Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Sci Signal ; 16(779): eabl4283, 2023 04 04.
Article in English | MEDLINE | ID: mdl-37014928

ABSTRACT

The chemotactic G protein-coupled receptor GPR183 and its most potent endogenous oxysterol ligand 7α,25-dihydroxycholesterol (7α,25-OHC) are important for immune cell positioning in secondary lymphoid tissues. This receptor-ligand pair is associated with various diseases, in some cases contributing favorably and in other cases adversely, making GPR183 an attractive target for therapeutic intervention. We investigated the mechanisms underlying GPR183 internalization and the role of internalization in the main biological function of the receptor, chemotaxis. We found that the C terminus of the receptor was important for ligand-induced internalization but less so for constitutive (ligand-independent) internalization. ß-arrestin potentiated ligand-induced internalization but was not required for ligand-induced or constitutive internalization. Caveolin and dynamin were the main mediators of both constitutive and ligand-induced receptor internalization in a mechanism independent of G protein activation. Clathrin-mediated endocytosis also contributed to constitutive GPR183 internalization in a ß-arrestin-independent manner, suggesting the existence of different pools of surface-localized GPR183. Chemotaxis mediated by GPR183 depended on receptor desensitization by ß-arrestins but could be uncoupled from internalization, highlighting an important biological role for the recruitment of ß-arrestin to GPR183. The role of distinct pathways in internalization and chemotaxis may aid in the development of GPR183-targeting drugs for specific disease contexts.


Subject(s)
Arrestin , Arrestins , Arrestin/metabolism , Arrestins/genetics , Arrestins/metabolism , Ligands , beta-Arrestins/metabolism , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism , Endocytosis
2.
PLoS One ; 18(3): e0279335, 2023.
Article in English | MEDLINE | ID: mdl-36862673

ABSTRACT

Weight loss through bariatric surgery is efficient for treatment or prevention of obesity related diseases such as type 2 diabetes and cardiovascular disease. Long term weight loss response does, however, vary among patients undergoing surgery. Thus, it is difficult to identify predictive markers while most obese individuals have one or more comorbidities. To overcome such challenges, an in-depth multiple omics analyses including fasting peripheral plasma metabolome, fecal metagenome as well as liver, jejunum, and adipose tissue transcriptome were performed for 106 individuals undergoing bariatric surgery. Machine leaning was applied to explore the metabolic differences in individuals and evaluate if metabolism-based patients' stratification is related to their weight loss responses to bariatric surgery. Using Self-Organizing Maps (SOMs) to analyze the plasma metabolome, we identified five distinct metabotypes, which were differentially enriched for KEGG pathways related to immune functions, fatty acid metabolism, protein-signaling, and obesity pathogenesis. The gut metagenome of the most heavily medicated metabotypes, treated simultaneously for multiple cardiometabolic comorbidities, was significantly enriched in Prevotella and Lactobacillus species. This unbiased stratification into SOM-defined metabotypes identified signatures for each metabolic phenotype and we found that the different metabotypes respond differently to bariatric surgery in terms of weight loss after 12 months. An integrative framework that utilizes SOMs and omics integration was developed for stratifying a heterogeneous bariatric surgery cohort. The multiple omics datasets described in this study reveal that the metabotypes are characterized by a concrete metabolic status and different responses in weight loss and adipose tissue reduction over time. Our study thus opens a path to enable patient stratification and hereby allow for improved clinical treatments.


Subject(s)
Bariatric Surgery , Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/surgery , Obesity/surgery , Adipose Tissue , Algorithms
3.
Metabolites ; 11(7)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201526

ABSTRACT

G-protein-coupled receptor-35 (GPR35) has been identified as a receptor for the tryptophan metabolite kynurenic acid (KynA) and suggested to modulate macrophage polarization in metabolic tissues. Whether GPR35 can influence vascular inflammation and atherosclerosis has however never been tested. Lethally irradiated LdlrKO mice were randomized to receive GPR35KO or wild type (WT) bone marrow transplants and fed a high cholesterol diet for eight weeks to develop atherosclerosis. GPR35KO and WT chimeric mice presented no difference in the size of atherosclerotic lesions in the aortic arch (2.37 ± 0.58% vs. 1.95 ± 0.46%, respectively) or in the aortic roots (14.77 ± 3.33% vs. 11.57 ± 2.49%, respectively). In line with these data, no changes in the percentage of VCAM-1+, IAb + cells, and CD3+ T cells, as well as alpha smooth muscle cell actin expression, was observed between groups. Interestingly, the GPR35KO group presented a small but significant increase in CD68+ macrophage infiltration in the plaque. However, in vitro culture experiments using bone marrow-derived macrophages from both groups indicated that GPR35 plays no role in modulating the secretion of major inflammatory cytokines. Our study indicates that GPR35 expression does not play a direct role in macrophage activation, vascular inflammation, and the development of atherosclerosis.

4.
Cell Rep ; 35(11): 109246, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34133934

ABSTRACT

Succinate functions both as a classical TCA cycle metabolite and an extracellular metabolic stress signal sensed by the mainly Gi-coupled succinate receptor SUCNR1. In the present study, we characterize and compare effects and signaling pathways activated by succinate and both classes of non-metabolite SUCNR1 agonists. By use of specific receptor and pathway inhibitors, rescue in G-protein-depleted cells and monitoring of receptor G protein activation by BRET, we identify Gq rather than Gi signaling to be responsible for SUCNR1-mediated effects on basic transcriptional regulation. Importantly, in primary human M2 macrophages, in which SUCNR1 is highly expressed, we demonstrate that physiological concentrations of extracellular succinate act through SUCNR1-activated Gq signaling to efficiently regulate transcription of immune function genes in a manner that hyperpolarizes their M2 versus M1 phenotype. Thus, sensing of stress-induced extracellular succinate by SUCNR1 is an important transcriptional regulator in human M2 macrophages through Gq signaling.


Subject(s)
Extracellular Space/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Macrophages/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Succinic Acid/metabolism , Arrestins/metabolism , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Expression Regulation , Gene Ontology , HEK293 Cells , Humans , Ligands , Macrophages/immunology , Male , Models, Biological , Protein Subunits/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Transcriptional Activation/genetics , Type C Phospholipases/metabolism
5.
Mol Metab ; 49: 101207, 2021 07.
Article in English | MEDLINE | ID: mdl-33711555

ABSTRACT

OBJECTIVES: Obesity is a complex disease associated with a high risk of comorbidities. Gastric bypass surgery, an invasive procedure with low patient eligibility, is currently the most effective intervention that achieves sustained weight loss. This beneficial effect is attributed to alterations in gut hormone signaling. An attractive alternative is to pharmacologically mimic the effects of bariatric surgery by targeting several gut hormonal axes. The G protein-coupled receptor 39 (GPR39) expressed in the gastrointestinal tract has been shown to mediate ghrelin signaling and control appetite, food intake, and energy homeostasis, but the broader effect on gut hormones is largely unknown. A potent and efficacious GPR39 agonist (Cpd1324) was recently discovered, but the in vivo function was not addressed. Herein we studied the efficacy of the GPR39 agonist, Cpd1324, on metabolism and gut hormone secretion. METHODS: Body weight, food intake, and energy expenditure in GPR39 agonist-treated mice and GPR39 KO mice were studied in calorimetric cages. Plasma ghrelin, glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), and peptide YY (PYY) levels were measured. Organoids generated from murine and human small intestine and mouse colon were used to study GLP-1 and PYY release. Upon GPR39 agonist administration, dynamic changes in intracellular GLP-1 content were studied via immunostaining and changes in ion transport across colonic mucosa were monitored in Ussing chambers. The G protein activation underlying GPR39-mediated selective release of gut hormones was studied using bioluminescence resonance energy transfer biosensors. RESULTS: The GPR39 KO mice displayed a significantly increased food intake without corresponding increases in respiratory exchange ratios or energy expenditure. Oral administration of a GPR39 agonist induced an acute decrease in food intake and subsequent weight loss in high-fat diet (HFD)-fed mice without affecting their energy expenditure. The tool compound, Cpd1324, increased GLP-1 secretion in the mice as well as in mouse and human intestinal organoids, but not in GPR39 KO mouse organoids. In contrast, the GPR39 agonist had no effect on PYY or GIP secretion. Transepithelial ion transport was acutely affected by GPR39 agonism in a GLP-1- and calcitonin gene-related peptide (CGRP)-dependent manner. Analysis of Cpd1324 signaling properties showed activation of Gαq and Gαi/o signaling pathways in L cells, but not Gαs signaling. CONCLUSIONS: The GPR39 agonist described in this study can potentially be used by oral administration as a weight-lowering agent due to its stimulatory effect on GLP-1 secretion, which is most likely mediated through a unique activation of Gα subunits. Thus, GPR39 agonism may represent a novel approach to effectively treat obesity through selective modulation of gastrointestinal hormonal axes.


Subject(s)
Gastrointestinal Hormones/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Animals , Appetite Regulation , Bariatric Surgery , Body Weight , Eating , Enteroendocrine Cells , Gastric Inhibitory Polypeptide/pharmacology , Ghrelin/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Peptide YY/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Gastrointestinal Hormone , Weight Loss
6.
Mol Metab ; 19: 49-64, 2019 01.
Article in English | MEDLINE | ID: mdl-30472415

ABSTRACT

OBJECTIVES: GPR142, which is highly expressed in pancreatic islets, has recently been deorphanized as a receptor for aromatic amino acids; however, its physiological role and pharmacological potential is unclear. METHODS AND RESULTS: We find that GPR142 is expressed not only in ß- but also in α-cells of the islets as well as in enteroendocrine cells, and we confirm that GPR142 is a highly selective sensor of essential aromatic amino acids, in particular Trp and oligopeptides with N-terminal Trp. GPR142 knock-out mice displayed a very limited metabolic phenotype but demonstrated that L-Trp induced secretion of pancreatic and gut hormones is mediated through GPR142 but that the receptor is not required for protein-induced hormone secretion. A synthetic GPR142 agonist stimulated insulin and glucagon as well as GIP, CCK, and GLP-1 secretion. In particular, GIP secretion was sensitive to oral administration of the GPR142 agonist an effect which in contrast to the other hormones was blocked by protein load. Oral administration of the GPR142 agonist increased [3H]-2-deoxyglucose uptake in muscle and fat depots mediated through insulin action while it lowered liver glycogen conceivably mediated through glucagon, and, consequently, it did not lower total blood glucose. Nevertheless, acute administration of the GPR142 agonist strongly improved oral glucose tolerance in both lean and obese mice as well as Zucker fatty rat. Six weeks in-feed chronic treatment with the GPR142 agonist did not affect body weight in DIO mice, but increased energy expenditure and carbohydrate utilization, lowered basal glucose, and improved insulin sensitivity. CONCLUSIONS: GPR142 functions as a sensor of aromatic amino acids, controlling GIP but also CCK and GLP-1 as well as insulin and glucagon in the pancreas. GPR142 agonists could have novel interesting potential in modifying metabolism through a balanced action of gut hormones as well as both insulin and glucagon.


Subject(s)
Islets of Langerhans/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acids, Aromatic/metabolism , Animals , Blood Glucose/metabolism , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Rats , Rats, Zucker , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Tryptophan/metabolism
7.
Mol Metab ; 6(12): 1585-1596, 2017 12.
Article in English | MEDLINE | ID: mdl-29157600

ABSTRACT

OBJECTIVE: Besides functioning as an intracellular metabolite, succinate acts as a stress-induced extracellular signal through activation of GPR91 (SUCNR1) for which we lack suitable pharmacological tools. METHODS AND RESULTS: Here we first determined that the cis conformation of the succinate backbone is preferred and that certain backbone modifications are allowed for GPR91 activation. Through receptor modeling over the X-ray structure of the closely related P2Y1 receptor, we discovered that the binding pocket is partly occupied by a segment of an extracellular loop and that succinate therefore binds in a very different mode than generally believed. Importantly, an empty side-pocket is identified next to the succinate binding site. All this information formed the basis for a substructure-based search query, which, combined with molecular docking, was used in virtual screening of the ZINC database to pick two serial mini-libraries of a total of only 245 compounds from which sub-micromolar, selective GPR91 agonists of unique structures were identified. The best compounds were backbone-modified succinate analogs in which an amide-linked hydrophobic moiety docked into the side-pocket next to succinate as shown by both loss- and gain-of-function mutagenesis. These compounds displayed GPR91-dependent activity in altering cytokine expression in human M2 macrophages similar to succinate, and importantly were devoid of any effect on the major intracellular target, succinate dehydrogenase. CONCLUSIONS: These novel, synthetic non-metabolite GPR91 agonists will be valuable both as pharmacological tools to delineate the GPR91-mediated functions of succinate and as leads for the development of GPR91-targeted drugs to potentially treat low grade metabolic inflammation and diabetic complications such as retinopathy and nephropathy.


Subject(s)
Molecular Docking Simulation , Receptors, G-Protein-Coupled/agonists , Small Molecule Libraries/pharmacology , Cells, Cultured , Drug Discovery/methods , HEK293 Cells , Humans , Protein Binding , Quantitative Structure-Activity Relationship , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Small Molecule Libraries/chemistry
8.
Cell Metab ; 25(4): 777-796, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28380372

ABSTRACT

In addition to their bioenergetic intracellular function, several classical metabolites act as extracellular signaling molecules activating cell-surface G-protein-coupled receptors (GPCRs), similar to hormones and neurotransmitters. "Signaling metabolites" generated from nutrients or by gut microbiota target primarily enteroendocrine, neuronal, and immune cells in the lamina propria of the gut mucosa and the liver and, through these tissues, the rest of the body. In contrast, metabolites from the intermediary metabolism act mainly as metabolic stress-induced autocrine and paracrine signals in adipose tissue, the liver, and the endocrine pancreas. Importantly, distinct metabolite GPCRs act as efficient pro- and anti-inflammatory regulators of key immune cells, and signaling metabolites may thus function as important drivers of the low-grade inflammation associated with insulin resistance and obesity. The concept of key metabolites as ligands for specific GPCRs has broadened our understanding of metabolic signaling significantly and provides a number of novel potential drug targets.


Subject(s)
Metabolome , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Enteroendocrine Cells/metabolism , Gastrointestinal Microbiome , Humans , Ligands
9.
J Immunol ; 194(4): 1417-21, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25595774

ABSTRACT

Triggering receptor expressed on myeloid cells (TREM)-1 is an orphan receptor implicated in innate immune activation. Inhibition of TREM-1 reduces sepsis in mouse models, suggesting a role for it in immune responses triggered by bacteria. However, the absence of an identified ligand has hampered a full understanding of TREM-1 function. We identified complexes between peptidoglycan recognition protein 1 (PGLYRP1) and bacterially derived peptidoglycan that constitute a potent ligand capable of binding TREM-1 and inducing known TREM-1 functions. Interestingly, multimerization of PGLYRP1 bypassed the need for peptidoglycan in TREM-1 activation, demonstrating that the PGLYRP1/TREM-1 axis can be activated in the absence of bacterial products. The role for PGLYRP1 as a TREM-1 activator provides a new mechanism by which bacteria can trigger myeloid cells, linking two known, but previously unrelated, pathways in innate immunity.


Subject(s)
Cytokines/immunology , Immunity, Innate/immunology , Membrane Glycoproteins/immunology , Neutrophils/immunology , Receptors, Immunologic/immunology , Humans , Immunoprecipitation , Ligands , Surface Plasmon Resonance , Triggering Receptor Expressed on Myeloid Cells-1
10.
J Biol Chem ; 287(12): 9454-60, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22235133

ABSTRACT

IL-21 is a class I cytokine that exerts pleiotropic effects on both innate and adaptive immune responses. It signals through a heterodimeric receptor complex consisting of the IL-21 receptor (IL-21R) and the common γ-chain. A hallmark of the class I cytokine receptors is the class I cytokine receptor signature motif (WSXWS). The exact role of this motif has not been determined yet; however, it has been implicated in diverse functions, including ligand binding, receptor internalization, proper folding, and export, as well as signal transduction. Furthermore, the WXXW motif is known to be a consensus sequence for C-mannosylation. Here, we present the crystal structure of IL-21 bound to IL-21R and reveal that the WSXWS motif of IL-21R is C-mannosylated at the first tryptophan. We furthermore demonstrate that a sugar chain bridges the two fibronectin domains that constitute the extracellular domain of IL-21R and anchors at the WSXWS motif through an extensive hydrogen bonding network, including mannosylation. The glycan thus transforms the V-shaped receptor into an A-frame. This finding offers a novel structural explanation of the role of the class I cytokine signature motif.


Subject(s)
Interleukins/chemistry , Interleukins/metabolism , Receptors, Interleukin-21/chemistry , Receptors, Interleukin-21/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Glycosylation , Humans , Interleukins/genetics , Mannose/metabolism , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Receptors, Interleukin-21/genetics
11.
J Biol Chem ; 285(16): 12223-31, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20167599

ABSTRACT

The cytokine interleukin (IL)-21 exerts pleiotropic effects acting through innate as well as adaptive immune responses. The activities of IL-21 are mediated through binding to its cognate receptor complex composed of the IL-21 receptor private chain (IL-21Ralpha) and the common gamma-chain (gammaC), the latter being shared by IL-2, IL-4, IL-7, IL-9, and IL-15. The binding energy of the IL-21 ternary complex is predominantly provided by the high affinity interaction between IL-21 and IL-21Ralpha, whereas the interaction between IL-21 and gammaC, albeit essential for signaling, is rather weak. The design of IL-21 analogues, which have lost most or all affinity toward the signaling gammaC chain, while simultaneously maintaining a tight interaction with the private chain, would in theory represent candidates for IL-21 antagonists. We predicted the IL-21 residues, which compose the gammaC binding epitope using homology modeling and alignment with the related cytokines, IL-2 and IL-4. Next we systematically analyzed the predicted binding epitope by a mutagenesis study. Indeed two mutants, which have significantly impaired gammaC affinity with undiminished IL-21Ralpha affinity, were successfully identified. Functional studies confirmed that these two novel hIL-21 double mutants do act as hIL-21 antagonists.


Subject(s)
Drug Design , Interleukins/antagonists & inhibitors , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Cell Line , Epitopes/chemistry , Epitopes/genetics , Epitopes/metabolism , Humans , In Vitro Techniques , Interleukin-2/chemistry , Interleukin-2/genetics , Interleukin-4/chemistry , Interleukin-4/genetics , Interleukins/chemistry , Interleukins/genetics , Interleukins/metabolism , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , Receptors, Interleukin-21/chemistry , Receptors, Interleukin-21/genetics , Receptors, Interleukin-21/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Structural Homology, Protein
12.
J Biol Chem ; 282(32): 23326-36, 2007 Aug 10.
Article in English | MEDLINE | ID: mdl-17565991

ABSTRACT

The high resolution three-dimensional structure of human interleukin (hIL)-21 has been resolved by heteronuclear NMR spectroscopy. Overall, the hIL-21 structure is dominated by a well defined central four-helical bundle, arranged in an up-up-down-down topology, as observed for other cytokines. A segment of the hIL-21 molecule that includes the third helical segment, helix C, is observed to exist in two distinct and interchangeable states. In one conformer, the helix C segment is presented in a regular, alpha-helical conformation, whereas in the other conformer, this segment is largely disordered. A structure-based sequence alignment of hIL-21 with receptor complexes of the related cytokines, interleukin-2 and -4, implied that this particular segment is involved in receptor binding. An hIL-21 analog was designed to stabilize the region around helix C through the introduction of a segment grafted from hIL-4. This novel hIL-21 analog was demonstrated to exhibit a 10-fold increase in potency in a cellular assay.


Subject(s)
Interleukins/chemistry , Amino Acid Sequence , Cell Line , Crystallography, X-Ray , Humans , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Protein Conformation , Protein Engineering , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Temperature
13.
Regul Pept ; 119(3): 151-4, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15120474

ABSTRACT

BACKGROUND: The enteroinsular-axis is abnormal in type 2 diabetics, which contributes to the diabetic phenotype. The effect of the incretin hormone gastric inhibitory polypeptide (GIP) and the secretion of the incretin hormone glucagon-like peptide-1 (GLP-1) are thus greatly diminished. The explanation for these changes could be changes in the structure of either of the hormones or their receptors. Thus, the aim of this study was to study the occurrence of genetic variants in the GIP and GLP-1 encoding regions of the proGIP and proglucagon genes in type 2 diabetic patients and matched control subjects. METHODS AND RESULTS: Genomic DNA was extracted from buffy coats from 12 Caucasian type 2 diabetics and 12 healthy subjects, matched with respect to sex, age and BMI. The GIP and GLP-1 sequences were amplified using specific primers using the polymerase chain reaction (PCR). The amplified products were then sequenced. No germ-line mutations were identified in the GIP and the GLP-1 encoding regions of the proGIP and proglucagon genes in either the type 2 diabetic or the control subjects. CONCLUSIONS: The perturbed incretin effect in type 2 diabetics is not commonly caused by genetic variants in either the GIP or the GLP-1 encoding genes in type 2 diabetics.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Gastric Inhibitory Polypeptide/genetics , Glucagon/genetics , Peptide Fragments/genetics , Phenotype , Protein Precursors/genetics , Aged , Base Sequence , DNA/analysis , Denmark , Diabetes Mellitus, Type 2/metabolism , Female , Gastric Inhibitory Polypeptide/metabolism , Genetic Predisposition to Disease , Glucagon/metabolism , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Humans , Male , Middle Aged , Molecular Sequence Data , Peptide Fragments/metabolism , Polymerase Chain Reaction , Predictive Value of Tests , Protein Precursors/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Sequence Analysis, DNA , White People
14.
Can J Physiol Pharmacol ; 80(5): 413-7, 2002 May.
Article in English | MEDLINE | ID: mdl-12056547

ABSTRACT

To identify residues of the rat AT1A angiotensin II receptor involved with signal transduction and binding of the non-peptide agonist L-162,313 (5,7-dimethyl-2-ethyl-3-[[4-[2(n-butyloxycarbonylsulfonamido)-5-isobutyl-3-thienyl]phenyl]methyl]imidazol[4,5,6]-pyridine) we have performed ligand binding and inositol phosphate turnover assays in COS-7 cells transiently transfected with the wild-type and mutant forms of the receptor. Mutant receptors bore modifications in the extracellular region: T88H, Y92H, G1961, G196W, and D278E. Compound L-162,313 displaced [125I]-Sar1,Leu8-AngII from the mutants G196I and G196W with IC50 values similar to that of the wild-type. The affinity was, however, slightly affected by the D278E mutation and more significantly by the T88H and Y92H mutations. In inositol phosphate turnover assays, the ability of L-162,313 to trigger the activation cascade was compared with that of angiotensin II. These assays showed that the G196W mutant reached a relative maximum activation exceeding that of the wild-type receptor; the efficacy was slightly reduced in the G1961 mutant and further reduced in the T88H, Y92H, and D278E mutants. Our data suggest that residues of the extracellular domain of the AT1 receptor are involved in the binding of the non-peptide ligand, or in a general receptor activation phenomenon that involves conformational modifications for a preferential binding of agonists or antagonists.


Subject(s)
Biphenyl Compounds/metabolism , Imidazoles/metabolism , Mutagenesis, Site-Directed , Receptors, Angiotensin/agonists , Receptors, Angiotensin/metabolism , Amino Acid Sequence/physiology , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Biphenyl Compounds/pharmacology , COS Cells , Chlorocebus aethiops , Dose-Response Relationship, Drug , Imidazoles/pharmacology , Molecular Sequence Data , Rats , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/chemistry , Receptors, Angiotensin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL