Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Cell ; 165(7): 1632-1643, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27315480

ABSTRACT

Ligand-directed signal bias offers opportunities for sculpting molecular events, with the promise of better, safer therapeutics. Critical to the exploitation of signal bias is an understanding of the molecular events coupling ligand binding to intracellular signaling. Activation of class B G protein-coupled receptors is driven by interaction of the peptide N terminus with the receptor core. To understand how this drives signaling, we have used advanced analytical methods that enable separation of effects on pathway-specific signaling from those that modify agonist affinity and mapped the functional consequence of receptor modification onto three-dimensional models of a receptor-ligand complex. This yields molecular insights into the initiation of receptor activation and the mechanistic basis for biased agonism. Our data reveal that peptide agonists can engage different elements of the receptor extracellular face to achieve effector coupling and biased signaling providing a foundation for rational design of biased agonists.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/chemistry , Peptides/pharmacology , Venoms/pharmacology , Animals , CHO Cells , Calcium/metabolism , Cell Line , Cricetulus , Cyclic AMP/metabolism , Exenatide , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Oxyntomodulin/chemistry , Oxyntomodulin/metabolism , Peptides/chemistry , Rats , Signal Transduction , Venoms/chemistry
2.
Mol Cell ; 80(3): 485-500.e7, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33027691

ABSTRACT

Peptide drugs targeting class B1 G-protein-coupled receptors (GPCRs) can treat multiple diseases; however, there remains substantial interest in the development of orally delivered non-peptide drugs. Here, we reveal unexpected overlap between signaling and regulation of the glucagon-like peptide-1 (GLP-1) receptor by the non-peptide agonist PF 06882961 and GLP-1 that was not observed for another compound, CHU-128. Compounds from these patent series, including PF 06882961, are currently in clinical trials for treatment of type 2 diabetes. High-resolution cryoelectron microscopy (cryo-EM) structures reveal that the binding sites for PF 06882961 and GLP-1 substantially overlap, whereas CHU-128 adopts a unique binding mode with a more open receptor conformation at the extracellular face. Structural differences involving extensive water-mediated hydrogen bond networks could be correlated to functional data to understand how PF 06882961, but not CHU-128, can closely mimic the pharmacological properties of GLP-1. These findings will facilitate rational structure-based discovery of non-peptide agonists targeting class B GPCRs.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Animals , Binding Sites/physiology , Cryoelectron Microscopy/methods , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/chemistry , Humans , Peptides/chemistry , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
3.
PLoS Biol ; 22(7): e3002673, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39083706

ABSTRACT

Development of optimal therapeutics for disease states that can be associated with increased membrane cholesterol requires better molecular understanding of lipid modulation of the drug target. Type 1 cholecystokinin receptor (CCK1R) agonist actions are affected by increased membrane cholesterol, enhancing ligand binding and reducing calcium signaling, while agonist actions of the closely related CCK2R are not. In this work, we identified a set of chimeric human CCK1R/CCK2R mutations that exchange the cholesterol sensitivity of these 2 receptors, providing powerful tools when expressed in CHO and HEK-293 model cell lines to explore mechanisms. Static, low energy, high-resolution structures of the mutant CCK1R constructs, stabilized in complex with G protein, were not substantially different, suggesting that alterations to receptor dynamics were key to altered function. We reveal that cholesterol-dependent dynamic changes in the conformation of the helical bundle of CCK receptors affects both ligand binding at the extracellular surface and G protein coupling at the cytosolic surface, as well as their interrelationships involved in stimulus-response coupling. This provides an ideal setting for potential allosteric modulators to correct the negative impact of membrane cholesterol on CCK1R.


Subject(s)
Cholesterol , GTP-Binding Proteins , Protein Binding , Receptor, Cholecystokinin A , Receptor, Cholecystokinin B , Animals , Humans , CHO Cells , Cholesterol/metabolism , Cricetulus , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , HEK293 Cells , Ligands , Mutation , Protein Conformation , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin B/metabolism , Receptor, Cholecystokinin B/genetics
4.
Nature ; 577(7790): 432-436, 2020 01.
Article in English | MEDLINE | ID: mdl-31915381

ABSTRACT

Class B G-protein-coupled receptors are major targets for the treatment of chronic diseases, including diabetes and obesity1. Structures of active receptors reveal peptide agonists engage deep within the receptor core, leading to an outward movement of extracellular loop 3 and the tops of transmembrane helices 6 and 7, an inward movement of transmembrane helix 1, reorganization of extracellular loop 2 and outward movement of the intracellular side of transmembrane helix 6, resulting in G-protein interaction and activation2-6. Here we solved the structure of a non-peptide agonist, TT-OAD2, bound to the glucagon-like peptide-1 (GLP-1) receptor. Our structure identified an unpredicted non-peptide agonist-binding pocket in which reorganization of extracellular loop 3 and transmembrane helices 6 and 7 manifests independently of direct ligand interaction within the deep transmembrane domain pocket. TT-OAD2 exhibits biased agonism, and kinetics of G-protein activation and signalling that are distinct from peptide agonists. Within the structure, TT-OAD2 protrudes beyond the receptor core to interact with the lipid or detergent, providing an explanation for the distinct activation kinetics that may contribute to the clinical efficacy of this compound series. This work alters our understanding of the events that drive the activation of class B receptors.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Isoquinolines/pharmacology , Phenylalanine/analogs & derivatives , Pyridines/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Glucagon-Like Peptide-1 Receptor/chemistry , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Isoquinolines/chemistry , Kinetics , Models, Molecular , Phenylalanine/chemistry , Phenylalanine/pharmacology , Protein Structure, Quaternary , Protein Structure, Tertiary , Pyridines/chemistry , Structural Homology, Protein
5.
PLoS Biol ; 19(6): e3001295, 2021 06.
Article in English | MEDLINE | ID: mdl-34086670

ABSTRACT

G protein-coupled receptors (GPCRs) are critical regulators of cellular function acting via heterotrimeric G proteins as their primary transducers with individual GPCRs capable of pleiotropic coupling to multiple G proteins. Structural features governing G protein selectivity and promiscuity are currently unclear. Here, we used cryo-electron microscopy (cryo-EM) to determine structures of the cholecystokinin (CCK) type 1 receptor (CCK1R) bound to the CCK peptide agonist, CCK-8 and 2 distinct transducer proteins, its primary transducer Gq, and the more weakly coupled Gs. As seen with other Gq/11-GPCR complexes, the Gq-α5 helix (αH5) bound to a relatively narrow pocket in the CCK1R core. Surprisingly, the backbone of the CCK1R and volume of the G protein binding pocket were essentially equivalent when Gs was bound, with the Gs αH5 displaying a conformation that arises from "unwinding" of the far carboxyl-terminal residues, compared to canonically Gs coupled receptors. Thus, integrated changes in the conformations of both the receptor and G protein are likely to play critical roles in the promiscuous coupling of individual GPCRs.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/metabolism , Cholecystokinin/metabolism , Cholesterol/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/ultrastructure , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/ultrastructure , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Receptors, Cholecystokinin/ultrastructure , Signal Transduction
6.
Nature ; 558(7711): 559-563, 2018 06.
Article in English | MEDLINE | ID: mdl-29925945

ABSTRACT

The class A adenosine A1 receptor (A1R) is a G-protein-coupled receptor that preferentially couples to inhibitory Gi/o heterotrimeric G proteins, has been implicated in numerous diseases, yet remains poorly targeted. Here we report the 3.6 Å structure of the human A1R in complex with adenosine and heterotrimeric Gi2 protein determined by Volta phase plate cryo-electron microscopy. Compared to inactive A1R, there is contraction at the extracellular surface in the orthosteric binding site mediated via movement of transmembrane domains 1 and 2. At the intracellular surface, the G protein engages the A1R primarily via amino acids in the C terminus of the Gαi α5-helix, concomitant with a 10.5 Å outward movement of the A1R transmembrane domain 6. Comparison with the agonist-bound ß2 adrenergic receptor-Gs-protein complex reveals distinct orientations for each G-protein subtype upon engagement with its receptor. This active A1R structure provides molecular insights into receptor and G-protein selectivity.


Subject(s)
Adenosine/chemistry , Adenosine/metabolism , Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/ultrastructure , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A1/ultrastructure , Binding Sites , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Models, Molecular , Receptor, Adenosine A1/metabolism , Rotation , Substrate Specificity
7.
Nature ; 555(7694): 121-125, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29466332

ABSTRACT

The class B glucagon-like peptide-1 (GLP-1) G protein-coupled receptor is a major target for the treatment of type 2 diabetes and obesity. Endogenous and mimetic GLP-1 peptides exhibit biased agonism-a difference in functional selectivity-that may provide improved therapeutic outcomes. Here we describe the structure of the human GLP-1 receptor in complex with the G protein-biased peptide exendin-P5 and a Gαs heterotrimer, determined at a global resolution of 3.3 Å. At the extracellular surface, the organization of extracellular loop 3 and proximal transmembrane segments differs between our exendin-P5-bound structure and previous GLP-1-bound GLP-1 receptor structure. At the intracellular face, there was a six-degree difference in the angle of the Gαs-α5 helix engagement between structures, which was propagated across the G protein heterotrimer. In addition, the structures differed in the rate and extent of conformational reorganization of the Gαs protein. Our structure provides insights into the molecular basis of biased agonism.


Subject(s)
Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/ultrastructure , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/ultrastructure , Binding Sites , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/chemistry , Humans , Models, Molecular , Protein Conformation
8.
J Physiol ; 601(23): 5195-5211, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37772438

ABSTRACT

Agonists of dopamine D2 receptors (D2R), 5-hydroxytryptamine (5-HT, serotonin) receptors (5-HTR) and ghrelin receptors (GHSR) activate neurons in the lumbosacral defecation centre, and act as 'colokinetics', leading to increased propulsive colonic motility, in vivo. In the present study, we investigated which neurons in the lumbosacral defecation centre express the receptors and whether dopamine, serotonin and ghrelin receptor agonists act on the same lumbosacral preganglionic neurons (PGNs). We used whole cell electrophysiology to record responses from neurons in the lumbosacral defecation centre, following colokinetic application, and investigated their expression profiles and the chemistries of their neural inputs. Fluorescence in situ hybridisation revealed Drd2, Ghsr and Htr2C transcripts were colocalised in lumbosacral PGNs of mice, and immunohistochemistry showed that these neurons have closely associated tyrosine hydroxylase and 5-HT boutons. Previous studies showed that they do not receive ghrelin inputs. Whole cell electrophysiology in adult mice spinal cord revealed that dopamine, serotonin, α-methylserotonin and capromorelin each caused inward, excitatory currents in overlapping populations of lumbosacral PGNs. Furthermore, dopamine caused increased frequency of both IPSCs and EPSCs in a cohort of D2R neurons. Tetrodotoxin blocked the IPSCs and EPSCs, revealing a post-synaptic excitatory action of dopamine. In lumbosacral PGNs of postnatal day 7-14 rats, only dopamine's postsynaptic effects were observed. Furthermore, inward, excitatory currents evoked by dopamine were reduced by the GHSR antagonist, YIL781. We conclude that lumbosacral PGNs are the site where the action of endogenous ligands of D2R and 5-HT2R converge, and that GHSR act as a cis-modulator of D2R expressed by the same neurons. KEY POINTS: Dopamine, 5-hydroxytryptamine (5-HT, serotonin) and ghrelin (GHSR) receptor agonists increase colorectal motility and have been postulated to act at receptors on parasympathetic preganglionic neurons (PGNs) in the lumbosacral spinal cord. We aimed to determine which neurons in the lumbosacral spinal cord express dopamine, serotonin and GHSR receptors, their neural inputs, and whether agonists at these receptors excite them. We show that dopamine, serotonin and ghrelin receptor transcripts are contained in the same PGNs and that these neurons have closely associated tyrosine hydroxylase and serotonin boutons. Whole cell electrophysiology revealed that dopamine, serotonin and GHSR receptor agonists induce an inward excitatory current in overlapping populations of lumbosacral PGNs. Dopamine-induced excitation was reversed by GHSR antagonism. The present study demonstrates that lumbosacral PGNs are the site at which actions of endogenous ligands of dopamine D2 receptors and 5-HT type 2 receptors converge. Ghrelin receptors are functional, but their role appears to be as modulators of dopamine effects at D2 receptors.


Subject(s)
Dopamine , Serotonin , Humans , Rats , Animals , Mice , Dopamine/pharmacology , Serotonin/pharmacology , Receptors, Ghrelin , Rats, Sprague-Dawley , Rodentia , Defecation/physiology , Ghrelin/pharmacology , Tyrosine 3-Monooxygenase/pharmacology , Receptors, Serotonin , Receptors, Dopamine D2
9.
Nature ; 546(7656): 118-123, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28437792

ABSTRACT

Class B G-protein-coupled receptors are major targets for the treatment of chronic diseases, such as osteoporosis, diabetes and obesity. Here we report the structure of a full-length class B receptor, the calcitonin receptor, in complex with peptide ligand and heterotrimeric Gαsßγ protein determined by Volta phase-plate single-particle cryo-electron microscopy. The peptide agonist engages the receptor by binding to an extended hydrophobic pocket facilitated by the large outward movement of the extracellular ends of transmembrane helices 6 and 7. This conformation is accompanied by a 60° kink in helix 6 and a large outward movement of the intracellular end of this helix, opening the bundle to accommodate interactions with the α5-helix of Gαs. Also observed is an extended intracellular helix 8 that contributes to both receptor stability and functional G-protein coupling via an interaction with the Gß subunit. This structure provides a new framework for understanding G-protein-coupled receptor function.


Subject(s)
Cryoelectron Microscopy , Heterotrimeric GTP-Binding Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/ultrastructure , Receptors, Calcitonin/classification , Receptors, Calcitonin/ultrastructure , Binding Sites , Cell Membrane/metabolism , Conserved Sequence , Heterotrimeric GTP-Binding Proteins/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Models, Molecular , Protein Conformation , Receptors, Calcitonin/agonists , Receptors, Calcitonin/metabolism
10.
J Pharmacol Exp Ther ; 377(3): 417-440, 2021 06.
Article in English | MEDLINE | ID: mdl-33727283

ABSTRACT

Obesity and associated comorbidities are a major health burden, and novel therapeutics to help treat obesity are urgently needed. There is increasing evidence that targeting the amylin receptors (AMYRs), heterodimers of the calcitonin G protein-coupled receptor (CTR) and receptor activity-modifying proteins, improves weight control and has the potential to act additively with other treatments such as glucagon-like peptide-1 receptor agonists. Recent data indicate that AMYR agonists, which can also independently activate the CTR, may have improved efficacy for treating obesity, even though selective activation of CTRs is not efficacious. AM833 (cagrilintide) is a novel lipidated amylin analog that is undergoing clinical trials as a nonselective AMYR and CTR agonist. In the current study, we have investigated the pharmacology of AM833 across 25 endpoints and compared this peptide with AMYR selective and nonselective lipidated analogs (AM1213 and AM1784), and the clinically used peptide agonists pramlintide (AMYR selective) and salmon CT (nonselective). We also profiled human CT and rat amylin as prototypical selective agonists of CTR and AMYRs, respectively. Our results demonstrate that AM833 has a unique pharmacological profile across diverse measures of receptor binding, activation, and regulation. SIGNIFICANCE STATEMENT: AM833 is a novel nonselective agonist of calcitonin family receptors that has demonstrated efficacy for the treatment of obesity in phase 2 clinical trials. This study demonstrates that AM833 has a unique pharmacological profile across diverse measures of receptor binding, activation, and regulation when compared with other selective and nonselective calcitonin receptor and amylin receptor agonists. The present data provide mechanistic insight into the actions of AM833.


Subject(s)
Calcitonin , Protein Precursors , Animals , Male , Rats , Receptors, Calcitonin
11.
Biochem Biophys Res Commun ; 533(3): 559-564, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32980116

ABSTRACT

Human ghrelin receptor (GHSR) is a recognized prospective target in the diagnosis and therapy of multiple cancer types. To gain a better understanding of this receptor signaling system, we have synthesized a novel full-length ghrelin analog that is fluorescently labeled at the side-chain of a C-terminal cysteine extension. This analog exhibited nanomolar affinity and potency for the ghrelin receptor. It shows comparable efficacy with that of endogenous ghrelin. The fluorescently-labeled ghrelin analog is a valuable tool for in vitro imaging of cell lines that express ghrelin receptor.


Subject(s)
Ghrelin/analogs & derivatives , Ghrelin/chemical synthesis , Luminescent Proteins/chemical synthesis , Luminescent Proteins/metabolism , Fluorescence , HEK293 Cells , Humans , Luminescent Proteins/chemistry , Receptors, Ghrelin/metabolism
12.
BMC Cancer ; 19(1): 157, 2019 Feb 18.
Article in English | MEDLINE | ID: mdl-30777055

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common and aggressive type of primary brain cancer. With median survival of less than 15 months, identification and validation of new GBM therapeutic targets is of critical importance. RESULTS: In this study we tested expression and performed pharmacological characterization of the calcitonin receptor (CTR) as well as other members of the calcitonin family of receptors in high-grade glioma (HGG) cell lines derived from individual patient tumours, cultured in defined conditions. Previous immunohistochemical data demonstrated CTR expression in GBM biopsies and we were able to confirm CALCR (gene encoding CTR) expression. However, as assessed by cAMP accumulation assay, only one of the studied cell lines expressed functional CTR, while the other cell lines have functional CGRP (CLR/RAMP1) receptors. The only CTR-expressing cell line (SB2b) showed modest coupling to the cAMP pathway and no activation of other known CTR signaling pathways, including ERK1/2 and p38 MAP kinases, and Ca2+ mobilization, supportive of low cell surface receptor expression. Exome sequencing data failed to account for the discrepancy between functional data and expression on the cell lines that do not respond to calcitonin(s) with no deleterious non-synonymous polymorphisms detected, suggesting that other factors may be at play, such as alternative splicing or rapid constitutive receptor internalisation. CONCLUSIONS: This study shows that GPCR signaling can display significant variation depending on cellular system used, and effects seen in model recombinant cell lines or tumour cell lines are not always reproduced in a more physiologically relevant system and vice versa.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Receptors, Calcitonin/genetics , Receptors, Calcitonin/metabolism , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Calcitonin Receptor-Like Protein/genetics , Calcitonin Receptor-Like Protein/metabolism , Cell Culture Techniques , Cell Proliferation , Glioblastoma/mortality , Humans , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptor Activity-Modifying Protein 1/genetics , Receptor Activity-Modifying Protein 2/genetics , Signal Transduction , Survival Analysis , Transcriptome , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Cancer Immunol Immunother ; 66(9): 1217-1228, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28501939

ABSTRACT

We have reported that calcitonin receptor (CTR) is widely expressed in biopsies from the lethal brain tumour glioblastoma by malignant glioma and brain tumour-initiating cells (glioma stem cells) using anti-human CTR antibodies. A monoclonal antibody against an epitope within the extracellular domain of CTR was raised (mAb2C4) and chemically conjugated to either plant ribosome-inactivating proteins (RIPs) dianthin-30 or gelonin, or the drug monomethyl auristatin E (MMAE), and purified. In the high-grade glioma cell line (HGG, representing glioma stem cells) SB2b, in the presence of the triterpene glycoside SO1861, the EC50 for mAb2C4:dianthin was 10.0 pM and for mAb2C4:MMAE [antibody drug conjugate (ADC)] 2.5 nM, 250-fold less potent. With the cell line U87MG, in the presence of SO1861, the EC50 for mAb2C4:dianthin was 20 pM, mAb2C4:gelonin, 20 pM, compared to the ADC (6.3 nM), which is >300 less potent. Several other HGG cell lines that express CTR were tested and the efficacies of mAb2C4:RIP (dianthin or gelonin) were similar. Co-administration of the enhancer SO1861 purified from plants enhances lysosomal escape. Enhancement with SO1861 increased potency of the immunotoxin (>3 log values) compared to the ADC (1 log). The uptake of antibody was demonstrated with the fluorescent conjugate mAb2C4:Alexa Fluor 568, and the release of dianthin-30:Alexa Fluor488 into the cytosol following addition of SO1861 supports our model. These data demonstrate that the immunotoxins are highly potent and that CTR is an effective target expressed by a large proportion of HGG cell lines representative of glioma stem cells and isolated from individual patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Oligopeptides/pharmacology , Receptors, Calcitonin/antagonists & inhibitors , Ribosome Inactivating Proteins, Type 1/pharmacology , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Humans , Receptors, Calcitonin/immunology , Tumor Cells, Cultured
14.
Mol Pharmacol ; 89(3): 335-47, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26700562

ABSTRACT

The glucagon-like peptide 1 (GLP-1) receptor is a class B G protein-coupled receptor (GPCR) that is a key target for treatments for type II diabetes and obesity. This receptor, like other class B GPCRs, displays biased agonism, though the physiologic significance of this is yet to be elucidated. Previous work has implicated R2.60(190), N3.43(240), Q7.49(394), and H6.52(363) as key residues involved in peptide-mediated biased agonism, with R2.60(190), N3.43(240), and Q7.49(394) predicted to form a polar interaction network. In this study, we used novel insight gained from recent crystal structures of the transmembrane domains of the glucagon and corticotropin releasing factor 1 (CRF1) receptors to develop improved models of the GLP-1 receptor that predict additional key molecular interactions with these amino acids. We have introduced E6.53(364)A, N3.43(240)Q, Q7.49(394)N, and N3.43(240)Q/Q7.49(394)N mutations to probe the role of predicted H-bonding and charge-charge interactions in driving cAMP, calcium, or extracellular signal-regulated kinase (ERK) signaling. A polar interaction between E6.53(364) and R2.60(190) was predicted to be important for GLP-1- and exendin-4-, but not oxyntomodulin-mediated cAMP formation and also ERK1/2 phosphorylation. In contrast, Q7.49(394), but not R2.60(190)/E6.53(364) was critical for calcium mobilization for all three peptides. Mutation of N3.43(240) and Q7.49(394) had differential effects on individual peptides, providing evidence for molecular differences in activation transition. Collectively, this work expands our understanding of peptide-mediated signaling from the GLP-1 receptor and the key role that the central polar network plays in these events.


Subject(s)
Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide 1/chemistry , Models, Molecular , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallization , Glucagon-Like Peptide 1/metabolism , Humans , Hydrogen Bonding , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding/physiology , Protein Structure, Secondary
15.
Proc Natl Acad Sci U S A ; 109(45): 18607-12, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23091034

ABSTRACT

The glucagon-like peptide-1 receptor (GLP-1R) is a family B G protein-coupled receptor and an important drug target for the treatment of type II diabetes, with activation of pancreatic GLP-1Rs eliciting glucose-dependent insulin secretion. Currently, approved therapeutics acting at this receptor are peptide based, and there is substantial interest in small molecule modulators for the GLP-1R. Using a variety of resonance energy transfer techniques, we demonstrate that the GLP-1R forms homodimers and that transmembrane helix 4 (TM4) provides the primary dimerization interface. We show that disruption of dimerization using a TM4 peptide, a minigene construct encoding TM4, or by mutation of TM4, eliminates G protein-dependent high-affinity binding to GLP-1(7-36)NH(2) but has selective effects on receptor signaling. There was <10-fold decrease in potency in cAMP accumulation or ERK1/2 phosphorylation assays but marked loss of intracellular calcium mobilization by peptide agonists. In contrast, there was near-complete abrogation of the cAMP response to an allosteric agonist, compound 2, but preservation of ERK phosphorylation. Collectively, this indicates that GLP-1R dimerization is important for control of signal bias. Furthermore, we reveal that two small molecule ligands are unaltered in their ability to allosterically modulate signaling from peptide ligands, demonstrating that these modulators act in cis within a single receptor protomer, and this has important implications for small molecule drug design.


Subject(s)
Protein Multimerization/drug effects , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Allosteric Regulation/drug effects , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cyclic AMP/metabolism , Glucagon-Like Peptide-1 Receptor , Humans , Peptides/pharmacology , Protein Subunits/metabolism
16.
Biochem Pharmacol ; 219: 115932, 2024 01.
Article in English | MEDLINE | ID: mdl-37989413

ABSTRACT

Bitter taste receptors (T2R) are a subfamily of G protein-coupled receptors that enable humans to detect aversive and toxic substances. The ability to discern bitter compounds varies between individuals and is attributed mainly to naturally occurring T2R polymorphisms. T2Rs are also expressed in numerous non-gustatory tissues, including the heart, indicating potential contributions to cardiovascular physiology. In this study. T2Rs that have previously been identified in human cardiac tissues (T2Rs - 10, 14, 30, 31, 46 and 50) and their naturally occurring polymorphisms were functionally characterised. The ligand-dependent signaling responses of some T2R variants were completely abolished (T2R30 Leu252 and T2R46 Met228), whereas other receptor variants had moderate changes in their maximal response, but not potency, relative to wild type. Using a cAMP fluorescent biosensor, we reveal the productive coupling of T2R14, but not the T2R14 Phe201 variant, to endogenous Gαi. Modeling revealed that these variants resulted in altered interactions that generally affected ligand binding (T2R30 Leu252) or Gα protein interactions (T2R46 Met228 and T2R14 Phe201), rather than receptor structural stability. Interestingly, this study is the first to show a difference in signaling for T2R50 Tyr203 (rs1376251) which has been associated with cardiovascular disease. The observation of naturally occurring functional variation in the T2Rs with the greatest expression in the heart is important, as their discovery should prove useful in deciphering the role of T2Rs within the cardiovascular system.


Subject(s)
Receptors, G-Protein-Coupled , Taste , Humans , Taste/physiology , Ligands , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
17.
J Biol Chem ; 287(24): 20674-88, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22535965

ABSTRACT

Caveolins act as scaffold proteins in multiprotein complexes and have been implicated in signaling by G protein-coupled receptors. Studies using knock-out mice suggest that ß(3)-adrenoceptor (ß(3)-AR) signaling is dependent on caveolin-1; however, it is not known whether caveolin-1 is associated with the ß(3)-AR or solely with downstream signaling proteins. We have addressed this question by examining the impact of membrane rafts and caveolin-1 on the differential signaling of mouse ß(3a)- and ß(3b)-AR isoforms that diverge at the distal C terminus. Only the ß(3b)-AR promotes pertussis toxin (PTX)-sensitive cAMP accumulation. When cells expressing the ß(3a)-AR were treated with filipin III to disrupt membrane rafts or transfected with caveolin-1 siRNA, the cyclic AMP response to the ß(3)-AR agonist CL316243 became PTX-sensitive, suggesting Gα(i/o) coupling. The ß(3a)-AR C terminus, SP(384)PLNRF(389)DGY(392)EGARPF(398)PT, resembles a caveolin interaction motif. Mutant ß(3a)-ARs (F389A/Y392A/F398A or P384S/F389A) promoted PTX-sensitive cAMP responses, and in situ proximity assays demonstrated an association between caveolin-1 and the wild type ß(3a)-AR but not the mutant receptors. In membrane preparations, the ß(3b)-AR activated Gα(o) and mediated PTX-sensitive cAMP responses, whereas the ß(3a)-AR did not activate Gα(i/o) proteins. The endogenous ß(3a)-AR displayed Gα(i/o) coupling in brown adipocytes from caveolin-1 knock-out mice or in wild type adipocytes treated with filipin III. Our studies indicate that interaction of the ß(3a)-AR with caveolin inhibits coupling to Gα(i/o) proteins and suggest that signaling is modulated by a raft-enriched complex containing the ß(3a)-AR, caveolin-1, Gα(s), and adenylyl cyclase.


Subject(s)
Caveolin 1/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Membrane Microdomains/metabolism , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction/physiology , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Adrenergic beta-Agonists/pharmacology , Amino Acid Motifs , Amino Acid Substitution , Animals , CHO Cells , Caveolin 1/genetics , Cricetinae , Cricetulus , Dioxoles/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Membrane Microdomains/genetics , Mice , Mice, Knockout , Mutation, Missense , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Adrenergic, beta-3/genetics , Signal Transduction/drug effects
18.
Biochem Soc Trans ; 41(1): 172-9, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23356279

ABSTRACT

Type 2 diabetes is a major global health problem and there is ongoing research for new treatments to manage the disease. The GLP-1R (glucagon-like peptide-1 receptor) controls the physiological response to the incretin peptide, GLP-1, and is currently a major target for the development of therapeutics owing to the broad range of potential beneficial effects in Type 2 diabetes. These include promotion of glucose-dependent insulin secretion, increased insulin biosynthesis, preservation of ß-cell mass, improved peripheral insulin sensitivity and promotion of weight loss. Despite this, our understanding of GLP-1R function is still limited, with the desired spectrum of GLP-1R-mediated signalling yet to be determined. We review the current understanding of GLP-1R function, in particular, highlighting recent contributions in the field on allosteric modulation, probe-dependence and ligand-directed signal bias and how these behaviours may influence future drug development.


Subject(s)
Receptors, Glucagon/physiology , Allosteric Regulation , Amino Acid Sequence , Diabetes Mellitus, Type 2/physiopathology , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide-1 Receptor , Humans , Incretins/physiology , Molecular Mimicry , Molecular Sequence Data , Receptors, Glucagon/metabolism , Sequence Homology, Amino Acid , Signal Transduction
19.
Mol Pharmacol ; 82(6): 1082-93, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22936816

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a signal-regulated transcription factor, which is canonically activated by the direct binding of xenobiotics. In addition, switching cells from adherent to suspension culture also activates the AhR, representing a nonxenobiotic, physiological activation of AhR signaling. Here, we show that the AhR is recruited to target gene enhancers in both ligand [isopropyl-2-(1,3-dithietane-2-ylidene)-2-[N-(4-methylthiazol-2-yl)carbamoyl]acetate (YH439)]-treated and suspension cells, suggesting a common mechanism of target gene induction between these two routes of AhR activation. However, gene expression profiles critically differ between xenobiotic- and suspension-activated AhR signaling. Por and Cldnd1 were regulated predominantly by ligand treatments, whereas, in contrast, ApoER2 and Ganc were regulated predominantly by the suspension condition. Classic xenobiotic-metabolizing AhR targets such as Cyp1a1, Cyp1b1, and Nqo1 were regulated by both ligand and suspension conditions. Temporal expression patterns of AhR target genes were also found to vary, with examples of transient activation, transient repression, or sustained alterations in expression. Furthermore, sequence analysis coupled with chromatin immunoprecipitation assays and reporter gene analysis identified a functional xenobiotic response element (XRE) in the intron 1 of the mouse Tiparp gene, which was also bound by hypoxia-inducible factor-1α during hypoxia and features a concatemer of four XRE cores (GCGTG). Our data suggest that this XRE concatemer site concurrently regulates the expression of both the Tiparp gene and its cis antisense noncoding RNA after ligand- or suspension-induced AhR activation. This work provides novel insights into how AhR signaling drives different transcriptional programs via the ligand versus suspension modes of activation.


Subject(s)
Cell Adhesion/genetics , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Transcriptional Activation/genetics , Xenobiotics/pharmacology , Animals , Cell Adhesion/drug effects , Cell Line , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression/drug effects , HEK293 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Ligands , Mice , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Transcriptome/drug effects
20.
Mol Pharmacol ; 82(6): 1066-73, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22930710

ABSTRACT

Identifying novel mechanisms to enhance glucagon-like peptide-1 (GLP-1) receptor signaling may enable nascent medicinal chemistry strategies with the aim of developing new orally available therapeutic agents for the treatment of type 2 diabetes mellitus. Therefore, we tested the hypothesis that selectively modulating the low-affinity GLP-1 receptor agonist, oxyntomodulin, would improve the insulin secretory properties of this naturally occurring hormone to provide a rationale for pursuing an unexplored therapeutic approach. Signal transduction and competition binding studies were used to investigate oxyntomodulin activity on the GLP-1 receptor in the presence of the small molecule GLP-1 receptor modulator, 4-(3-benzyloxyphenyl)-2-ethylsulfinyl-6-(trifluoromethyl)pyrimidine (BETP). In vivo, the intravenous glucose tolerance test characterized oxyntomodulin-induced insulin secretion in animals administered the small molecule. BETP increased oxyntomodulin binding affinity for the GLP-1 receptor and enhanced oxyntomodulin-mediated GLP-1 receptor signaling as measured by activation of the α subunit of heterotrimeric G protein and cAMP accumulation. In addition, oxyntomodulin-induced insulin secretion was enhanced in the presence of the compound. BETP was pharmacologically characterized to induce biased signaling by oxyntomodulin. These studies demonstrate that small molecules targeting the GLP-1 receptor can increase binding and receptor activation of the endogenous peptide oxyntomodulin. The biased signaling engendered by BETP suggests that GLP-1 receptor mobilization of cAMP is the critical insulinotropic signaling event. Because of the unique metabolic properties of oxyntomodulin, identifying molecules that enhance its activity should be pursued to assess the efficacy and safety of this novel mechanism.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin/metabolism , Oxyntomodulin/pharmacology , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Animals , CHO Cells , Cell Line , Cricetinae , Cyclic AMP/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Drug Synergism , GTP-Binding Proteins/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , HEK293 Cells , Humans , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL