Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Res ; 32(8): 761-772, 2022 08.
Article in English | MEDLINE | ID: mdl-35739238

ABSTRACT

Somatostatin receptors (SSTRs) play versatile roles in inhibiting the secretion of multiple hormones such as growth hormone and thyroid-stimulating hormone, and thus are considered as targets for treating multiple tumors. Despite great progress made in therapeutic development against this diverse receptor family, drugs that target SSTRs still show limited efficacy with preferential binding affinity and conspicuous side-effects. Here, we report five structures of SSTR2 and SSTR4 in different states, including two crystal structures of SSTR2 in complex with a selective peptide antagonist and a non-peptide agonist, respectively, a cryo-electron microscopy (cryo-EM) structure of Gi1-bound SSTR2 in the presence of the endogenous ligand SST-14, as well as two cryo-EM structures of Gi1-bound SSTR4 in complex with SST-14 and a small-molecule agonist J-2156, respectively. By comparison of the SSTR structures in different states, molecular mechanisms of agonism and antagonism were illustrated. Together with computational and functional analyses, the key determinants responsible for ligand recognition and selectivity of different SSTR subtypes and multiform binding modes of peptide and non-peptide ligands were identified. Insights gained in this study will help uncover ligand selectivity of various SSTRs and accelerate the development of new molecules with better efficacy by targeting SSTRs.


Subject(s)
Neoplasms , Receptors, Somatostatin , Cryoelectron Microscopy , Humans , Ligands , Neoplasms/metabolism , Receptors, Somatostatin/agonists , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Somatostatin/pharmacology , Somatostatin/therapeutic use
2.
Biochem J ; 479(3): 385-399, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35084016

ABSTRACT

The apelin receptor (APLNR) regulates many biological processes including metabolism, angiogenesis, circulating blood volume and cardiovascular function. Additionally, APLNR is overexpressed in various types of cancer and influences cancer progression. APLNR is reported to regulate tumor recognition during immune surveillance by modulating the IFN-γ response. However, the mechanism of APLNR cross-talk with intratumoral IFN-γ signaling remains unknown. Here, we show that activation of APLNR up-regulates IFN-γ signaling in melanoma cells through APLNR mediated ß-arrestin 1 but not ß-arrestin 2 recruitment. Our data suggests that ß-arrestin 1 directly interacts with STAT1 to inhibit STAT1 phosphorylation to attenuate IFN-γ signaling. The APLNR mutant receptor, I109A, which is deficient in ß-arrestins recruitment, is unable to enhance intratumoral IFN-γ signaling. While APLNR N112G, a constitutively active mutant receptor, increases intratumoral sensitivity to IFN-γ signaling by enhancing STAT1 phosphorylation upon IFN-γ exposure. We also demonstrate in a co-culture system that APLNR regulates tumor survival rate. Taken together, our findings reveal that APLNR modulates IFN-γ signaling in melanoma cells and suggest that APLNR may be a potential target to enhance the efficacy of immunotherapy.


Subject(s)
Apelin Receptors/physiology , Interferon-gamma/physiology , Janus Kinases/physiology , Melanoma/metabolism , Neoplasm Proteins/physiology , STAT1 Transcription Factor/physiology , Signal Transduction/physiology , beta-Arrestin 1/physiology , Apelin Receptors/antagonists & inhibitors , Apelin Receptors/chemistry , Apelin Receptors/genetics , Cell Line, Tumor , Cytotoxicity, Immunologic , HEK293 Cells , Humans , Janus Kinases/antagonists & inhibitors , Melanoma/immunology , Models, Molecular , Protein Interaction Mapping , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , T-Lymphocytes/immunology , beta-Arrestin 2/analysis
3.
Theranostics ; 11(4): 1901-1917, 2021.
Article in English | MEDLINE | ID: mdl-33408788

ABSTRACT

Rationale: Fc engineering has become the focus of antibody drug development. The current mutagenesis and in silico protein design methods are confined by the limited throughput and high cost, while the high-throughput phage display and yeast display technologies are not suitable for screening glycosylated Fc variants. Here we developed a mammalian cell display-based Fc engineering platform. Methods: By using mammalian cell display and next generation sequencing, we screened millions of Fc variants for optimized affinity and specificity for FcγRIIIa or FcγRIIb. The identified Fc variants with improved binding to FcγRIIIa were substituted into trastuzumab and rituximab and the effector function of antibodies were examined in the PBMC-based assay. On the other hand, the identified Fc variants with selectively enhanced FcγRIIb binding were applied to CD40 agonist antibody and the activities of the antibodies were measured on different cell assays. The immunostimulatory activity of CD40 antibodies was also evaluated by OVA-specific CD8+ T cell response model in FcγR/CD40-humanized mice. Results: Using this approach, we screened millions of Fc variant and successfully identified several novel Fc variants with enhanced FcγRIIIa or FcγRIIb binding. These identified Fc variants displayed a dramatic increase in antibody-dependent cellular cytotoxicity in PBMC-based assay. Novel variants with selectively enhanced FcγRIIb binding were also identified. CD40 agonist antibodies substituted with these Fc variants displayed activity more potent than the parental antibody in the in vitro and in vivo models.Conclusions: This approach increased the throughput of Fc variant screening from thousands to millions magnitude, enabled screening variants containing multiple mutations and could be integrated with glycoengineering technology, represents an ideal platform for Fc engineering. The initial efforts demonstrated the capability of the platform and the novel Fc variants could be substituted into nearly any antibody for the next generation of antibody therapeutics.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Breast Neoplasms/drug therapy , Immunoglobulin Fc Fragments/immunology , Leukocytes, Mononuclear/immunology , Receptors, IgG/metabolism , Trastuzumab/pharmacology , Animals , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Leukocytes, Mononuclear/drug effects , Mice , Receptors, IgG/immunology , Tumor Cells, Cultured
4.
Biochem J ; 477(17): 3313-3327, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32779693

ABSTRACT

Apelin receptor (APJ) is a G protein-coupled receptor that contributes to many physiological processes and is emerging as a therapeutic target to treat a variety of diseases. For most disease indications the role of G protein vs ß-arrestin signalling in mitigating disease pathophysiology remains poorly understood. This hinders the development of G protein biased APJ agonists, which have been proposed to have several advantages over balanced APJ signalling agonists. To elucidate the contribution of APJ ß-arrestin signalling, we generated a transgenic mouse harbouring a point mutation (APJ I107A) that maintains full G protein activity but fails to recruit ß-arrestin following receptor activation. APJ I107A mutant mice did not alter cardiac function at rest, following exercise challenge or in response to pressure overload induced cardiac hypertrophy. Additionally, APJ I107A mice have comparable body weights, plasma glucose and lipid levels relative to WT mice when fed a chow diet. However, APJ I107A mice showed significantly lower body weight, blood insulin levels, improved glucose tolerance and greater insulin sensitivity when fed a high-fat diet. Furthermore, loss of APJ ß-arrestin signalling also affected fat composition and the expression of lipid metabolism related genes in adipose tissue from high-fat fed mice. Taken together, our results suggest that G protein biased APJ activation may be more effective for certain disease indications given that loss of APJ mediated ß-arrestin signalling appears to mitigate several aspects of diet induced metabolic dysfunction.


Subject(s)
Adipose Tissue/metabolism , Apelin Receptors/deficiency , Diet, High-Fat/adverse effects , Metabolic Diseases/metabolism , Myocardium/metabolism , Point Mutation , Signal Transduction , beta-Arrestins/metabolism , Adipose Tissue/pathology , Amino Acid Substitution , Animals , Apelin Receptors/metabolism , HEK293 Cells , Humans , Metabolic Diseases/chemically induced , Metabolic Diseases/genetics , Metabolic Diseases/pathology , Mice , Myocardium/pathology , beta-Arrestins/genetics
5.
Commun Biol ; 3(1): 146, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32218528

ABSTRACT

Hybridoma and phage display are two powerful technologies for isolating target-specific monoclonal antibodies based on the binding. However, for complex membrane proteins, such as G protein-coupled receptors (GPCRs), binding-based screening rarely results in functional antibodies. Here we describe a function-based high-throughput screening method for quickly identifying antibody antagonists and agonists against GPCRs by combining glycosylphosphatidylinositol-anchored antibody cell display with ß-arrestin recruitment-based cell sorting and screening. This method links antibody genotype with phenotype and is applicable to all GPCR targets. We validated this method by identifying a panel of antibody antagonists and an antibody agonist to the human apelin receptor from an immune antibody repertoire. In contrast, we obtained only neutral binders and antibody antagonists from the same repertoire by phage display, suggesting that the new approach described here is more efficient than traditional methods in isolating functional antibodies. This new method may create a new paradigm in antibody drug discovery.


Subject(s)
Antibodies/pharmacology , Apelin Receptors/agonists , Apelin Receptors/antagonists & inhibitors , Drug Discovery , High-Throughput Screening Assays , Animals , Apelin Receptors/genetics , Apelin Receptors/metabolism , CHO Cells , Cell Line, Tumor , Cell Surface Display Techniques , Cricetulus , Flow Cytometry , Genes, Reporter , HEK293 Cells , Humans , Hybridomas , Proof of Concept Study , Signal Transduction , beta-Arrestins/genetics , beta-Arrestins/metabolism
6.
JCI Insight ; 5(8)2020 04 23.
Article in English | MEDLINE | ID: mdl-32208384

ABSTRACT

Heart failure (HF) remains a grievous illness with poor prognosis even with optimal care. The apelin receptor (APJ) counteracts the pressor effect of angiotensin II, attenuates ischemic injury, and has the potential to be a novel target to treat HF. Intravenous administration of apelin improves cardiac function acutely in patients with HF. However, its short half-life restricts its use to infusion therapy. To identify a longer acting APJ agonist, we conducted a medicinal chemistry campaign, leading to the discovery of potent small-molecule APJ agonists with comparable activity to apelin by mimicking the C-terminal portion of apelin-13. Acute infusion increased systolic function and reduced systemic vascular resistance in 2 rat models of impaired cardiac function. Similar results were obtained in an anesthetized but not a conscious canine HF model. Chronic oral dosing in a rat myocardial infarction model reduced myocardial collagen content and improved diastolic function to a similar extent as losartan, a RAS antagonist standard-of-care therapy, but lacked additivity with coadministration. Collectively, this work demonstrates the feasibility of developing clinical, viable, potent small-molecule agonists that mimic the endogenous APJ ligand with more favorable drug-like properties and highlights potential limitations for APJ agonism for this indication.


Subject(s)
Apelin Receptors/agonists , Heart/drug effects , Animals , Dogs , Drug Discovery , Heart Failure , Intercellular Signaling Peptides and Proteins , Rats
7.
Cell Res ; 30(5): 455, 2020 May.
Article in English | MEDLINE | ID: mdl-32203135

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Cell Res ; 30(5): 436-445, 2020 05.
Article in English | MEDLINE | ID: mdl-32047270

ABSTRACT

The pituitary adenylate cyclase-activating polypeptide type I receptor (PAC1R) belongs to the secretin receptor family and is widely distributed in the central neural system and peripheral organs. Abnormal activation of the receptor mediates trigeminovascular activation and sensitization, which is highly related to migraine, making PAC1R a potential therapeutic target. Elucidation of PAC1R activation mechanism would benefit discovery of therapeutic drugs for neuronal disorders. PAC1R activity is governed by pituitary adenylate cyclase-activating polypeptide (PACAP), known as a major vasodilator neuropeptide, and maxadilan, a native peptide from the sand fly, which is also capable of activating the receptor with similar potency. These peptide ligands have divergent sequences yet initiate convergent PAC1R activity. It is of interest to understand the mechanism of PAC1R ligand recognition and receptor activity regulation through structural biology. Here we report two near-atomic resolution cryo-EM structures of PAC1R activated by PACAP38 or maxadilan, providing structural insights into two distinct ligand binding modes. The structures illustrate flexibility of the extracellular domain (ECD) for ligands with distinct conformations, where ECD accommodates ligands in different orientations while extracellular loop 1 (ECL1) protrudes to further anchor the ligand bound in the orthosteric site. By structure-guided molecular modeling and mutagenesis, we tested residues in the ligand-binding pockets and identified clusters of residues that are critical for receptor activity. The structures reported here for the first time elucidate the mechanism of specificity and flexibility of ligand recognition and binding for PAC1R, and provide insights toward the design of therapeutic molecules targeting PAC1R.


Subject(s)
Insect Proteins/metabolism , Models, Molecular , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , Cell Line , Cryoelectron Microscopy , Humans , Ligands , Migraine Disorders/metabolism , Protein Binding , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/antagonists & inhibitors , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism
9.
Sci Adv ; 6(3): eaax7379, 2020 01.
Article in English | MEDLINE | ID: mdl-31998837

ABSTRACT

Developing antibody agonists targeting the human apelin receptor (APJ) is a promising therapeutic approach for the treatment of chronic heart failure. Here, we report the structure-guided discovery of a single-domain antibody (sdAb) agonist JN241-9, based on the cocrystal structure of APJ with an sdAb antagonist JN241, the first cocrystal structure of a class A G protein-coupled receptor (GPCR) with a functional antibody. As revealed by the structure, JN241 binds to the extracellular side of APJ, makes critical contacts with the second extracellular loop, and inserts the CDR3 into the ligand-binding pocket. We converted JN241 into a full agonist JN241-9 by inserting a tyrosine into the CDR3. Modeling and molecular dynamics simulation shed light on JN241-9-stimulated receptor activation, providing structural insights for finding agonistic antibodies against class A GPCRs.


Subject(s)
Apelin Receptors/agonists , Apelin Receptors/chemistry , Drug Discovery/methods , Quantitative Structure-Activity Relationship , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/pharmacology , Animals , Binding Sites , Drug Design , Humans , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding
10.
J Chem Inf Model ; 60(9): 4339-4349, 2020 09 28.
Article in English | MEDLINE | ID: mdl-31652060

ABSTRACT

G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors, which is arguably the most important family of drug target. With the technology breakthroughs in X-ray crystallography and cryo-electron microscopy, more than 300 GPCR-ligand complex structures have been publicly reported since 2007, covering about 60 unique GPCRs. Such abundant structural information certainly will facilitate the structure-based drug design by targeting GPCRs. In this study, we have developed a fragment-based computational method for designing novel GPCR ligands. We first extracted the characteristic interaction patterns (CIPs) on the binding interfaces between GPCRs and their ligands. The CIPs were used as queries to search the chemical fragments derived from GPCR ligands, which were required to form similar interaction patterns with GPCR. Then, the selected chemical fragments were assembled into complete molecules by using the AutoT&T2 software. In this work, we chose ß-adrenergic receptor (ß-AR) and muscarinic acetylcholine receptor (mAChR) as the targets to validate this method. Based on the designs suggested by our method, samples of 63 compounds were purchased and tested in a cell-based functional assay. A total of 15 and 22 compounds were identified as active antagonists for ß2-AR and mAChR M1, respectively. Molecular dynamics simulations and binding free energy analysis were performed to explore the key interactions (e.g., hydrogen bonds and π-π interactions) between those active compounds and their target GPCRs. In summary, our work presents a useful approach to the de novo design of GPCR ligands based on the relevant 3D structural information.


Subject(s)
Receptors, G-Protein-Coupled , Signal Transduction , Cryoelectron Microscopy , Crystallography, X-Ray , Ligands , Receptors, Adrenergic, beta-2
11.
Cell Rep ; 29(10): 2936-2943.e4, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31801061

ABSTRACT

Adrenergic G-protein-coupled receptors (GPCRs) mediate different cellular signaling pathways in the presence of endogenous catecholamines and play important roles in both physiological and pathological conditions. Extensive studies have been carried out to investigate the structure and function of ß adrenergic receptors (ßARs). However, the structure of α adrenergic receptors (αARs) remains to be determined. Here, we report the structure of the human α2C adrenergic receptor (α2CAR) with the non-selective antagonist, RS79948, at 2.8 Å. Our structure, mutations, modeling, and functional experiments indicate that a α2CAR-specific D206ECL2-R409ECL3-Y4056.58 network plays a role in determining α2 adrenergic subtype selectivity. Furthermore, our results show that a specific loosened helix at the top of TM4 in α2CAR is involved in receptor activation. Together, our structure of human α2CAR-RS79948 provides key insight into the mechanism underlying the α2 adrenergic receptor activation and subtype selectivity.


Subject(s)
Receptors, Adrenergic, alpha-2/metabolism , Animals , CHO Cells , Cell Line , Cricetulus , HEK293 Cells , Humans , Isoquinolines/pharmacology , Ligands , Naphthyridines/pharmacology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
12.
Biochem J ; 475(23): 3813-3826, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30409826

ABSTRACT

Biased ligands of G protein-coupled receptors (GPCRs) may have improved therapeutic benefits and safety profiles. However, the molecular mechanism of GPCR biased signaling remains largely unknown. Using apelin receptor (APJ) as a model, we systematically investigated the potential effects of amino acid residues around the orthosteric binding site on biased signaling. We discovered that a single residue mutation I109A (I1093.32) in the transmembrane domain 3 (TM3) located in the deep ligand-binding pocket was sufficient to convert a balanced APJ into a G protein signaling biased receptor. APJ I109A mutant receptor retained full capabilities in ligand binding and G protein activation, but was defective in GRK recruitment, ß-arrestin recruitment, and downstream receptor-mediated ERK activation. Based on molecular dynamics simulations, we proposed a molecular mechanism for biased signaling of I109A mutant receptor. We postulate that due to the extra space created by I109A mutation, the phenyl group of the last residue (Phe-13) of apelin rotates down and initiates a cascade of conformational changes in TM3. Phe-13 formed a new cluster of hydrophobic interactions with the sidechains of residues in TM3, including F1103.33 and M1133.36, which stabilizes the mutant receptor in a conformation favoring biased signaling. Interruption of these stabilizing interactions by double mutation F110A/I109A or M113A/I109A largely restored the ß-arrestin-mediated signaling. Taken together, we describe herein the discovery of a biased APJ mutant receptor and provide detailed molecular insights into APJ signaling selectivity, facilitating the discovery of novel therapeutics targeting APJ.


Subject(s)
Amino Acids/chemistry , Apelin Receptors/chemistry , Protein Domains , Receptors, G-Protein-Coupled/chemistry , Amino Acid Sequence , Amino Acids/genetics , Amino Acids/metabolism , Apelin/chemistry , Apelin/metabolism , Apelin Receptors/genetics , Apelin Receptors/metabolism , Binding Sites/genetics , Cell Line, Tumor , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Ligands , Molecular Dynamics Simulation , Mutation, Missense , Protein Binding , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
13.
Structure ; 25(6): 858-866.e4, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28528775

ABSTRACT

Apelin receptor (APJR) is a key regulator of human cardiovascular function and is activated by two different endogenous peptide ligands, apelin and Elabela, each with different isoforms diversified by length and amino acid sequence. Here we report the 2.6-Å resolution crystal structure of human APJR in complex with a designed 17-amino-acid apelin mimetic peptide agonist. The structure reveals that the peptide agonist adopts a lactam constrained curved two-site ligand binding mode. Combined with mutation analysis and molecular dynamics simulations with apelin-13 binding to the wild-type APJR, this structure provides a mechanistic understanding of apelin recognition and binding specificity. Comparison of this structure with that of other peptide receptors suggests that endogenous peptide ligands with a high degree of conformational flexibility may bind and modulate the receptors via a similar two-site binding mechanism.


Subject(s)
Apelin Receptors/chemistry , Alanine , Apelin/chemistry , Apelin Receptors/agonists , Apelin Receptors/genetics , Crystallography, X-Ray , Humans , Molecular Dynamics Simulation , Molecular Mimicry , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptides, Cyclic/chemistry , Protein Conformation , Signal Transduction
14.
Mol Cancer Ther ; 15(6): 1227-37, 2016 06.
Article in English | MEDLINE | ID: mdl-27196749

ABSTRACT

Aberrant hepatocyte growth factor (HGF)/MET signaling has been implicated in hepatocarcinogenesis, suggesting that MET may serve as an attractive therapeutic target in hepatocellular carcinoma. We sought to investigate the in vitro and in vivo antitumor activity of AMG 337, a potent and highly selective small molecule MET kinase inhibitor, in preclinical models of hepatocellular carcinoma. The antiproliferative activity of AMG 337 was evaluated across a panel of hepatocellular carcinoma cell lines in a viability assay. Daily oral administration was used to evaluate the in vivo antitumor activity of AMG 337 in two patient-derived xenograft (PDX) models of hepatocellular carcinoma (LI0612 and LI1078). AMG 337 exerted potent antiproliferative activity against 2 of 40 hepatocellular carcinoma cell lines, namely, MHCC97H (IC50, 0.015 µmol/L) and HCCLM3 (IC50, 0.025 µmol/L). Both sensitive cell lines showed MET amplification (MET/CEN-7 >2.0) assessed by FISH, and high MET expression (3+ IHC) assessed by IHC. AMG 337 potently inhibited p-MET in all cell lines with detectable levels of total MET. However, the dose-dependent inhibition of downstream effectors of HGF/MET signaling, including p-GAB1, p-AKT, and p-ERK, was limited to those cell lines sensitive to AMG 337 in a viability assay (MHCC97H and HCCLM3). AMG 337 significantly inhibited tumor growth at all doses tested in the MET-amplified and MET-high-expressing hepatocellular carcinoma PDX model LI0612 and had no effect on tumor growth in the non-MET-amplified and MET-low-expressing hepatocellular carcinoma PDX model LI1078. AMG 337 represents a promising and novel therapeutic strategy for targeting hepatocellular carcinomas with a dependence on HGF/MET signaling. Mol Cancer Ther; 15(6); 1227-37. ©2016 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridones/administration & dosage , Small Molecule Libraries/administration & dosage , Triazoles/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Amplification , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/genetics , Mice , Proto-Oncogene Proteins c-met/genetics , Pyridones/pharmacology , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Triazoles/pharmacology , Xenograft Model Antitumor Assays
15.
Biochem J ; 424(1): 39-45, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19725810

ABSTRACT

TAARs (trace amine-associated receptors) are G-protein-coupled receptors that respond to low abundance, endogenous amines such as tyramine and tryptamine, and represent potential targets for neuropsychiatric diseases. However, some members of this receptor subfamily either have no ligand identified or remain difficult to express and characterize using recombinant systems. In the present paper we report the successful expression of human and mouse TAAR1, and the characterization of their responses to various natural and synthetic agonists. In HEK (human embryonic kidney)-293/CRE-bla cells, mouse TAAR1 showed a robust response to trace amines as measured using either a cAMP assay or a beta-lactamase reporter assay, whereas human TAAR1 showed a weaker, but still measurable, response. When certain fragments of human TAAR1 were replaced with the corresponding regions of mouse TAAR1, the chimaeric receptor showed a much stronger response in cAMP production. Examination of a series of agonists on these receptors revealed that the human and the chimaeric receptor are almost identical in pharmacology, but distinct from the mouse receptor. We also screened small libraries of pharmacologically active agents on TAAR1 and identified a series of synthetic agonists, some of which are also ligands of the enigmatic imidazoline receptor. The findings of the present study not only shed light on the pharmacological species difference of TAAR1, but also raise new possibilities about the mechanism of some of the imidazoline-related agents.


Subject(s)
Imidazoline Receptors/agonists , Imidazoline Receptors/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Animals , COS Cells , Cell Line , Central Nervous System Stimulants/pharmacology , Chlorocebus aethiops , Cyclic AMP/metabolism , Humans , Mice , Octopamine/pharmacology , Phenethylamines/pharmacology , Psychotropic Drugs/pharmacology , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synephrine/pharmacology , Tryptamines/pharmacology , Tyramine/pharmacology
16.
J Biomol Screen ; 14(7): 789-97, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19525486

ABSTRACT

GPR139 is an orphan G-protein-coupled receptor (GPCR) that is expressed nearly exclusively in the central nervous system and may play a role in the control of locomotor activity. The signal transduction pathway and pharmacological function of GPR139, however, are still controversial due to the lack of natural or synthetic ligands. The authors report the characterization of human GPR139 signaling pathway and identification of surrogate agonists and antagonists. In both transient and stable transfections of HEK293F cells, overexpression of GPR139 increased basal intracellular cAMP concentrations compared to control cells. Furthermore, forskolin and isoproterenol-stimulated cAMP responses were enhanced in GPR139-expressing cells, suggesting that GPR139 is predominantly coupled to Galpha(s). The authors screened a large library of small molecules for compounds that increase cAMP levels in GPR139-expressing cells and identified a compound with GPR139 agonist activity. This compound increased cAMP production specifically in cells expressing GPR139 but not in cells expressing its highly homologous receptor GPR142. Furthermore, this compound did not induce calcium mobilization in GPR139 cells, indicating no Galpha(q)-mediated response. In addition, antagonist screening with the identified agonist yielded 2 classes of compounds as antagonists. The identification of surrogate agonists and antagonists of human GPR139 provides important tools for further study of this orphan GPCR.


Subject(s)
Drug Evaluation, Preclinical/methods , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Cell Line , Clone Cells , Humans , Transfection
17.
Assay Drug Dev Technol ; 6(4): 543-50, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18699727

ABSTRACT

Fluorescence resonance energy transfer (FRET) has emerged as a powerful tool to the study of protein-protein interactions, such as receptor-ligand binding. However, the application of FRET to the study of G protein-coupled receptors (GPCRs) has been limited by the method of labeling receptor with fluorescence probes. Here we described a novel time-resolved (TR)-FRET method to study GPCR-ligand binding by using human complement 5a (C5a) receptor (C5aR) as a model system. Human C5aR was expressed in human embryonic kidney 293 cells with a hemagglutinin (HA) epitope at the N-terminus. Purified human C5a was labeled with terbium chelate and used as the fluorescence donor. Monoclonal anti-HA antibody conjugated with Alexa Fluor 488 was used as the fluorescence acceptor. Robust FRET signal was observed when the labeled ligand and C5aR membrane were mixed in the presence of the conjugated anti-HA antibody. This FRET signal was specific and saturable. C5a binding affinity to C5aR measured by the FRET assay was consistent with the data as determined by competition binding analysis using radiolabeled C5a. The FRET assay was also used to determine affinity of C5aR antagonists by competition binding analysis, and the data are similar to those from radioligand binding studies. Compared to the commonly used radioligand binding assay, this TR-FRET-based assay provides a nonradioactive, faster, and sensitive homogeneous assay format that could be easily adapted to high-throughput screening. The principle of this assay should also be applicable to other GPCRs, especially to those receptors with peptide or protein ligands.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Receptors, G-Protein-Coupled/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Chelating Agents/chemistry , Complement C5a/chemistry , Cyclic AMP/metabolism , Humans , Ligands , Radioligand Assay , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Terbium/chemistry
18.
Biochem J ; 376(Pt 3): 645-53, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-12956623

ABSTRACT

The Escherichia coli GABA (gamma-aminobutyric acid) permease GabP is a prototypical APC (amine/polyamine/choline) super-family transporter that has a CAR (consensus amphipathic region) containing multiple specificity determinants, ostensibly organized on two helical surfaces, one hydrophobic [SHS (sensitive hydrophobic surface)] and the other hydrophilic [SPS (sensitive polar surface)]. To gauge the functional effects of placing alanine insertions at close intervals across the entire GabP CAR, 64 insertion variants were constructed. Insertions, particularly those in the SHS and the SPS, were highly detrimental to steady-state [(3)H]GABA accumulation. TSR (transport specificity ratio) analysis, employing [(3)H]nipecotic acid and [(14)C]GABA, showed that certain alanine insertions were associated with a specificity shift (i.e. a change in k (cat)/ K (m)). An insertion (INS Ala-269) located N-terminal to the SHS increased specificity for [(3)H]nipecotic acid relative to [(14)C]GABA, whereas an insertion (INS Ala-321) located C-terminal to the SPS had the opposite effect. Overall, the results are consistent with a working hypothesis that the GabP CAR contains extensive functional surfaces that may be manipulated by insertion mutagenesis to alter the specificity ( k (cat)/ K (m)) phenotype. The thermodynamic basis of TSR analysis provides generality, suggesting that amino acid insertions could affect specificity in many other transporters, particularly those such as the E. coli phenylalanine permease PheP [Pi, Chow and Pittard (2002) J. Bacteriol. 184, 5842-5847] that have a functionally significant CAR-like domain.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membrane Transport Proteins , Organic Anion Transporters/chemistry , Organic Anion Transporters/metabolism , Alanine/genetics , Amino Acid Sequence , Biological Transport/drug effects , Carrier Proteins/genetics , Consensus Sequence , Escherichia coli Proteins/genetics , GABA Plasma Membrane Transport Proteins , Genes, Reporter , Hydrophobic and Hydrophilic Interactions , Immunoblotting , Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Insertional , Organic Anion Transporters/genetics , Protein Structure, Secondary , Substrate Specificity , beta-Galactosidase/genetics , beta-Galactosidase/metabolism , gamma-Aminobutyric Acid/metabolism
19.
Science ; 301(5638): 1391-4, 2003 Sep 05.
Article in English | MEDLINE | ID: mdl-12958364

ABSTRACT

Wnt proteins, regulators of development in many organisms, bind to seven transmembrane-spanning (7TMS) receptors called frizzleds, thereby recruiting the cytoplasmic molecule dishevelled (Dvl) to the plasma membrane.Frizzled-mediated endocytosis of Wg (a Drosophila Wnt protein) and lysosomal degradation may regulate the formation of morphogen gradients. Endocytosis of Frizzled 4 (Fz4) in human embryonic kidney 293 cells was dependent on added Wnt5A protein and was accomplished by the multifunctional adaptor protein beta-arrestin 2 (betaarr2), which was recruited to Fz4 by binding to phosphorylated Dvl2. These findings provide a previously unrecognized mechanism for receptor recruitment of beta-arrestin and demonstrate that Dvl plays an important role in the endocytosis of frizzled, as well as in promoting signaling.


Subject(s)
Arrestins/metabolism , Endocytosis , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Arrestins/genetics , Cell Line , Cell Membrane/metabolism , Clathrin/metabolism , Cytoplasm/metabolism , Dishevelled Proteins , Drosophila Proteins , Frizzled Receptors , Humans , Mice , Phosphoproteins/metabolism , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Proteins/genetics , Proto-Oncogene Proteins/pharmacology , RNA, Small Interfering , Recombinant Fusion Proteins/metabolism , Signal Transduction , Wnt Proteins , Wnt-5a Protein , beta-Arrestin 2 , beta-Arrestins
20.
J Biol Chem ; 278(28): 26295-301, 2003 Jul 11.
Article in English | MEDLINE | ID: mdl-12724327

ABSTRACT

Beta1-adrenergic receptors, expressed at high levels in the human heart, have a carboxyl-terminal ESKV motif that can directly interact with PDZ domain-containing proteins. Using the beta1-adrenergic receptor carboxyl terminus as bait, we identified the novel beta1-adrenergic receptor-binding partner GIPC in a yeast two-hybrid screen of a human heart cDNA library. Here we demonstrate that the PDZ domain-containing protein, GIPC, co-immunoprecipitates with the beta1-adrenergic receptor in COS-7 cells. Essential for this interaction is the Ser residue of the beta1-adrenergic receptor carboxyl-terminal ESKV motif. Our data also demonstrate that beta1-adrenergic receptor stimulation activates the mitogen-activated protein kinase, ERK1/2. beta1-adrenergic receptor-mediated ERK1/2 activation was inhibited by pertussis toxin, implicating Gi, and was substantially decreased by the expression of GIPC. Expression of GIPC had no observable effect on beta1-adrenergic receptor sequestration or receptor-mediated cAMP accumulation. This GIPC effect was specific for the beta1-adrenergic receptor and was dependent on an intact PDZ binding motif. These data suggest that GIPC can regulate beta1-adrenergic receptor-stimulated, Gi-mediated, ERK activation while having no effect on receptor internalization or Gs-mediated cAMP signaling.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation, Enzymologic , Mitogen-Activated Protein Kinases/metabolism , Neuropeptides/metabolism , Receptors, Adrenergic, beta-1/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Animals , COS Cells , Cell Line , Cyclic AMP/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Gene Library , Humans , Immunoblotting , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Myocardium/metabolism , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Serine/metabolism , Signal Transduction , Time Factors , Transfection , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...