institute for Drug Discovery, Medical Faculty, Leipzig University, 04103 Leipzig, Germany; (F.L.)
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, USA
- Center for Scalable Data Analytics and Artificial Intelligence, Leipzig University, 04105 Leipzig, Germany
| |
Collapse
|
3
|
Kumar L, Brenner N, Sledzieski S, Olaosebikan M, Roger LM, Lynn-Goin M, Klein-Seetharaman R, Berger B, Putnam H, Yang J, Lewinski NA, Singh R, Daniels NM, Cowen L, Klein-Seetharaman J. Transfer of knowledge from model organisms to evolutionarily distant non-model organisms: The coral Pocillopora damicornis membrane signaling receptome. PLoS One 2023; 18:e0270965. [PMID: 36735673 PMCID: PMC9897584 DOI: 10.1371/journal.pone.0270965] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
With the ease of gene sequencing and the technology available to study and manipulate non-model organisms, the extension of the methodological toolbox required to translate our understanding of model organisms to non-model organisms has become an urgent problem. For example, mining of large coral and their symbiont sequence data is a challenge, but also provides an opportunity for understanding functionality and evolution of these and other non-model organisms. Much more information than for any other eukaryotic species is available for humans, especially related to signal transduction and diseases. However, the coral cnidarian host and human have diverged over 700 million years ago and homologies between proteins in the two species are therefore often in the gray zone, or at least often undetectable with traditional BLAST searches. We introduce a two-stage approach to identifying putative coral homologues of human proteins. First, through remote homology detection using Hidden Markov Models, we identify candidate human homologues in the cnidarian genome. However, for many proteins, the human genome alone contains multiple family members with similar or even more divergence in sequence. In the second stage, therefore, we filter the remote homology results based on the functional and structural plausibility of each coral candidate, shortlisting the coral proteins likely to have conserved some of the functions of the human proteins. We demonstrate our approach with a pipeline for mapping membrane receptors in humans to membrane receptors in corals, with specific focus on the stony coral, P. damicornis. More than 1000 human membrane receptors mapped to 335 coral receptors, including 151 G protein coupled receptors (GPCRs). To validate specific sub-families, we chose opsin proteins, representative GPCRs that confer light sensitivity, and Toll-like receptors, representative non-GPCRs, which function in the immune response, and their ability to communicate with microorganisms. Through detailed structure-function analysis of their ligand-binding pockets and downstream signaling cascades, we selected those candidate remote homologues likely to carry out related functions in the corals. This pipeline may prove generally useful for other non-model organisms, such as to support the growing field of synthetic biology.
Collapse
Affiliation(s)
- Lokender Kumar
- Department of Chemistry, Colorado School of Mines, Golden, CO, United States of America
| | - Nathanael Brenner
- Department of Chemistry, Colorado School of Mines, Golden, CO, United States of America
| | - Samuel Sledzieski
- MIT Computer Science & Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Monsurat Olaosebikan
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | - Liza M. Roger
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Matthew Lynn-Goin
- Department of Chemistry, Colorado School of Mines, Golden, CO, United States of America
| | | | - Bonnie Berger
- MIT Computer Science & Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Hollie Putnam
- Department of Biological Sciences, University of Rhode Island, South Kingstown, RI, United States of America
| | - Jinkyu Yang
- Department of Department of Aeronautics & Astronautics, University of Washington, Seattle, WA, United States of America
| | - Nastassja A. Lewinski
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Rohit Singh
- MIT Computer Science & Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Noah M. Daniels
- Department of Computer Science and Statistics, University of Rhode Island, South Kingstown, RI, United States of America
| | - Lenore Cowen
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | - Judith Klein-Seetharaman
- Department of Chemistry, Colorado School of Mines, Golden, CO, United States of America
- * E-mail:
| |
Collapse
|
4
|
Mind the Gap—Deciphering GPCR Pharmacology Using 3D Pharmacophores and Artificial Intelligence. Pharmaceuticals (Basel) 2022; 15:ph15111304. [DOI: 10.3390/ph15111304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are amongst the most pharmaceutically relevant and well-studied protein targets, yet unanswered questions in the field leave significant gaps in our understanding of their nuanced structure and function. Three-dimensional pharmacophore models are powerful computational tools in in silico drug discovery, presenting myriad opportunities for the integration of GPCR structural biology and cheminformatics. This review highlights success stories in the application of 3D pharmacophore modeling to de novo drug design, the discovery of biased and allosteric ligands, scaffold hopping, QSAR analysis, hit-to-lead optimization, GPCR de-orphanization, mechanistic understanding of GPCR pharmacology and the elucidation of ligand–receptor interactions. Furthermore, advances in the incorporation of dynamics and machine learning are highlighted. The review will analyze challenges in the field of GPCR drug discovery, detailing how 3D pharmacophore modeling can be used to address them. Finally, we will present opportunities afforded by 3D pharmacophore modeling in the advancement of our understanding and targeting of GPCRs.
Collapse
|
5
|
Dubovski N, Fierro F, Margulis E, Ben Shoshan-Galeczki Y, Peri L, Niv MY. Taste GPCRs and their ligands. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 193:177-193. [PMID: 36357077 DOI: 10.1016/bs.pmbts.2022.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Taste GPCRs are expressed in taste buds on the tongue and play a key role in food choice and consumption. They are also expressed extra-orally, with various physiological roles that are currently under study. Unraveling the roles of these receptors relies on the knowledge of their ligands. Combining sensory, cell-based and computational approaches enabled the discovery of numerous agonists and several antagonists. Here we provide a short overview of taste receptor families, main recent methods for ligands discovery, and current sources of information about known ligands. The future directions that are likely to impact the taste GPCR field include focus on ligand interactions with naturally occurring polymorphisms, as well as harnessing the power of CryoEM and of multiple signaling readout techniques.
Collapse
Affiliation(s)
- Nitzan Dubovski
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Fabrizio Fierro
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Eitan Margulis
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yaron Ben Shoshan-Galeczki
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Lior Peri
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Masha Y Niv
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.
| |
Collapse
|
6
|
G-protein-coupled receptor P2Y10 facilitates chemokine-induced CD4 T cell migration through autocrine/paracrine mediators. Nat Commun 2021; 12:6798. [PMID: 34815397 PMCID: PMC8611058 DOI: 10.1038/s41467-021-26882-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
G-protein-coupled receptors (GPCRs), especially chemokine receptors, play a central role in the regulation of T cell migration. Various GPCRs are upregulated in activated CD4 T cells, including P2Y10, a putative lysophospholipid receptor that is officially still considered an orphan GPCR, i.e., a receptor with unknown endogenous ligand. Here we show that in mice lacking P2Y10 in the CD4 T cell compartment, the severity of experimental autoimmune encephalomyelitis and cutaneous contact hypersensitivity is reduced. P2Y10-deficient CD4 T cells show normal activation, proliferation and differentiation, but reduced chemokine-induced migration, polarization, and RhoA activation upon in vitro stimulation. Mechanistically, CD4 T cells release the putative P2Y10 ligands lysophosphatidylserine and ATP upon chemokine exposure, and these mediators induce P2Y10-dependent RhoA activation in an autocrine/paracrine fashion. ATP degradation impairs RhoA activation and migration in control CD4 T cells, but not in P2Y10-deficient CD4 T cells. Importantly, the P2Y10 pathway appears to be conserved in human T cells. Taken together, P2Y10 mediates RhoA activation in CD4 T cells in response to auto-/paracrine-acting mediators such as LysoPS and ATP, thereby facilitating chemokine-induced migration and, consecutively, T cell-mediated diseases.
Collapse
|
7
|
Free fatty acid receptor 1: a ray of hope in the therapy of type 2 diabetes mellitus. Inflammopharmacology 2021; 29:1625-1639. [PMID: 34669065 DOI: 10.1007/s10787-021-00879-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/21/2021] [Indexed: 12/25/2022]
Abstract
Free fatty acid receptor 1 (FFAR1) is a G-protein coupled receptor with prominent expression on pancreatic beta cells, bones, intestinal cells as well as the nerve cells. This receptor mediates a multitude of functions in the body including release of incretins, secretion of insulin as well as sensation of pain. Since FFAR1 causes secretion of insulin and regulates glucose metabolism, efforts were made to unfold its structure followed by discovering agonists for the receptor and the utilization of these agonists in the therapy of type 2 diabetes mellitus. Development of such functional FFAR1 agonists is a necessity because the currently available therapy for type 2 diabetes mellitus has numerous drawbacks, of which, the major one is hypoglycemia. Since the most prominent effect of the FFAR1 agonists is on glucose concentration in the body, so the major research is focused on treating type 2 diabetes mellitus, though the agonists could benefit other metabolic disorders and neurological disorders as well. The agonists developed so far had one major limitation, i.e., hepatotoxicity. Although, the only agonist that could reach phase 3 clinical trials was TAK-875 developed by Takeda Pharmaceuticals but it was also withdrawn due to toxic effects on the liver. Thus, there are numerous agonists for the varied binding sites of the receptor but no drug available yet. There does seem to be a ray of hope in the drugs that target FFAR1 but a lot more efforts towards drug discovery would result in the successful management of type 2 diabetes mellitus.
Collapse
|
8
|
Ichimura A. Elucidation of the Physiological Functions of Membrane Proteins as Novel Drug Target Candidate Molecules. Biol Pharm Bull 2021; 44:1167-1173. [PMID: 34471043 DOI: 10.1248/bpb.b21-00296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
For pharmaceutical research focused on identifying novel drug target candidate molecules, it is essential to explore unknown biological phenomena, elucidate underlying molecular mechanisms and regulate biological processes based on these findings. Proteins expressed on the plasma membrane and endoplasmic reticulum (ER) membrane play important roles in linking extracellular environmental information to intracellular processes. Stimulating membranous proteins induces various kinds of changes in cells, such as alterations in gene expression levels and enzymatic activities. However, the physiological functions and endogenous ligands of many G-protein-coupled receptors (GPCRs) have not been determined, although GPCRs already constitute a large class of drug-target membrane proteins. Furthermore, the precise physiological roles played by many ER membrane proteins have not been elucidated to date. In this review article, I summarize the results of our recent studies, including the observations that the lipid sensor FFAR4/GPR120 controlled systemic energy homeostasis and that the ER membrane monovalent cation channel trimeric intracellular cation (TRIC)-B and the plasma membrane divalent cation channel transient receptor potential melastatin 7 (TRPM7) regulated bone formation. I further describe the therapeutic significance of these membranous protein-related biological processes.
Collapse
Affiliation(s)
- Atsuhiko Ichimura
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
9
|
Yin YL, Ye C, Zhou F, Wang J, Yang D, Yin W, Wang MW, Xu HE, Jiang Y. Molecular basis for kinin selectivity and activation of the human bradykinin receptors. Nat Struct Mol Biol 2021; 28:755-761. [PMID: 34518695 DOI: 10.1038/s41594-021-00645-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Bradykinin and kallidin are endogenous kinin peptide hormones that belong to the kallikrein-kinin system and are essential to the regulation of blood pressure, inflammation, coagulation and pain control. Des-Arg10-kallidin, the carboxy-terminal des-Arg metabolite of kallidin, and bradykinin selectively activate two G protein-coupled receptors, type 1 and type 2 bradykinin receptors (B1R and B2R), respectively. The hyperactivation of bradykinin receptors, termed 'bradykinin storm', is associated with pulmonary edema in COVID-19 patients, suggesting that bradykinin receptors are important targets for COVID-19 intervention. Here we report two G protein-coupled complex structures of human B1R and B2R bound to des-Arg10-kallidin and bradykinin, respectively. Combined with functional analysis, our structures reveal the mechanism of ligand selectivity and specific activation of the bradykinin receptor. These findings also provide a framework for guiding drug design targeting bradykinin receptors for the treatment of inflammation, cardiovascular disorders and COVID-19.
Collapse
Affiliation(s)
- Yu-Ling Yin
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chenyu Ye
- School of Pharmacy, Fudan University, Shanghai, China
| | - Fulai Zhou
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jia Wang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Dehua Yang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wanchao Yin
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Ming-Wei Wang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,School of Pharmacy, Fudan University, Shanghai, China. .,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China. .,School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - H Eric Xu
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Yi Jiang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
10
|
Wu Y, Zeng L, Zhao S. Ligands of Adrenergic Receptors: A Structural Point of View. Biomolecules 2021; 11:936. [PMID: 34202543 PMCID: PMC8301793 DOI: 10.3390/biom11070936] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/09/2021] [Accepted: 06/12/2021] [Indexed: 01/14/2023] Open
Abstract
Adrenergic receptors are G protein-coupled receptors for epinephrine and norepinephrine. They are targets of many drugs for various conditions, including treatment of hypertension, hypotension, and asthma. Adrenergic receptors are intensively studied in structural biology, displayed for binding poses of different types of ligands. Here, we summarized molecular mechanisms of ligand recognition and receptor activation exhibited by structure. We also reviewed recent advances in structure-based ligand discovery against adrenergic receptors.
Collapse
Affiliation(s)
- Yiran Wu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; (Y.W.); (L.Z.)
| | - Liting Zeng
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; (Y.W.); (L.Z.)
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; (Y.W.); (L.Z.)
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
11
|
Environment-Sensitive Fluorescence of 7-Nitrobenz-2-oxa-1,3-diazol-4-yl (NBD)-Labeled Ligands for Serotonin Receptors. Molecules 2021; 26:molecules26133848. [PMID: 34202630 PMCID: PMC8270269 DOI: 10.3390/molecules26133848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/21/2022] Open
Abstract
Serotonin is a neurotransmitter that plays a crucial role in the regulation of several behavioral and cognitive functions by binding to a number of different serotonin receptors present on the cell surface. We report here the synthesis and characterization of several novel fluorescent analogs of serotonin in which the fluorescent NBD (7-nitrobenz-2-oxa-1,3-diazol-4-yl) group is covalently attached to serotonin. The fluorescent ligands compete with the serotonin1A receptor specific radiolabeled agonist for binding to the receptor. Interestingly, these fluorescent ligands display a high environmental sensitivity of their fluorescence. Importantly, the human serotonin1A receptor stably expressed in CHO-K1 cells could be specifically labeled with one of the fluorescent ligands with minimal nonspecific labeling. Interestingly, we show by spectral imaging that the NBD-labeled ligand exhibits a red edge excitation shift (REES) of 29 nm when bound to the receptor, implying that it is localized in a restricted microenvironment. Taken together, our results show that NBD-labeled serotonin analogs offer an attractive fluorescent approach for elucidating the molecular environment of the serotonin binding site in serotonin receptors. In view of the multiple roles played by the serotonergic systems in the central and peripheral nervous systems, these fluorescent ligands would be useful in future studies involving serotonin receptors.
Collapse
|
12
|
Rahman MT, Decker AM, Langston TL, Mathews KM, Laudermilk L, Maitra R, Ma W, Darcq E, Kieffer BL, Jin C. Design, Synthesis, and Structure-Activity Relationship Studies of (4-Alkoxyphenyl)glycinamides and Bioisosteric 1,3,4-Oxadiazoles as GPR88 Agonists. J Med Chem 2020; 63:14989-15012. [PMID: 33205975 PMCID: PMC7737621 DOI: 10.1021/acs.jmedchem.0c01581] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Increasing evidence implicates the orphan G protein-coupled receptor 88 (GPR88) in a number of striatal-associated disorders. In this study, we report the design and synthesis of a series of novel (4-alkoxyphenyl)glycinamides (e.g., 31) and the corresponding 1,3,4-oxadiazole bioisosteres derived from the 2-AMPP scaffold (1) as GPR88 agonists. The 5-amino-1,3,4-oxadiazole derivatives (84, 88-90) had significantly improved potency and lower lipophilicity compared to 2-AMPP. Compound 84 had an EC50 of 59 nM in the GPR88 overexpressing cell-based cAMP assay. In addition, 84 had an EC50 of 942 nM in the [35S]GTPγS binding assay using mouse striatal membranes but was inactive in membranes from GPR88 knockout mice, even at a concentration of 100 μM. In vivo pharmacokinetic testing of 90 in rats revealed that the 5-amino-1,3,4-oxadiazole analogues may have limited brain permeability. Taken together, these results provide the basis for further optimization to develop a suitable agonist to probe GPR88 functions in the brain.
Collapse
Affiliation(s)
- Md Toufiqur Rahman
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Ann M. Decker
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Tiffany L. Langston
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Kelly M. Mathews
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Lucas Laudermilk
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Rangan Maitra
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States
| | - Weiya Ma
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada
| | - Emmanuel Darcq
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada,INSERM U1114, University of Strasbourg, Strasbourg 67085, France
| | - Brigitte L. Kieffer
- Douglas Research Center, Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada,INSERM U1114, University of Strasbourg, Strasbourg 67085, France
| | - Chunyang Jin
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, North Carolina, 27709, United States,Corresponding author: Dr. Chunyang Jin, Research Triangle Institute, Post Office Box 12194, Research Triangle Park, NC 27709, Telephone: 919 541-6328, Fax: 919 541-8868,
| |
Collapse
|
13
|
Molecular evolution of a collage of cholesterol interaction motifs in transmembrane helix V of the serotonin 1A receptor. Chem Phys Lipids 2020; 232:104955. [PMID: 32846149 DOI: 10.1016/j.chemphyslip.2020.104955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/08/2020] [Accepted: 08/16/2020] [Indexed: 12/20/2022]
Abstract
The human serotonin1A receptor is a representative member of the superfamily of G protein-coupled receptors (GPCRs) and an important drug target for neurological disorders. Using a combination of biochemical, biophysical and molecular dynamics simulation approaches, we and others have shown that membrane cholesterol modulates the organization, dynamics and function of vertebrate serotonin1A receptors. Previous studies have shown that the cytoplasmic portion of transmembrane helix V (TM V) and the extramembraneous intracellular loop 3 are critical for G-protein coupling, phosphorylation and desensitization of the receptor. We have recently resolved a collage of putative cholesterol interaction motifs from the amino acid sequence overlapping this region. In this paper, we explore the sequence plasticity of this fragment that may have adapted to altered membrane lipidome, after vertebrates evolved from primordial invertebrates. Since invertebrates have lower levels of membrane cholesterol relative to vertebrates, we compared TM V sequence fragments from invertebrate serotonin1 receptors with vertebrate orthologs to infer the sequence plasticity in TM V. We report that the average number of cholesterol interaction motifs in TM V for diverse phyla represents an increasing trend that could mirror vertebrate evolution from primordial invertebrates. By statistical modeling, we propose that the collage of cholesterol interaction motifs in TM V of the human serotonin1A receptor may have evolved from rudimentary collages, reminiscent of primordial invertebrate orthologs. Taken together, we propose that a repertoire of cholesterol-philic nonsynonymous substitutions may have enhanced collage complexity in TM V during vertebrate evolution.
Collapse
|
14
|
Li Z, Li J, Liu L, Deng W, Liu Q, Liu R, Zhang W, He Z, Fan L, Yang Y, Duan Y, Hou H, Wang X, Yang Z, Wang X, Chen S, Wang Y, Huang N, Chen J. Structural Insight into the Mechanism of 4-Aminoquinolines Selectivity for the alpha2A-Adrenoceptor. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2585-2594. [PMID: 32694911 PMCID: PMC7340475 DOI: 10.2147/dddt.s214157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/19/2019] [Indexed: 11/23/2022]
Abstract
Background α2A-adrenoceptor (AR) is a potential target for the treatment of degenerative diseases of the central nervous system, and α2A-AR agonists are effective drugs for this condition. However, the lack of high selectivity for α2A-AR subtype of traditional drugs greatly limits their clinic usage. Methods A series of homobivalent 4-aminoquinolines conjugated by two 4-aminoquinoline moieties via varying alkane linker length (C2-C12) were characterized for their affinities for each α2-AR subtype. Subsequently, docking, molecular dynamics and mutagenesis were applied to uncover the molecular mechanism. Results Most 4-aminoquinolines (4-aminoquinoline monomer, C2-C6, C8-C10) were selective for α2A-AR over α2B- and α2C-ARs. Besides, the affinities are of similar linker length-dependence for each α2-AR subtype. Among all the compounds tested, C10 has the highest affinity for α2A-AR (pKi=−7.45±0.62), which is 12-fold and 60-fold selective over α2B-AR and α2C-AR, respectively. Docking and molecular dynamics suggest that C10 simultaneously interacts with orthosteric and “allosteric” sites of the α2A-AR. The mutation of F205 decreases the affinity by 2-fold. The potential allosteric residues include S90, N93, E94 and W99. Conclusion The specificity of C10 for the α2A-AR and the potential orthosteric and allosteric binding sites proposed in this study provide valuable guidance for the development of novel α2A-AR subtype selective compounds.
Collapse
Affiliation(s)
- Zaibing Li
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China.,Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Jingyu Li
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Liyan Liu
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Wenyi Deng
- West China Medical School, Sichuan University, Chengdu 610041, People's Republic of China
| | - Qingrong Liu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Ruofan Liu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Wen Zhang
- West China Medical School, Sichuan University, Chengdu 610041, People's Republic of China
| | - Zaiqing He
- Department of Pathology, Nuclear of Industry 416 Hospital, Chengdu, Sichuan 610051, People's Republic of China
| | - Lei Fan
- Department of Occupational Medicine, Nuclear of Industry 416 Hospital, Chengdu, Sichuan 610051, People's Republic of China
| | - Yingzhuo Yang
- Department of Nuclear Medicine, Sichuan Cancer Hospital, Chengdu 610041, People's Republic of China
| | - Yun Duan
- Department of Nuclear Medicine, Sichuan Cancer Hospital, Chengdu 610041, People's Republic of China
| | - Huifang Hou
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xinyuan Wang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Zhimei Yang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xiaoying Wang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Shanze Chen
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yi Wang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Ning Huang
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| | - Junli Chen
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
15
|
An X, Bai Q, Bing Z, Liu H, Zhang Q, Liu H, Yao X. Revealing the Positive Binding Cooperativity Mechanism between the Orthosteric and the Allosteric Antagonists of CCR2 by Metadynamics and Gaussian Accelerated Molecular Dynamics Simulations. ACS Chem Neurosci 2020; 11:628-637. [PMID: 31968162 DOI: 10.1021/acschemneuro.9b00630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
CC chemokine receptor 2 (CCR2) and its endogenous CC chemokine ligands are associated with numerous inflammatory, neurodegenerative diseases, and cancer. CCR2 is becoming an attractive target in the treatment of autoimmune disease and neurodegenerative diseases. The orthosteric antagonist BMS-681 and allosteric antagonist CCR2-RA-[R] of CCR2 show positive binding cooperativity. We performed well-tempered metadynamics simulations and Gaussian accelerated MD simulations to reveal the influence of the orthosteric antagonist on the unbinding of allosteric antagonist of CCR2. We revealed different unbinding pathways of CCR2-RA-[R] in binary complex CCR2-VT5 and ternary complex CCR2-73R-VT5. The different unbinding pathways of CCR2-RA-[R] are due to the conformational dynamics of TM6. We obtained the significant conformational differences of the intracellular side of TM6 upon CCR2 binding to different ligands by GaMD simulation. The conformational dynamics of TM6 are consistent with the unbinding pathway analysis. GaMD simulations indicate that BMS-681 binding restricts the bend of intracellular side of TM6 by stabilizing the extracellular sides of TM6 and TM7. The charged residues Arg2065.43 of TM5 and Glu2917.39 of TM7 play key roles in stabling TM7 and TM6. TM6 and TM7 are crucial components in the orthosteric and allosteric binding sites. Our results illustrate the conformational details about the effect of the orthosteric antagonist on the allosteric antagonist of CCR2. The conformational dynamics of CCR2 upon binding to different ligands can provide a rational basis for development of allosteric ligands of CCR2.
Collapse
Affiliation(s)
- Xiaoli An
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Qifeng Bai
- School of Basic Medical Science, Lanzhou University, Lanzhou 730000, China
| | - Zhitong Bing
- School of Basic Medical Science, Lanzhou University, Lanzhou 730000, China
- Institute of Modern Physics of Chinese Academy of Sciences, Lanzhou, Gansu 730000, China
| | - Hongli Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P. R. China
| | - Qianqian Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P. R. China
| | - Huanxiang Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P. R. China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou 730000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| |
Collapse
|
16
|
Laboute T, Gandía J, Pellissier LP, Corde Y, Rebeillard F, Gallo M, Gauthier C, Léauté A, Diaz J, Poupon A, Kieffer BL, Le Merrer J, Becker JA. The orphan receptor GPR88 blunts the signaling of opioid receptors and multiple striatal GPCRs. eLife 2020; 9:50519. [PMID: 32003745 PMCID: PMC7012601 DOI: 10.7554/elife.50519] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/30/2020] [Indexed: 12/16/2022] Open
Abstract
GPR88 is an orphan G protein-coupled receptor (GPCR) considered as a promising therapeutic target for neuropsychiatric disorders; its pharmacology, however, remains scarcely understood. Based on our previous report of increased delta opioid receptor activity in Gpr88 null mice, we investigated the impact of GPR88 co-expression on the signaling of opioid receptors in vitro and revealed that GPR88 inhibits the activation of both their G protein- and β-arrestin-dependent signaling pathways. In Gpr88 knockout mice, morphine-induced locomotor sensitization, withdrawal and supra-spinal analgesia were facilitated, consistent with a tonic inhibitory action of GPR88 on µOR signaling. We then explored GPR88 interactions with more striatal versus non-neuronal GPCRs, and revealed that GPR88 can decrease the G protein-dependent signaling of most receptors in close proximity, but impedes β-arrestin recruitment by all receptors tested. Our study unravels an unsuspected buffering role of GPR88 expression on GPCR signaling, with intriguing consequences for opioid and striatal functions.
Collapse
Affiliation(s)
- Thibaut Laboute
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France
| | - Jorge Gandía
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France
| | - Lucie P Pellissier
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France.,Biology and Bioinformatics of Signalling Systems, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Nouzilly, France
| | - Yannick Corde
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France
| | - Florian Rebeillard
- Cellular Biology and Molecular Pharmacology of central Receptors, Centre de Psychiatrie et Neurosciences, Inserm UMR_S894 - Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria Gallo
- Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Christophe Gauthier
- Biology and Bioinformatics of Signalling Systems, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Nouzilly, France
| | - Audrey Léauté
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France
| | - Jorge Diaz
- Cellular Biology and Molecular Pharmacology of central Receptors, Centre de Psychiatrie et Neurosciences, Inserm UMR_S894 - Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Poupon
- Biology and Bioinformatics of Signalling Systems, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Nouzilly, France
| | - Brigitte L Kieffer
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Canada.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, Inserm U1258, Université de Strasbourg, 1 rue Laurent Fries, Illkirch, France
| | - Julie Le Merrer
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, Inserm U1258, Université de Strasbourg, 1 rue Laurent Fries, Illkirch, France
| | - Jérôme Aj Becker
- Deficits of Reward GPCRs and Sociability, Physiologie de la Reproduction et des Comportements, INRA UMR-0085, CNRS UMR-7247, Université de Tours, Inserm, Nouzilly, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, Inserm U1258, Université de Strasbourg, 1 rue Laurent Fries, Illkirch, France
| |
Collapse
|
17
|
Xu Y. Targeting Lysophosphatidic Acid in Cancer: The Issues in Moving from Bench to Bedside. Cancers (Basel) 2019; 11:E1523. [PMID: 31658655 PMCID: PMC6826372 DOI: 10.3390/cancers11101523] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/02/2019] [Accepted: 10/08/2019] [Indexed: 12/16/2022] Open
Abstract
Since the clear demonstration of lysophosphatidic acid (LPA)'s pathological roles in cancer in the mid-1990s, more than 1000 papers relating LPA to various types of cancer were published. Through these studies, LPA was established as a target for cancer. Although LPA-related inhibitors entered clinical trials for fibrosis, the concept of targeting LPA is yet to be moved to clinical cancer treatment. The major challenges that we are facing in moving LPA application from bench to bedside include the intrinsic and complicated metabolic, functional, and signaling properties of LPA, as well as technical issues, which are discussed in this review. Potential strategies and perspectives to improve the translational progress are suggested. Despite these challenges, we are optimistic that LPA blockage, particularly in combination with other agents, is on the horizon to be incorporated into clinical applications.
Collapse
Affiliation(s)
- Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 950 W. Walnut Street R2-E380, Indianapolis, IN 46202, USA.
| |
Collapse
|
18
|
Apel AK, Cheng RK, Tautermann CS, Brauchle M, Huang CY, Pautsch A, Hennig M, Nar H, Schnapp G. Crystal Structure of CC Chemokine Receptor 2A in Complex with an Orthosteric Antagonist Provides Insights for the Design of Selective Antagonists. Structure 2019; 27:427-438.e5. [DOI: 10.1016/j.str.2018.10.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/08/2018] [Accepted: 10/25/2018] [Indexed: 12/23/2022]
|
19
|
Fatakia SN, Sarkar P, Chattopadhyay A. A collage of cholesterol interaction motifs in the serotonin 1A receptor: An evolutionary implication for differential cholesterol interaction. Chem Phys Lipids 2019; 221:184-192. [PMID: 30822391 DOI: 10.1016/j.chemphyslip.2019.02.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 12/22/2022]
Abstract
The serotonin1A receptor is a representative member of the G protein-coupled receptor (GPCR) superfamily and acts as an important drug target. In our previous work, we comprehensively demonstrated that membrane cholesterol is necessary in the organization, dynamics and function of the serotonin1A receptor. In this context, analysis of high-resolution GPCR crystal structures in general and in silico studies of the serotonin1A receptor in particular, have suggested the presence of cholesterol interaction sites (hotspots) in various regions of the receptor. In this work, we have identified an evolutionarily conserved collage of four categories of cholesterol interaction motifs associated with transmembrane helix V and the adjacent intracellular loop 3 fragment of the vertebrate serotonin1A receptor. This collage of motifs represents a total of twenty diverse context-dependent cholesterol interaction configurations. We envision that the gamut of cholesterol interaction sites, characterized by sequence plasticity in cholesterol interaction, could be relevant in receptor-cholesterol interaction in membranes of varying cholesterol content and organization, as found in diverse cell types. We conclude that an evolutionarily conserved mechanism of GPCR-cholesterol interaction allows the serotonin1A receptor to adapt to diverse membrane cholesterol levels during natural evolution.
Collapse
Affiliation(s)
- Sarosh N Fatakia
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India.
| | - Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
20
|
Molecular features characterizing non-peptide selectivity to the human B1 and B2 bradykinin receptors. Bioorg Med Chem Lett 2018; 29:11-14. [PMID: 30466897 DOI: 10.1016/j.bmcl.2018.11.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/08/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
Abstract
Bradykinin is produced in response to inflammation, trauma, burns, shock, allergy and some cardiovascular diseases. Actions of this peptide are mediated through two different G-protein coupled receptors, named B1 and B2 that have different pharmacological characteristics. The former is up-regulated during inflammation episodes or tissue trauma whereas, the latter is constitutively expressed in a variety of cell types. In a previous work we have characterized the molecular features that explain the observed structure-activity results for both receptors by means of molecular modeling studies, using diverse ligands for both receptors. These results were summarized in the form of two different pharmacophores that provided new insights to be used for the design of novel molecules with antagonistic profile. In the present work, we compare these pharmacophores to understand the features that characterize ligand selectivity to the two bradykinin receptors. The study shows that most of the residues involved in the binding pocket are similar in both receptors and consequently are the pharmacophores obtained. The main difference between the two pharmacophores remains on point #5 that involves a polar moiety for the B1 receptor and an aromatic ring for the B2 receptor. Accordingly, analysis of the prospective bound conformation of several non-selective small molecule ligands of the bradykinin receptors permits to conclude that fulfilment of point#5 is a requirement to produce selective ligands. However, the study also shows that this is a necessary condition only, since ligands need also to be bulky enough to avoid binding to these receptors in diverse poses. These results provide new insights for a better understanding of the molecular features that ligands are required to exhibit to be selective bradykinin ligands.
Collapse
|
21
|
Kurczab R, Śliwa P, Rataj K, Kafel R, Bojarski AJ. Salt Bridge in Ligand-Protein Complexes-Systematic Theoretical and Statistical Investigations. J Chem Inf Model 2018; 58:2224-2238. [PMID: 30351056 DOI: 10.1021/acs.jcim.8b00266] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although the salt bridge is the strongest among all known noncovalent molecular interactions, no comprehensive studies have been conducted to date to examine its role and significance in drug design. Thus, a systematic study of the salt bridge in biological systems is reported herein, with a broad analysis of publicly available data from Protein Data Bank, DrugBank, ChEMBL, and GPCRdb. The results revealed the distance and angular preferences as well as privileged molecular motifs of salt bridges in ligand-receptor complexes, which could be used to design the strongest interactions. Moreover, using quantum chemical calculations at the MP2 level, the energetic, directionality, and spatial variabilities of salt bridges were investigated using simple model systems mimicking salt bridges in a biological environment. Additionally, natural orbitals for chemical valence (NOCV) combined with the extended-transition-state (ETS) bond-energy decomposition method (ETS-NOCV) were analyzed and indicated a strong covalent contribution to the salt bridge interaction. The present results could be useful for implementation in rational drug design protocols.
Collapse
Affiliation(s)
- Rafał Kurczab
- Department of Medicinal Chemistry, Institute of Pharmacology , Polish Academy of Sciences , Smetna 12 , 31-343 Cracow , Poland
| | - Paweł Śliwa
- Faculty of Chemical Engineering and Technology , Cracow University of Technology , Warszawska 24 , 31-155 Cracow , Poland
| | - Krzysztof Rataj
- Department of Medicinal Chemistry, Institute of Pharmacology , Polish Academy of Sciences , Smetna 12 , 31-343 Cracow , Poland
| | - Rafał Kafel
- Department of Medicinal Chemistry, Institute of Pharmacology , Polish Academy of Sciences , Smetna 12 , 31-343 Cracow , Poland
| | - Andrzej J Bojarski
- Department of Medicinal Chemistry, Institute of Pharmacology , Polish Academy of Sciences , Smetna 12 , 31-343 Cracow , Poland
| |
Collapse
|
22
|
GPCR-SAS: A web application for statistical analyses on G protein-coupled receptors sequences. PLoS One 2018; 13:e0199843. [PMID: 30044824 PMCID: PMC6059404 DOI: 10.1371/journal.pone.0199843] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/14/2018] [Indexed: 11/19/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are one of the largest protein families in mammals. They mediate signal transduction across cell membranes and are important targets for the pharmaceutical industry. The G Protein-Coupled Receptors-Sequence Analysis and Statistics (GPCR-SAS) web application provides a set of tools to perform comparative analysis of sequence positions between receptors, based on a curated structural-informed multiple sequence alignment. The analysis tools include: (i) percentage of occurrence of an amino acid or motif and entropy at a position or range of positions, (ii) covariance of two positions, (iii) correlation between two amino acids in two positions (or two sequence motifs in two ranges of positions), and (iv) snake-plot representation for a specific receptor or for the consensus sequence of a group of selected receptors. The analysis of conservation of residues and motifs across transmembrane (TM) segments may guide the design of more selective ligands or help to rationalize activation mechanisms, among others. As an example, here we analyze the amino acids of the "transmission switch", that initiates receptor activation following ligand binding. The tool is freely accessible at http://lmc.uab.cat/gpcrsas/.
Collapse
|
23
|
Szpakowska M, Meyrath M, Reynders N, Counson M, Hanson J, Steyaert J, Chevigné A. Mutational analysis of the extracellular disulphide bridges of the atypical chemokine receptor ACKR3/CXCR7 uncovers multiple binding and activation modes for its chemokine and endogenous non-chemokine agonists. Biochem Pharmacol 2018. [DOI: 10.1016/j.bcp.2018.03.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
24
|
Chemical Diversity in the G Protein-Coupled Receptor Superfamily. Trends Pharmacol Sci 2018; 39:494-512. [PMID: 29576399 DOI: 10.1016/j.tips.2018.02.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/05/2018] [Accepted: 02/06/2018] [Indexed: 12/20/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell signaling transmembrane proteins that can be modulated by a plethora of chemical compounds. Systematic cheminformatics analysis of structurally and pharmacologically characterized GPCR ligands shows that cocrystallized GPCR ligands cover a significant part of chemical ligand space, despite their limited number. Many GPCR ligands and substructures interact with multiple receptors, providing a basis for polypharmacological ligand design. Experimentally determined GPCR structures represent a variety of binding sites and receptor-ligand interactions that can be translated to chemically similar ligands for which structural data are lacking. This integration of structural, pharmacological, and chemical information on GPCR-ligand interactions enables the extension of the structural GPCR-ligand interactome and the structure-based design of novel modulators of GPCR function.
Collapse
|
25
|
Shankar V, Goddard WA, Kim SK, Abrol R, Liu F. The 3D Structure of Human DP Prostaglandin G-Protein-Coupled Receptor Bound to Cyclopentanoindole Antagonist, Predicted Using the DuplexBiHelix Modification of the GEnSeMBLE Method. J Chem Theory Comput 2018; 14:1624-1642. [PMID: 29268008 DOI: 10.1021/acs.jctc.7b00842] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Prostaglandins play a critical physiological role in both cardiovascular and immune systems, acting through their interactions with 9 prostanoid G protein-coupled receptors (GPCRs). These receptors are important therapeutic targets for a variety of diseases including arthritis, allergies, type 2 diabetes, and cancer. The DP prostaglandin receptor is of interest because it has unique structural and physiological properties. Most notably, DP does not have the 3-6 ionic lock common to Class A GPCRs. However, the lack of X-ray structures for any of the 9 prostaglandin GPCRs hampers the application of structure-based drug design methods to develop more selective and active medications to specific receptors. We predict here 3D structures for the DP prostaglandin GPCR, based on the GEnSeMBLE complete sampling with hierarchical scoring (CS-HS) methodology. This involves evaluating the energy of 13 trillion packings to finally select the best 20 that are stable enough to be relevant for binding to antagonists, agonists, and modulators. To validate the predicted structures, we predict the binding site for the Merck cyclopentanoindole (CPI) selective antagonist docked to DP. We find that the CPI binds vertically in the 1-2-7 binding pocket, interacting favorably with residues R3107.40 and K762.54 with additional interactions with S3137.43, S3167.46, S191.35, etc. This binding site differs significantly from that of antagonists to known Class A GPCRs where the ligand binds in the 3-4-5-6 region. We find that the predicted binding site leads to reasonable agreement with experimental Structure-Activity Relationship (SAR). We suggest additional mutation experiments including K762.54, E1293.49, L1233.43, M2706.40, F2746.44 to further validate the structure, function, and activation mechanism of receptors in the prostaglandin family. Our structures and binding sites are largely consistent and improve upon the predictions by Li et al. ( J. Am. Chem. Soc. 2007 , 129 ( 35 ), 10720 ) that used our earlier MembStruk prediction methodology.
Collapse
Affiliation(s)
- Vishnu Shankar
- Materials and Process Simulation Center (139-74) , California Institute of Technology , 1200 E. California Blvd. , Pasadena , California 91125 , United States
| | - William A Goddard
- Materials and Process Simulation Center (139-74) , California Institute of Technology , 1200 E. California Blvd. , Pasadena , California 91125 , United States
| | - Soo-Kyung Kim
- Materials and Process Simulation Center (139-74) , California Institute of Technology , 1200 E. California Blvd. , Pasadena , California 91125 , United States
| | - Ravinder Abrol
- Materials and Process Simulation Center (139-74) , California Institute of Technology , 1200 E. California Blvd. , Pasadena , California 91125 , United States
| | - Fan Liu
- Materials and Process Simulation Center (139-74) , California Institute of Technology , 1200 E. California Blvd. , Pasadena , California 91125 , United States
| |
Collapse
|
26
|
Kaczor AA, Żuk J, Matosiuk D. Comparative molecular field analysis and molecular dynamics studies of the dopamine D 2 receptor antagonists without a protonatable nitrogen atom. Med Chem Res 2018; 27:1149-1166. [PMID: 29576721 PMCID: PMC5854747 DOI: 10.1007/s00044-018-2137-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/20/2018] [Indexed: 11/24/2022]
Abstract
The dopaminergic hypothesis of schizophrenia is the main concept explaining the direct reasons of schizophrenia and the effectiveness of current antipsychotics. All antipsychotics present on the market are potent dopamine D2 receptor antagonists or partial agonists. In this work we investigate a series of dopamine D2 receptor antagonists which do not fulfill the criteria of the classical pharmacophore model as they do not possess a protonatable nitrogen atom necessary to interact with the conserved Asp(3.32). Such compounds are interesting, inter alia, due to possible better pharmacokinetic profile when compared to basic, ionizable molecules. By means of homology modeling, molecular docking and molecular dynamics we determined that the compounds investigated interact with Asp(3.32) via their amide nitrogen atom. It was found that the studied compounds stabilize the receptor inactive conformation through the effect on the ionic lock, which is typical for GPCR antagonists. We constructed a CoMFA model for the studied compounds with the following statistics: R2 = 0.95, Q2 = 0.63. The quality of the CoMFA model was confirmed by high value of R2 of the test set, equal 0.96. The CoMFA model indicated two regions where bulky substituents are favored and two regions where bulky substituents are not beneficial. Two red contour regions near carbonyl groups were identified meaning that negative charge would be favored here. Furthermore, the S-oxide group is connected with blue contour region meaning that positive charge is favored in this position. These findings may be applied for further optimization of the studied compound series. ![]()
Collapse
Affiliation(s)
- Agnieszka A Kaczor
- 1Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modelling Lab, Faculty of Pharmacy with Division of Medical Analytics, Medical University of Lublin, 4A Chodźki St., 20093 Lublin, Poland.,2School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, P.O. Box 1627, 70211 Kuopio, Finland
| | - Justyna Żuk
- 1Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modelling Lab, Faculty of Pharmacy with Division of Medical Analytics, Medical University of Lublin, 4A Chodźki St., 20093 Lublin, Poland
| | - Dariusz Matosiuk
- 1Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modelling Lab, Faculty of Pharmacy with Division of Medical Analytics, Medical University of Lublin, 4A Chodźki St., 20093 Lublin, Poland
| |
Collapse
|
27
|
Stoit AR, Lange JH, Coolen HK, Rensink A, van den Hoogenband A, den Hartog AP, van Schaik S, Kruse CG. Spiro-1-benzofuranpiperidinylalkanoic acids as a novel and selective sphingosine S1P5 receptor agonist chemotype. Bioorg Med Chem Lett 2018; 28:459-465. [DOI: 10.1016/j.bmcl.2017.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/08/2017] [Indexed: 10/18/2022]
|
28
|
The molecular basis of subtype selectivity of human kinin G-protein-coupled receptors. Nat Chem Biol 2018; 14:284-290. [PMID: 29334381 DOI: 10.1038/nchembio.2551] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 11/21/2017] [Indexed: 12/16/2022]
Abstract
G-protein-coupled receptors (GPCRs) are the most important signal transducers in higher eukaryotes. Despite considerable progress, the molecular basis of subtype-specific ligand selectivity, especially for peptide receptors, remains unknown. Here, by integrating DNP-enhanced solid-state NMR spectroscopy with advanced molecular modeling and docking, the mechanism of the subtype selectivity of human bradykinin receptors for their peptide agonists has been resolved. The conserved middle segments of the bound peptides show distinct conformations that result in different presentations of their N and C termini toward their receptors. Analysis of the peptide-receptor interfaces reveals that the charged N-terminal residues of the peptides are mainly selected through electrostatic interactions, whereas the C-terminal segments are recognized via both conformations and interactions. The detailed molecular picture obtained by this approach opens a new gateway for exploring the complex conformational and chemical space of peptides and peptide analogs for designing GPCR subtype-selective biochemical tools and drugs.
Collapse
|
29
|
Vass M, Kooistra AJ, Verhoeven S, Gloriam D, de Esch IJP, de Graaf C. A Structural Framework for GPCR Chemogenomics: What's In a Residue Number? Methods Mol Biol 2018; 1705:73-113. [PMID: 29188559 DOI: 10.1007/978-1-4939-7465-8_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The recent surge of crystal structures of G protein-coupled receptors (GPCRs), as well as comprehensive collections of sequence, structural, ligand bioactivity, and mutation data, has enabled the development of integrated chemogenomics workflows for this important target family. This chapter will focus on cross-family and cross-class studies of GPCRs that have pinpointed the need for, and the implementation of, a generic numbering scheme for referring to specific structural elements of GPCRs. Sequence- and structure-based numbering schemes for different receptor classes will be introduced and the remaining caveats will be discussed. The use of these numbering schemes has facilitated many chemogenomics studies such as consensus binding site definition, binding site comparison, ligand repurposing (e.g. for orphan receptors), sequence-based pharmacophore generation for homology modeling or virtual screening, and class-wide chemogenomics studies of GPCRs.
Collapse
Affiliation(s)
- Márton Vass
- Department of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV, Amsterdam, The Netherlands
| | - Albert J Kooistra
- Department of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV, Amsterdam, The Netherlands
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Stefan Verhoeven
- Netherlands eScience Center, 1098 XG, Amsterdam, The Netherlands
| | - David Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Iwan J P de Esch
- Department of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV, Amsterdam, The Netherlands
| | - Chris de Graaf
- Department of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
30
|
Vasile S, Esguerra M, Jespers W, Oliveira A, Sallander J, Åqvist J, Gutiérrez-de-Terán H. Characterization of Ligand Binding to GPCRs Through Computational Methods. Methods Mol Biol 2018; 1705:23-44. [PMID: 29188557 DOI: 10.1007/978-1-4939-7465-8_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The recent increase in available G protein-coupled receptor structures now contributes decisively to the structure-based ligand design. In this context, computational approaches in combination with medicinal chemistry and pharmacology are extremely helpful. Here, we provide an update on our structure-based computational protocols, used to answer key questions related to GPCR-ligand binding. All combined, these techniques can shed light on ligand binding modes, determine the molecular basis of conformational selection, for agonists and antagonists, as well as of subtype selectivity. To illustrate each of these questions, we will consider examples from existing projects on three families of class A (rhodopsin-like) GPCRs: one small-molecule (nucleotide-like) family, i.e., the adenosine receptors, and two peptide-binding receptors: neuropeptide-Y and angiotensin II receptors. The successful application of the same computational protocols to investigate this diverse group of receptor families gives an idea of the general applicability of our methodology in the characterization of GPCR-ligand binding.
Collapse
Affiliation(s)
- Silvana Vasile
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Center, Box 596, Uppsala, SE-751 24, Sweden
| | - Mauricio Esguerra
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Center, Box 596, Uppsala, SE-751 24, Sweden
| | - Willem Jespers
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Center, Box 596, Uppsala, SE-751 24, Sweden
| | - Ana Oliveira
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Center, Box 596, Uppsala, SE-751 24, Sweden
| | - Jessica Sallander
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Center, Box 596, Uppsala, SE-751 24, Sweden
| | - Johan Åqvist
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Center, Box 596, Uppsala, SE-751 24, Sweden
| | - Hugo Gutiérrez-de-Terán
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Center, Box 596, Uppsala, SE-751 24, Sweden.
| |
Collapse
|
31
|
Abstract
Despite tremendous efforts, approximately 120 GPCRs remain orphan. Their physiological functions and their potential roles in diseases are poorly understood. Orphan GPCRs are extremely important because they may provide novel therapeutic targets for unmet medical needs. As a complement to experimental approaches, molecular modeling and virtual screening are efficient techniques to discover synthetic surrogate ligands which can help to elucidate the role of oGPCRs. Constitutively activated mutants and recently published active structures of GPCRs provide stimulating opportunities for building active molecular models for oGPCRs and identifying activators using virtual screening of compound libraries. We describe the molecular modeling and virtual screening process we have applied in the discovery of surrogate ligands, and provide examples for CCKA, a simulated oGPCR, and for two oGPCRs, GPR52 and GPR34.
Collapse
Affiliation(s)
- Constantino Diaz
- Research Informatics, Evotec (France) SAS, 195 Route d'Espagne, 31036, Toulouse, France.
| | | | - Emilie Pihan
- Research Informatics, Evotec (France) SAS, 195 Route d'Espagne, 31036, Toulouse, France
| |
Collapse
|
32
|
Rodrigues DA, Pinheiro PDSM, Ferreira TTDSC, Thota S, Fraga CAM. Structural basis for the agonist action at free fatty acid receptor 1 (FFA1R or GPR40). Chem Biol Drug Des 2017; 91:668-680. [DOI: 10.1111/cbdd.13131] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 09/02/2017] [Accepted: 10/14/2017] [Indexed: 01/24/2023]
Affiliation(s)
- Daniel Alencar Rodrigues
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio); Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
- Programa de Pós-Graduação em Química; Instituto de Química; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | - Pedro de Sena Murteira Pinheiro
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio); Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal; Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | - Thayssa Tavares da Silva Cunha Ferreira
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio); Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal; Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | - Sreekanth Thota
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio); Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN); Center for Technological Development in Health (CDTS); Fundação Oswaldo Cruz - Ministério da Saúde; Rio de Janeiro RJ Brazil
| | - Carlos Alberto Manssour Fraga
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio); Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
- Programa de Pós-Graduação em Química; Instituto de Química; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal; Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| |
Collapse
|
33
|
Zhang C, Shao YM, Ma X, Cheong SL, Qin C, Tao L, Zhang P, Chen S, Zeng X, Liu H, Pastorin G, Jiang Y, Chen YZ. Pharmacological relationships and ligand discovery of G protein-coupled receptors revealed by simultaneous ligand and receptor clustering. J Mol Graph Model 2017; 76:136-142. [PMID: 28728042 DOI: 10.1016/j.jmgm.2017.06.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 06/17/2017] [Accepted: 06/19/2017] [Indexed: 12/18/2022]
Abstract
Conventional ligand and receptor similarity methods have been extensively used for exposing pharmacological relationships and drug lead discovery. They may in some cases neglect minor relationships useful for target hopping particularly against the remote family members. To complement the conventional methods for capturing these minor relationships, we developed a new method that uses a SLARC (Simultaneous Ligand And Receptor Clustering) 2D map to simultaneously characterize the ligand structural and receptor binding-site sequence relationships of a receptor family. The SLARC maps of the rhodopsin-like GPCR family comprehensively revealed scaffold hopping, target hopping, and multi-target relationships for the ligands of both homologous and remote family members. Their usefulness in new ligand discovery was validated by guiding the prospective discovery of novel indole piperazinylpyrimidine dual-targeting adenosine A2A receptor antagonist and dopamine D2 agonist compounds. The SLARC approach is useful for revealing pharmacological relationships and discovering new ligands at target family levels.
Collapse
Affiliation(s)
- Cheng Zhang
- Ministry-Province Jointly Constructed Base for State Key Lab and Shenzhen Technology and Engineering Lab for Personalized Cancer Diagnostics and Therapeutics, Tsinghua University Shenzhen Graduate School, and Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen 518055, PR China; Department of Molecular Pharmacology and Experimental Therapeutics, Center for Individualized Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Yi-Ming Shao
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Xiaohua Ma
- School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore
| | - Siew Lee Cheong
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Chu Qin
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Lin Tao
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Peng Zhang
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Shangying Chen
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Xian Zeng
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Hongxia Liu
- Ministry-Province Jointly Constructed Base for State Key Lab and Shenzhen Technology and Engineering Lab for Personalized Cancer Diagnostics and Therapeutics, Tsinghua University Shenzhen Graduate School, and Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen 518055, PR China
| | - Giorgia Pastorin
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School for Integrative Sciences and Engineering, 117456, Singapore.
| | - Yuyang Jiang
- Ministry-Province Jointly Constructed Base for State Key Lab and Shenzhen Technology and Engineering Lab for Personalized Cancer Diagnostics and Therapeutics, Tsinghua University Shenzhen Graduate School, and Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen 518055, PR China.
| | - Yu Zong Chen
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore; NUS Graduate School for Integrative Sciences and Engineering, 117456, Singapore.
| |
Collapse
|
34
|
Structure and Function of Peptide-Binding G Protein-Coupled Receptors. J Mol Biol 2017; 429:2726-2745. [PMID: 28705763 DOI: 10.1016/j.jmb.2017.06.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors and are important human drug targets. Of the 826 human GPCRs, 118 of them recognize endogenous peptide or protein ligands, and 30 of the 118 are targeted by approved drug molecules, including the very high-profile class B glucagon-like peptide 1 receptor. In this review, we analyze the 21 experimentally determined three-dimensional structures of the known peptide-binding GPCRs in relation to the endogenous peptides and drug molecules that modulate their cell signaling processes. Our integrated analyses reveal that half of the marketed drugs and most of the drugs in clinical trials that interact with peptide GPCRs are small molecules with a wide range of binding modes distinct from those of large peptide ligands. As we continue to collect additional data on these receptors from orthogonal approaches, including nuclear magnetic resonance and electron microscopy, we are beginning to understand how these receptors interact with their ligands at the molecular level and how improving the pharmacology of GPCR signal transduction requires us to study these receptors using multiple biophysical techniques.
Collapse
|
35
|
Lin F, Li F, Wang C, Wang J, Yang Y, Yang L, Li Y. Mechanism Exploration of Arylpiperazine Derivatives Targeting the 5-HT 2A Receptor by In Silico Methods. Molecules 2017; 22:molecules22071064. [PMID: 28672848 PMCID: PMC6152085 DOI: 10.3390/molecules22071064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 11/16/2022] Open
Abstract
As a G-protein coupled receptor, the 5-hydroxytryptamine 2A (5-HT2A) receptor is known for its critical role in the cognitive, behavioural and physiological functions, and thus is a primary molecular target to treat psychiatric diseases, including especially depression. With purpose to explore the structural traits affecting the inhibitory activity, currently a dataset of 109 arylpiperazine derivatives as promising 5-HT2A antagonists was built, based on which the ligand-based three-dimensional quantitative structure-activity relationship (3D-QSAR) study by using both comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA) approaches was carried out. The resultant optimal CoMSIA model displays proper validity and predictability with cross-validated correlation coefficient Q² = 0.587, non-cross-validated correlation coefficient R²ncv = 0.900 and predicted correlation coefficient for the test set of compounds R²pre = 0.897, respectively. Besides, molecular docking was also conducted to investigate the binding mode between these ligands and the active site of the 5-HT2A receptor. Meanwhile, as a docking supplementary tool to study the antagonists' conformation in the binding cavity, molecular dynamics (MD) simulation was also performed, providing further elucidation about the changes in the ligand-receptor complex. Lastly, some new molecules were also newly-designed based on the above results that are potential arylpiperazine antagonists of 5-HT2A receptor. We hope that the present models and derived information may be of help for facilitating the optimization and design of novel potent antagonists as antidepressant drugs as well as exploring the interaction mechanism of 5-HT2A antagonists.
Collapse
Affiliation(s)
- Feng Lin
- Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Pharmacy School, Shihezi University, Shihezi 832002, Xinjiang, China.
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian 116024, Liaoning, China.
| | - Feng Li
- Department of Civil Engineering, Henan Institute of Engineering, Zhengzhou 451191, Henan, China.
| | - Chao Wang
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian 116024, Liaoning, China.
| | - Jinghui Wang
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian 116024, Liaoning, China.
| | - Yinfeng Yang
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian 116024, Liaoning, China.
| | - Ling Yang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yan Li
- Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Pharmacy School, Shihezi University, Shihezi 832002, Xinjiang, China.
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian 116024, Liaoning, China.
| |
Collapse
|
36
|
Development and validation of a high-throughput calcium mobilization assay for the orphan receptor GPR88. J Biomed Sci 2017; 24:23. [PMID: 28347302 PMCID: PMC5369193 DOI: 10.1186/s12929-017-0330-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/20/2017] [Indexed: 12/02/2022] Open
Abstract
Background GPR88 is an orphan G protein-coupled receptor highly expressed in the striatum and is implicated in basal ganglia-associated disorders. However, the receptor functions of GPR88 are still largely unknown due to the lack of potent and selective ligands appropriate for central nervous system investigation. Development of a high-throughput screening assay for GPR88 should facilitate the discovery of novel ligands to probe GPR88 functions. Methods In this paper, we describe the development of a CHO-Gαqi5-GPR88 cell-based calcium mobilization assay. The assay takes advantage of functional coupling of GPR88 with the promiscuous Gαqi5 protein and consequent mobilization of intracellular calcium, which can be measured in a 384-well format with a Fluorescent Imaging Plate Reader. Results The CHO-Gαqi5-GPR88 cell-based calcium mobilization assay was validated by the structure-activity relationship study of known GPR88 agonist (1R,2R)-2-PCCA analogues. The assay was automated and miniaturized to a 384-well format, and was deemed robust and reproducible with a Z’-factor of 0.72 and tolerated dimethyl sulfoxide to a final concentration of 2%. Screening a pilot neurotransmitter library consisting of 228 compounds yielded 10 hits, but none of the hits were confirmed as GPR88 agonists in follow-up assays. Conclusions We have developed a high-throughput calcium mobilization assay for the orphan receptor GPR88. This calcium mobilization assay can be used to identify several different types of GPR88 ligands including agonists, competitive and noncompetitive antagonists, inverse agonists, and allosteric modulators. These ligands will serve as valuable tools to probe signaling mechanisms and in vivo functions of GPR88, and could expedite development of novel therapies for diseases potentially mediated by GPR88. Electronic supplementary material The online version of this article (doi:10.1186/s12929-017-0330-3) contains supplementary material, which is available to authorized users.
Collapse
|
37
|
Abstract
Drug discovery is a multidisciplinary and multivariate optimization endeavor. As such, in silico screening tools have gained considerable importance to archive, analyze and exploit the vast and ever-increasing amount of experimental data generated throughout the process. The current review will focus on the computer-aided prediction of the numerous properties that need to be controlled during the discovery of a preliminary hit and its promotion to a viable clinical candidate. It does not pretend to the almost impossible task of an exhaustive report but will highlight a few key points that need to be collectively addressed both by chemists and biologists to fuel the drug discovery pipeline with innovative and safe drug candidates.
Collapse
Affiliation(s)
- Didier Rognan
- Laboratoire d'Innovation Thérapeutique, UMR 7200 CNRS-Université de Strasbourg, 74 route du Rhin, 67400 Illkirch, France.
| |
Collapse
|
38
|
Sakai T, Shiraishi A, Kawada T, Matsubara S, Aoyama M, Satake H. Invertebrate Gonadotropin-Releasing Hormone-Related Peptides and Their Receptors: An Update. Front Endocrinol (Lausanne) 2017; 8:217. [PMID: 28932208 PMCID: PMC5592718 DOI: 10.3389/fendo.2017.00217] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/14/2017] [Indexed: 12/16/2022] Open
Abstract
Gonadotropin-releasing hormones (GnRHs) play pivotal roles in reproductive functions via the hypothalamus, pituitary, and gonad axis, namely, HPG axis in vertebrates. GnRHs and their receptors (GnRHRs) are likely to be conserved in invertebrate deuterostomes and lophotrochozoans. All vertebrate and urochordate GnRHs are composed of 10 amino acids, whereas protostome, echinoderm, and amphioxus GnRH-like peptides are 11- or 12-residue peptide containing two amino acids after an N-terminal pyro-Glu. In urochordates, Halocynthia roretzi GnRH gene encodes two GnRH peptide sequences, whereas two GnRH genes encode three different GnRH peptides in Ciona intestinalis. These findings indicate the species-specific diversification of GnRHs. Intriguingly, the major signaling pathway for GnRHRs is intracellular Ca2+ mobilization in chordates, echinoderms, and protostomes, whereas Ciona GnRHRs (Ci-GnRHRs) are endowed with multiple GnRHergic cAMP production pathways in a ligand-selective manner. Moreover, the ligand-specific modulation of signal transduction via heterodimerization among Ci-GnRHR paralogs suggests the species-specific development of fine-tuning of gonadal functions in ascidians. Echinoderm GnRH-like peptides show high sequence differences compared to those of protostome counterparts, leading to the difficulty in classification of peptides and receptors. These findings also show both the diversity and conservation of GnRH signaling systems in invertebrates. The lack of the HPG axis in invertebrates indicates that biological functions of GnRHs are not release of gonadotropins in current invertebrates and common ancestors of vertebrates and invertebrates. To date, authentic or putative GnRHRs have been characterized from various echinoderms and protostomes as well as chordates and the mRNAs have been found to be distributed not only reproductive organs but also other tissues. Collectively, these findings further support the notion that invertebrate GnRHs have biological roles other than the regulation of reproductive functions. Moreover, recent molecular phylogenetic analysis suggests that adipokinetic hormone (AKH), corazonin (CRZ), and AKH/CRZ-related peptide (ACP) belong to the GnRH superfamily but has led to the different classifications of these peptides and receptors using different datasets including the number of sequences and structural domains. In this review, we provide current knowledge of, and perspectives in, molecular basis and evolutionary aspects of the GnRH, AKH, CRZ, and ACP.
Collapse
Affiliation(s)
- Tsubasa Sakai
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Akira Shiraishi
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Tsuyoshi Kawada
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Shin Matsubara
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Masato Aoyama
- Faculty of Science, Department of Biological Sciences, Nara Women’s University, Nara, Japan
| | - Honoo Satake
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
- *Correspondence: Honoo Satake,
| |
Collapse
|
39
|
Orphan receptor ligand discovery by pickpocketing pharmacological neighbors. Nat Chem Biol 2016; 13:235-242. [PMID: 27992882 DOI: 10.1038/nchembio.2266] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 10/11/2016] [Indexed: 12/20/2022]
Abstract
Understanding the pharmacological similarity of G protein-coupled receptors (GPCRs) is paramount for predicting ligand off-target effects, drug repurposing, and ligand discovery for orphan receptors. Phylogenetic relationships do not always correctly capture pharmacological similarity. Previous family-wide attempts to define pharmacological relationships were based on three-dimensional structures and/or known receptor-ligand pairings, both unavailable for orphan GPCRs. Here, we present GPCR-CoINPocket, a novel contact-informed neighboring pocket metric of GPCR binding-site similarity that is informed by patterns of ligand-residue interactions observed in crystallographically characterized GPCRs. GPCR-CoINPocket is applicable to receptors with unknown structure or ligands and accurately captures known pharmacological relationships between GPCRs, even those undetected by phylogeny. When applied to orphan receptor GPR37L1, GPCR-CoINPocket identified its pharmacological neighbors, and transfer of their pharmacology aided in discovery of the first surrogate ligands for this orphan with a 30% success rate. Although primarily designed for GPCRs, the method is easily transferable to other protein families.
Collapse
|
40
|
Kuhne S, Kooistra AJ, Bosma R, Bortolato A, Wijtmans M, Vischer HF, Mason JS, de Graaf C, de Esch IJP, Leurs R. Identification of Ligand Binding Hot Spots of the Histamine H1 Receptor following Structure-Based Fragment Optimization. J Med Chem 2016; 59:9047-9061. [DOI: 10.1021/acs.jmedchem.6b00981] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Sebastiaan Kuhne
- Amsterdam
Institute for Molecules, Medicines and Systems (AIMMS), Division of
Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Albert J. Kooistra
- Amsterdam
Institute for Molecules, Medicines and Systems (AIMMS), Division of
Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Reggie Bosma
- Amsterdam
Institute for Molecules, Medicines and Systems (AIMMS), Division of
Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Andrea Bortolato
- Heptares Therapeutics Ltd., BioPark,
Broadwater Road, Welwyn Garden City, Herts AL7 3AX, U.K
| | - Maikel Wijtmans
- Amsterdam
Institute for Molecules, Medicines and Systems (AIMMS), Division of
Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Henry F. Vischer
- Amsterdam
Institute for Molecules, Medicines and Systems (AIMMS), Division of
Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Jonathan S. Mason
- Heptares Therapeutics Ltd., BioPark,
Broadwater Road, Welwyn Garden City, Herts AL7 3AX, U.K
| | - Chris de Graaf
- Amsterdam
Institute for Molecules, Medicines and Systems (AIMMS), Division of
Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Iwan J. P. de Esch
- Amsterdam
Institute for Molecules, Medicines and Systems (AIMMS), Division of
Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Rob Leurs
- Amsterdam
Institute for Molecules, Medicines and Systems (AIMMS), Division of
Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
41
|
Kleinau G, Müller A, Biebermann H. Oligomerization of GPCRs involved in endocrine regulation. J Mol Endocrinol 2016; 57:R59-80. [PMID: 27151573 DOI: 10.1530/jme-16-0049] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 05/04/2016] [Indexed: 12/27/2022]
Abstract
More than 800 different human membrane-spanning G-protein-coupled receptors (GPCRs) serve as signal transducers at biological barriers. These receptors are activated by a wide variety of ligands such as peptides, ions and hormones, and are able to activate a diverse set of intracellular signaling pathways. GPCRs are of central importance in endocrine regulation, which underpins the significance of comprehensively studying these receptors and interrelated systems. During the last decade, the capacity for multimerization of GPCRs was found to be a common and functionally relevant property. The interaction between GPCR monomers results in higher order complexes such as homomers (identical receptor subtype) or heteromers (different receptor subtypes), which may be present in a specific and dynamic monomer/oligomer equilibrium. It is widely accepted that the oligomerization of GPCRs is a mechanism for determining the fine-tuning and expansion of cellular processes by modification of ligand action, expression levels, and related signaling outcome. Accordingly, oligomerization provides exciting opportunities to optimize pharmacological treatment with respect to receptor target and tissue selectivity or for the development of diagnostic tools. On the other hand, GPCR heteromerization may be a potential reason for the undesired side effects of pharmacological interventions, faced with numerous and common mutual signaling modifications in heteromeric constellations. Finally, detailed deciphering of the physiological occurrence and relevance of specific GPCR/GPCR-ligand interactions poses a future challenge. This review will tackle the aspects of GPCR oligomerization with specific emphasis on family A GPCRs involved in endocrine regulation, whereby only a subset of these receptors will be discussed in detail.
Collapse
Affiliation(s)
- Gunnar Kleinau
- Institute of Experimental Pediatric Endocrinology (IEPE)Charité-Universitätsmedizin, Berlin, Germany
| | - Anne Müller
- Institute of Experimental Pediatric Endocrinology (IEPE)Charité-Universitätsmedizin, Berlin, Germany
| | - Heike Biebermann
- Institute of Experimental Pediatric Endocrinology (IEPE)Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
42
|
Sharma K, Ahuja G, Hussain A, Balfanz S, Baumann A, Korsching SI. Elimination of a ligand gating site generates a supersensitive olfactory receptor. Sci Rep 2016; 6:28359. [PMID: 27323929 PMCID: PMC4914996 DOI: 10.1038/srep28359] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/01/2016] [Indexed: 11/18/2022] Open
Abstract
Olfaction poses one of the most complex ligand-receptor matching problems in biology due to the unparalleled multitude of odor molecules facing a large number of cognate olfactory receptors. We have recently deorphanized an olfactory receptor, TAAR13c, as a specific receptor for the death-associated odor cadaverine. Here we have modeled the cadaverine/TAAR13c interaction, exchanged predicted binding residues by site-directed mutagenesis, and measured the activity of the mutant receptors. Unexpectedly we observed a binding site for cadaverine at the external surface of the receptor, in addition to an internal binding site, whose mutation resulted in complete loss of activity. In stark contrast, elimination of the external binding site generated supersensitive receptors. Modeling suggests this site to act as a gate, limiting access of the ligand to the internal binding site and thereby downregulating the affinity of the native receptor. This constitutes a novel mechanism to fine-tune physiological sensitivity to socially relevant odors.
Collapse
Affiliation(s)
- Kanika Sharma
- Institute of Genetics, Biocenter, University at Cologne, Zülpicherstrasse 47a, 50674 Cologne, Germany
| | - Gaurav Ahuja
- Institute of Genetics, Biocenter, University at Cologne, Zülpicherstrasse 47a, 50674 Cologne, Germany
| | - Ashiq Hussain
- Institute of Genetics, Biocenter, University at Cologne, Zülpicherstrasse 47a, 50674 Cologne, Germany
| | - Sabine Balfanz
- Institute of Complex Systems (ICS-4), Research Center Jülich, 52428 Jülich, Germany
| | - Arnd Baumann
- Institute of Complex Systems (ICS-4), Research Center Jülich, 52428 Jülich, Germany
| | - Sigrun I Korsching
- Institute of Genetics, Biocenter, University at Cologne, Zülpicherstrasse 47a, 50674 Cologne, Germany
| |
Collapse
|
43
|
Narayanan S, Maitra R, Deschamps JR, Bortoff K, Thomas JB, Zhang Y, Warner K, Vasukuttan V, Decker A, Runyon SP. Discovery of a novel small molecule agonist scaffold for the APJ receptor. Bioorg Med Chem 2016; 24:3758-70. [PMID: 27369451 DOI: 10.1016/j.bmc.2016.06.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 11/30/2022]
Abstract
The apelinergic system includes a series of endogenous peptides apelin, ELABELA/TODDLER and their 7-transmembrane G-protein coupled apelin receptor (APJ, AGTRL-1, APLNR). The APJ receptor is an attractive therapeutic target because of its involvement in cardiovascular diseases and potentially other disorders including liver fibrosis, obesity, diabetes, and neuroprotection. To date, pharmacological characterization of the APJ receptor has been limited due to the lack of small molecule functional agonists or antagonists. Through focused screening we identified a drug-like small molecule agonist hit 1 with a functional EC50 value of 21.5±5μM and binding affinity (Ki) of 5.2±0.5μM. Initial structure-activity studies afforded compound 22 having a 27-fold enhancement in potency and the first sub-micromolar full agonist with an EC50 value of 800±0.1nM and Ki of 1.3±0.3μM. Preliminary SAR, synthetic methodology, and in vitro pharmacological characterization indicate this scaffold will serve as a favorable starting point for further refinement of APJ potency and selectivity.
Collapse
Affiliation(s)
- Sanju Narayanan
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, United States
| | - Rangan Maitra
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, United States
| | - Jeffery R Deschamps
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, Code 6930, 4555 Overlook Avenue SW, Washington, DC 20375, United States
| | - Katherine Bortoff
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, United States
| | - James B Thomas
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, United States
| | - Yanyan Zhang
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, United States
| | - Keith Warner
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, United States
| | - Vineetha Vasukuttan
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, United States
| | - Ann Decker
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, United States
| | - Scott P Runyon
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, United States.
| |
Collapse
|
44
|
Kleist AB, Getschman AE, Ziarek JJ, Nevins AM, Gauthier PA, Chevigné A, Szpakowska M, Volkman BF. New paradigms in chemokine receptor signal transduction: Moving beyond the two-site model. Biochem Pharmacol 2016; 114:53-68. [PMID: 27106080 DOI: 10.1016/j.bcp.2016.04.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/13/2016] [Indexed: 10/21/2022]
Abstract
Chemokine receptor (CKR) signaling forms the basis of essential immune cellular functions, and dysregulated CKR signaling underpins numerous disease processes of the immune system and beyond. CKRs, which belong to the seven transmembrane domain receptor (7TMR) superfamily, initiate signaling upon binding of endogenous, secreted chemokine ligands. Chemokine-CKR interactions are traditionally described by a two-step/two-site mechanism, in which the CKR N-terminus recognizes the chemokine globular core (i.e. site 1 interaction), followed by activation when the unstructured chemokine N-terminus is inserted into the receptor TM bundle (i.e. site 2 interaction). Several recent studies challenge the structural independence of sites 1 and 2 by demonstrating physical and allosteric links between these supposedly separate sites. Others contest the functional independence of these sites, identifying nuanced roles for site 1 and other interactions in CKR activation. These developments emerge within a rapidly changing landscape in which CKR signaling is influenced by receptor PTMs, chemokine and CKR dimerization, and endogenous non-chemokine ligands. Simultaneous advances in the structural and functional characterization of 7TMR biased signaling have altered how we understand promiscuous chemokine-CKR interactions. In this review, we explore new paradigms in CKR signal transduction by considering studies that depict a more intricate architecture governing the consequences of chemokine-CKR interactions.
Collapse
Affiliation(s)
- Andrew B Kleist
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Anthony E Getschman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Joshua J Ziarek
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Ave, Boston, MA 02115, USA.
| | - Amanda M Nevins
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Pierre-Arnaud Gauthier
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg.
| | - Andy Chevigné
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg.
| | - Martyna Szpakowska
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg.
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
45
|
Massart R, Mignon V, Stanic J, Munoz-Tello P, Becker JAJ, Kieffer BL, Darmon M, Sokoloff P, Diaz J. Developmental and adult expression patterns of the G-protein-coupled receptor GPR88 in the rat: Establishment of a dual nuclear-cytoplasmic localization. J Comp Neurol 2016; 524:2776-802. [PMID: 26918661 DOI: 10.1002/cne.23991] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 01/31/2023]
Abstract
GPR88 is a neuronal cerebral orphan G-protein-coupled receptor (GPCR) that has been linked to various psychiatric disorders. However, no extensive description of its localization has been provided so far. Here, we investigate the spatiotemporal expression of the GPR88 in prenatal and postnatal rat tissues by using in situ hybridization and immunohistochemistry. GPR88 protein was initially detected at embryonic day 16 (E16) in the striatal primordium. From E16-E20 to adulthood, the highest expression levels of both protein and mRNA were observed in striatum, olfactory tubercle, nucleus accumbens, amygdala, and neocortex, whereas in spinal cord, pons, and medulla GPR88 expression remains discrete. We observed an intracellular redistribution of GPR88 during cortical lamination. In the cortical plate of the developing cortex, GPR88 presents a classical GPCR plasma membrane/cytoplasmic localization that shifts, on the day of birth, to nuclei of neurons progressively settling in layers V to II. This intranuclear localization remains throughout adulthood and was also detected in monkey and human cortex as well as in the amygdala and hypothalamus of rats. Apart from the central nervous system, GPR88 was transiently expressed at high levels in peripheral tissues, including adrenal cortex (E16-E21) and cochlear ganglia (E19-P3), and also at moderate levels in retina (E18-E19) and spleen (E21-P7). The description of the GPR88 anatomical expression pattern may provide precious functional insights into this novel receptor. Furthermore, the GRP88 nuclear localization suggests nonclassical GPCR modes of action of the protein that could be relevant for cortical development and psychiatric disorders. J. Comp. Neurol. 524:2776-2802, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Renaud Massart
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France.,Neurology-Psychiatry Department, Pierre Fabre Research Institute, 81100, Castres, France
| | - Virginie Mignon
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France
| | - Jennifer Stanic
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France
| | - Paola Munoz-Tello
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France
| | - Jerôme A J Becker
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, INSERM, 67400, Illkirch-Graffenstaden, France
| | - Brigitte L Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, INSERM, 67400, Illkirch-Graffenstaden, France
| | - Michèle Darmon
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France
| | - Pierre Sokoloff
- Neurology-Psychiatry Department, Pierre Fabre Research Institute, 81100, Castres, France
| | - Jorge Diaz
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France
| |
Collapse
|
46
|
Auvynet C, Baudesson de Chanville C, Hermand P, Dorgham K, Piesse C, Pouchy C, Carlier L, Poupel L, Barthélémy S, Felouzis V, Lacombe C, Sagan S, Chemtob S, Quiniou C, Salomon B, Deterre P, Sennlaub F, Combadière C. ECL1i, d(LGTFLKC), a novel, small peptide that specifically inhibits CCL2-dependent migration. FASEB J 2016; 30:2370-81. [PMID: 26979087 DOI: 10.1096/fj.201500116] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/25/2016] [Indexed: 11/11/2022]
Abstract
CC chemokine receptor type 2 (CCR2) is a key molecule in inflammatory diseases and is an obvious drug target for the treatment of inflammation. A number of nonpeptidic, competitive CCR2 antagonists have been developed, but none has yet been approved for clinical use. Our aim was to identify a short peptide that showed allosteric antagonism against human and mouse CCR2. On the basis of sequence analysis and 3-dimensional modeling, we identified an original 7-d-amino acid peptidic CCR2 inhibitor that we have called extracellular loop 1 inverso (ECL1i), d(LGTFLKC). In vitro, ECL1i selectively and potently inhibits CC chemokine ligand type 2 (CCL2)-triggered chemotaxis (IC50, 2 µM) but no other conventional CCL2-associated events. We used the classic competitive CCR2 antagonist, BMS22 {2-[(isopropylaminocarbonyl)amino]-N-[2-[[cis-2-[[4-(methylthio)benzoyl]amino]cyclohexyl]amino]-2-oxoethyl]-5-(trifluoromethyl)benzamide}, as positive control and inhibited CCL2-dependent chemotaxis with an IC50 of 18 nM. As negative control, we used a peptide with the same composition as ECL1i, but in a different sequence, d(FKLTLCG). In vivo, ECL1i (4 mg/kg) interfered with CCR2-positive cell recruitment and attenuated disease progression in experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis. This study establishes ECL1i as the first allosteric inhibitor of CCR2 with functional selectivity. ECL1i is a promising new agent in therapeutic development, and it may, by its selective effect, increase our understanding of CCR2 signaling pathways and functions.-Auvynet, C., Baudesson de Chanville, C., Hermand, P., Dorgham, K., Piesse, C., Pouchy, C., Carlier, L., Poupel, L., Barthélémy, S., Felouzis, V., Lacombe, C., Sagan, S., Salomon, B., Deterre, P., Sennlaub, F., Combadière, C. ECL1i, d(LGTFLKC), a novel, small peptide that specifically inhibits CCL2-dependent migration.
Collapse
Affiliation(s)
- Constance Auvynet
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Camille Baudesson de Chanville
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Patricia Hermand
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Karim Dorgham
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Christophe Piesse
- Sorbonne Universités, UPMC/Univ Paris 06, Institut de Biologie Paris-Seine (IBPS) 3631, CNRS, Service de Synthése Peptidique, Paris, France
| | - Charlotte Pouchy
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Ludovic Carlier
- Sorbonne Universités, UPMC/Univ Paris 06, CNRS, UMR 7203, Laboratoire des Biomolécules, Paris, France
| | - Lucie Poupel
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Sandrine Barthélémy
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Virginie Felouzis
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Claire Lacombe
- Sorbonne Universités, UPMC/Univ Paris 06, CNRS, UMR 7203, Laboratoire des Biomolécules, Paris, France; Ecole Normale Supérieure-Université de Recherche Paris Sciences et Lettres, Département de Chimie, Paris, France; Faculté des Sciences et Technologie, Université Paris Est Créteil-Val de Marne, Créteil, France
| | - Sandrine Sagan
- Sorbonne Universités, UPMC/Univ Paris 06, CNRS, UMR 7203, Laboratoire des Biomolécules, Paris, France
| | | | | | - Benoit Salomon
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Philippe Deterre
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Florian Sennlaub
- Sorbonne Universités, UPMC/ Univ Paris 06, UMRS 968, INSERM, U968, Institut de la Vision, Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-Direction des Hôpitaux et de l'Offre de Soins (DHOS), Centre d'Investigation Clinique 503, Paris, France
| | - Christophe Combadière
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France;
| |
Collapse
|
47
|
Li Q, Tachie-Baffour Y, Liu Z, Baldwin MW, Kruse AC, Liberles SD. Non-classical amine recognition evolved in a large clade of olfactory receptors. eLife 2015; 4:e10441. [PMID: 26519734 PMCID: PMC4695389 DOI: 10.7554/elife.10441] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/28/2015] [Indexed: 11/13/2022] Open
Abstract
Biogenic amines are important signaling molecules, and the structural basis for their recognition by G Protein-Coupled Receptors (GPCRs) is well understood. Amines are also potent odors, with some activating olfactory trace amine-associated receptors (TAARs). Here, we report that teleost TAARs evolved a new way to recognize amines in a non-classical orientation. Chemical screens de-orphaned eleven zebrafish TAARs, with agonists including serotonin, histamine, tryptamine, 2-phenylethylamine, putrescine, and agmatine. Receptors from different clades contact ligands through aspartates on transmembrane α-helices III (canonical Asp3.32) or V (non-canonical Asp5.42), and diamine receptors contain both aspartates. Non-classical monoamine recognition evolved in two steps: an ancestral TAAR acquired Asp5.42, gaining diamine sensitivity, and subsequently lost Asp3.32. Through this transformation, the fish olfactory system dramatically expanded its capacity to detect amines, ecologically significant aquatic odors. The evolution of a second, alternative solution for amine detection by olfactory receptors highlights the tremendous structural versatility intrinsic to GPCRs. DOI:http://dx.doi.org/10.7554/eLife.10441.001 Many organisms make molecules called biogenic amines. These molecules, which include the human hormones adrenaline and histamine, have important roles in regulating the biology and behaviour of many animals. Some biogenic amines bind to receptor proteins called GPCRs on the surface of cells. Many drugs can affect the activity of GPCRs, so understanding how different GPCRs work is an important goal of the pharmaceutical industry. Like all proteins, GPCRs are made of chains of molecules called amino acids. The GPCRs that can detect biogenic amines use a particular amino acid named Asp3.32, and when this amino acid is mutated, these GPCRs become unable to bind to their target amine. Trace amine-associated receptors (TAARs) are a type of GPCR that are found in many animals to detect odors. Most TAARs in mammals contain the Asp3.32 residue, and recognize amine odors. However, many fish TAARs do not contain Asp3.32, and it was not clear what molecules these fish receptors detect. Here Li et al. find that these fish TAARs also recognize amines, and use a different amino acid called Asp5.42. Also, some TAARs contain both Asp3.32 and Asp5.42, and recognize chemicals with two amines named diamines. Some diamines that bind to TAARs are foul smelling odors; for example, cadaverine and putrescine are repulsive smells emitted by decomposing flesh. In total, the experiments identified amines that can bind to eleven zebrafish TAARs that previously had no odor partner. Li et al. propose that some fish TAARs lost the Asp3.32 during the course of evolution to leave the Asp5.42 as the main interaction site for amines. This change dramatically altered how these TAARs interact with amines, which probably expanded the number of different amines that fish can detect. These findings open up new ways to study how the fish brain processes information about its surroundings. DOI:http://dx.doi.org/10.7554/eLife.10441.002
Collapse
Affiliation(s)
- Qian Li
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Yaw Tachie-Baffour
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Zhikai Liu
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Maude W Baldwin
- Department of Organismic and Evolutionary Biology, Museum of Comparative Zoology, Harvard University, Cambridge, United States
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, United States
| | - Stephen D Liberles
- Department of Cell Biology, Harvard Medical School, Boston, United States
| |
Collapse
|
48
|
Jang JW, Cho NC, Min SJ, Cho YS, Park KD, Seo SH, No KT, Pae AN. Novel Scaffold Identification of mGlu1 Receptor Negative Allosteric Modulators Using a Hierarchical Virtual Screening Approach. Chem Biol Drug Des 2015; 87:239-56. [DOI: 10.1111/cbdd.12654] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/15/2015] [Accepted: 08/18/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Jae Wan Jang
- Center for Neuro-Medicine; Brain Science Institute; Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 5 Seongbuk-gu, Seoul 136-791 Korea
- Department of Biological Chemistry; School of Science; Korea University of Science and Technology; 52 Eoeun dong Yuseong-gu, Daejeon 305-333 Korea
| | - Nam-Chul Cho
- Center for Neuro-Medicine; Brain Science Institute; Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 5 Seongbuk-gu, Seoul 136-791 Korea
- Department of Biotechnology; Yonsei University; Seodaemun-gu, Seoul 120-749 Korea
| | - Sun-Joon Min
- Department of Applied Chemistry; Hanyang University; Ansan, Gyeonggi-do 15588 Korea
| | - Yong Seo Cho
- Center for Neuro-Medicine; Brain Science Institute; Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 5 Seongbuk-gu, Seoul 136-791 Korea
- Department of Biological Chemistry; School of Science; Korea University of Science and Technology; 52 Eoeun dong Yuseong-gu, Daejeon 305-333 Korea
| | - Ki Duk Park
- Center for Neuro-Medicine; Brain Science Institute; Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 5 Seongbuk-gu, Seoul 136-791 Korea
- Department of Biological Chemistry; School of Science; Korea University of Science and Technology; 52 Eoeun dong Yuseong-gu, Daejeon 305-333 Korea
| | - Seon Hee Seo
- Center for Neuro-Medicine; Brain Science Institute; Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 5 Seongbuk-gu, Seoul 136-791 Korea
| | - Kyoung Tai No
- Department of Biotechnology; Yonsei University; Seodaemun-gu, Seoul 120-749 Korea
| | - Ae Nim Pae
- Center for Neuro-Medicine; Brain Science Institute; Korea Institute of Science and Technology (KIST); Hwarangno 14-gil 5 Seongbuk-gu, Seoul 136-791 Korea
- Department of Biological Chemistry; School of Science; Korea University of Science and Technology; 52 Eoeun dong Yuseong-gu, Daejeon 305-333 Korea
| |
Collapse
|
49
|
Weichert D, Gmeiner P. Covalent molecular probes for class A G protein-coupled receptors: advances and applications. ACS Chem Biol 2015; 10:1376-86. [PMID: 25860503 DOI: 10.1021/acschembio.5b00070] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Covalent modification of G protein-coupled receptors (GPCRs) by employing specific molecular probes has for decades provided a successful strategy to facilitate the elucidation of the structure and function of this pharmacologically important class of membrane proteins. The ligands typically comprise a pharmacophore that generates affinity for a given GPCR and contain a reactive functionality that may form a covalent bond with a suitably positioned amino acid residue. Covalent ligands have been successfully applied to circumvent poor affinity of compounds when stable labeling of receptor populations was required, and they have been used in the isolation, purification, and pharmacological characterization of specific subtypes of GPCRs. Recently, structural studies have demonstrated that covalent molecular probes are effective at stabilizing GPCRs to obtain X-ray crystal structures, thus providing valuable insights for the development of novel therapeutics. Herein, we review covalently binding molecular probes for class A GPCRs with a focus on ligands comprising cross-linking groups that do not require photoactivation and further highlight their significant and diverse applications.
Collapse
Affiliation(s)
- Dietmar Weichert
- Department
of Chemistry and
Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Peter Gmeiner
- Department
of Chemistry and
Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| |
Collapse
|
50
|
Planesas JM, Pérez-Nueno VI, Borrell JI, Teixidó J. Studying the binding interactions of allosteric agonists and antagonists of the CXCR4 receptor. J Mol Graph Model 2015; 60:1-14. [PMID: 26080355 DOI: 10.1016/j.jmgm.2015.05.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 05/06/2015] [Accepted: 05/07/2015] [Indexed: 12/01/2022]
Abstract
Several examples of allosteric modulators of GPCRs have been reported recently in the literature, but understanding their molecular mechanism presents a new challenge for medicinal chemistry. For the specific case of the cellular receptor CXCR4, it is known that pepducins (lipidated fragments of intracellular GPCR loops) such as ATI-2341 modulate CXCR4 activity agonistically via an allosteric mechanism. Moreover, there are also examples of small organic molecules such as AMD11070 and GSK812397 which may also act as allosteric antagonists. However, incomplete knowledge of the ligand-binding sites has hampered a detailed molecular understanding of how these inhibitors work. Here, we attempt to answer this question by analysing the binding interactions between the CXCR4 receptor and the above-mentioned allosteric modulators. We propose two different allosteric binding sites, one located in the intracellular loops 1, 2 and 3 (ICL1, ICL2 and ICL3) which binds the pepducin agonist ATI-2341, and the other at a subsite of the main extracellular orthosteric binding pocket between extracellular loops 1 and 2 and the N-terminus, which binds the antagonists AMD11070 and GSK812397. Allosteric interactions between the CXCR4 and ATI-2341 were predicted by combining different modeling approaches. First, a rotational blind docking search was applied and the best poses were subsequently refined using flexible docking methods and molecular dynamic simulations. For the AMD11070 and GSK812397 antagonists, the entire CXCR4 protein surface was explored by blind docking in order to define the binding region. A second docking analysis by subsites was then performed to refine the allosteric interactions. Finally, we identified the binding residues that appear to be essential for CXCR4 allosteric modulators.
Collapse
Affiliation(s)
- Jesús M Planesas
- Grup d'Enginyeria Molecular, Institut Químic de Sarriá (IQS), Universitat Ramon Llull, Barcelona, Spain
| | - Violeta I Pérez-Nueno
- Grup d'Enginyeria Molecular, Institut Químic de Sarriá (IQS), Universitat Ramon Llull, Barcelona, Spain; Harmonic Pharma, Espace Transfert, 615 rue du Jardin Botanique, 54600 Villers lès Nancy, France.
| | - José I Borrell
- Grup d'Enginyeria Molecular, Institut Químic de Sarriá (IQS), Universitat Ramon Llull, Barcelona, Spain
| | - Jordi Teixidó
- Grup d'Enginyeria Molecular, Institut Químic de Sarriá (IQS), Universitat Ramon Llull, Barcelona, Spain.
| |
Collapse
|